fulltext - DiVA Portal

Digital Comprehensive Summaries of Uppsala Dissertations
from the Faculty of Medicine 1095
The Effects of SSRI Treatment on
Human Placenta and Embryo
HELENA KAIHOLA
ACTA
UNIVERSITATIS
UPSALIENSIS
UPPSALA
2015
ISSN 1651-6206
ISBN 978-91-554-9227-4
urn:nbn:se:uu:diva-248527
Dissertation presented at Uppsala University to be publicly examined in Sal IV,
Universitetshuset, Uppsala, Thursday, 28 May 2015 at 09:15 for the degree of Doctor of
Philosophy (Faculty of Medicine). The examination will be conducted in Swedish. Faculty
examiner: Professor Olav Spigset (Department of Laboratory Medicine, Children's and
Women's Health, Norwegian University of Science and Technology, Trondheim, Norway).
Abstract
Kaihola, H. 2015. The Effects of SSRI Treatment on Human Placenta and Embryo. Digital
Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 1095.
90 pp. Uppsala: Acta Universitatis Upsaliensis. ISBN 978-91-554-9227-4.
During pregnancy, 4 - 7% of women suffer from major depressive disorder. When antidepressive
treatment is needed, selective serotonin reuptake inhibitors (SSRIs) are the most commonly
used. Although severe complications from SSRI treatment are rare, association with a number
of adverse pregnancy and fetal outcomes has been found. Also, antenatal depression per se has
been shown to affect pregnancy outcomes. The overall aim of this thesis was to examine the
effects of SSRIs on human placenta and embryo.
In the first study, gene expression was investigated in placenta from depressed, SSRItreated and healthy pregnant women, using microarray analysis. Antenatal depression and SSRI
treatment induced alterations in gene expression, but only 20 genes in common were noted.
Validation with qRT-PCR showed that six out of seven selected genes were altered in SSRItreated women compared with controls, and two genes were altered between depressed women
and controls.
In study two, the protein levels in placenta from depressed, SSRI-treated and healthy pregnant
women were investigated, focusing on the NGF signaling pathway. NGF, phosphorylated Raf-1,
ROCK2 and phosphorylated ROCK2, were altered in both SSRI-treated and depressed women,
although the proteins were regulated differently in the two groups.
In the third study, human embryos were treated with fluoxetine. Embryo development
and protein expression were studied. Fluoxetine had some effect on the timing of embryo
developmental stages. Also, several proteins were uniquely found in fluoxetine-treated embryos
compared with untreated embryos. Fluoxetine also altered the levels of proteins secreted from
the embryo.
In the fourth study, the human neuroblastoma cell line SH-SY5Y/TrkA was treated with TPA
and NGF. The activation of Raf-1 was investigated and the involvement of Ras and PKC was
studied. Both NGF and TPA activated Raf-1, but to a different extent and via different pathways.
The NGF-induced activation of Raf-1 was mediated via Ras, while TPA induced signaling via
PKC.
In conclusion, SSRI treatment and antenatal depression influence placental gene and protein
expression. These findings may affect placental development and function, which in turn could
affect fetal development. Also, direct exposure of embryos to fluoxetine has some effects on
embryo development and protein expression, which may affect the development of the fetus.
Keywords: antenatal depression, embryo, embryo development, fluoxetine, gene expression,
NGF, placenta, protein expression, Raf-1, ROCK, signaling pathways, SSRI
Helena Kaihola, Department of Women's and Children's Health, Akademiska sjukhuset,
Uppsala University, SE-75185 Uppsala, Sweden.
© Helena Kaihola 2015
ISSN 1651-6206
ISBN 978-91-554-9227-4
urn:nbn:se:uu:diva-248527 (http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-248527)
To my family
List of Papers
This thesis is based on the following studies, which are referred to in the text
by their Roman numerals. To be noted: my maiden name was Söderholm.
I
Olivier, J.D.A., Åkerud, H., Skalkidou, A., Kaihola, H.,
Sundström Poromaa, I. (2015) The effects of antenatal depression and antidepressant treatment on placental gene expression. Front Cell Neurosci 8:465.
II
Kaihola, H., Olivier, J., Sundström Poromaa, I., Åkerud, H.
(2015) The effect of antenatal depression and selective serotonin reuptake inhibitor treatment on nerve growth factor signaling in human placenta. PLOS ONE 10(1):e0116459.
III
Kaihola, H., Gülen Yaldir, F., Hreinson J., Hörnaeus, K., Bergquist, J., Olivier. J., Sundström-Poromaa, I., Åkerud, H.
(2015) Effects of fluoxetine on human embryo development.
Manuscript.
IV
Söderholm, H., Olsson, A-K., Lavenius, E., Rönnstrand, L.,
Nånberg, E. (2001) Activation of Ras, Raf-1 and protein kinase C in differentiating human neuroblastoma cells after
treatment with phorbolester and NGF. Cell Signal 13:95-104.
Reprints were made with permission from the respective publishers.
Contents
Introduction ................................................................................................... 11 Antenatal depression and anxiety ............................................................. 11 Causes of antenatal depression ............................................................ 13 The effect of antenatal depression on pregnancy outcomes, fetal
development and offspring .................................................................. 14 Selective serotonin reuptake inhibitors (SSRIs) ....................................... 15 Serotonin and its role during embryo development ............................. 17 Serotonin and its role during placental development........................... 20 The effects of SSRIs on pregnancy and fetal development ................. 21 The effects of SSRIs on the offspring.................................................. 22 Treatment of mental health problems during pregnancy ..................... 23 The role of NGF in neuronal and non-neuronal development ................. 24 Signaling pathways downstream of the NGF receptor ........................ 24 Neuroblastoma cell line SH-SY5Y as a model system ............................ 26 Aims .............................................................................................................. 27 Materials and methods .................................................................................. 28 Material .................................................................................................... 28 Women and placentas (study I and II) ................................................. 28 Embryo collection (study III) .............................................................. 29 Cell culture and treatments (study IV)................................................. 29 Methods .................................................................................................... 32 Study I.................................................................................................. 32 Study II ................................................................................................ 32 Study III ............................................................................................... 34 Study IV ............................................................................................... 36 Statistics and bioinformatics .................................................................... 37 Ethical approvals (study I to III) .............................................................. 38 Summary of Results ...................................................................................... 39 Study I ...................................................................................................... 39 Study II ..................................................................................................... 45 Study III ................................................................................................... 47 Study IV ................................................................................................... 50 Discussion ..................................................................................................... 54 Methodological considerations................................................................. 54 Choice of control group ....................................................................... 54 Sample size .......................................................................................... 56 Human embryos ................................................................................... 57 Experimental considerations ................................................................ 57 The effects of SSRI and antenatal depression on placenta ....................... 58 ROCK1 and ROCK2 ........................................................................... 59 Trophoblasts, placenta and pregnancy complications ......................... 60 Developmental programming .............................................................. 62 Additive or separate effects? ............................................................... 63 Fluoxetine affects embryo development .................................................. 63 NGF signaling pathways .......................................................................... 64 Conclusions and remarks ......................................................................... 66 Summary of conclusions ............................................................................... 67 Sammanfattning på svenska (Summary in Swedish) .................................... 68 Acknowledgements ....................................................................................... 70 References ..................................................................................................... 74 Abbreviations
ACOG
BASIC
BDNF
BMI
cDNA
ECL
EGF
EPDS
ERK
FLX
GST-MEK
HPA
IHC
IPA
IVF
MAPK
MARCKS
MEK
NGF
NICE
PBS
PCR
PKC
qRT-PCR
Raf
Ras
RhoA
RNA
ROCK
SDS-PAGE
SSRI
TPA
TrkA
uPA
WB
The American college of obstetricians and gynecologists
Biology, affect, stress, imaging and cognition in pregnancy
and puerperium
Brain-derived neurotrophic factor
Body mass index
Complementary deoxyribonucleic acid (DNA)
Enhanced chemiluminescence
Epidermal growth factor
Edinburgh postnatal depression scale
Extracellular signal-regulated kinase
Fluoxetine
Glutathione S-transferase-MEK
Hypothalamus-pituitary-adrenal
Immunohistochemistry
Ingenuity pathway analysis
In vitro fertilization
Mitogen-activated protein kinase
Myristoylated alanine-rich C-kinase substrate
Mitogen/extracellular signal-regulated kinase
Nerve growth factor
The national institute of health and care excellence
Phosphate-buffered saline
Polymerase chain reaction
Protein kinase C
Quantitative reverse transcriptase-polymerase chain reaction
Rapidly accelerated fibrosarcoma
Rat sarcoma
Ras homolog gene family, member A
Ribonucleic acid
Rho-associated, coiled-coil-containing protein kinase
Sodium dodecyl sulphate-polyacrylamide gel electrophoresis
Selective serotonin reuptake inhibitor
12-O-tetradecanoylphorbol-13-acetate
Tropomyosin receptor kinase A
Urokinase-type plasminogen activator
Western blot
Introduction
In 2014, 10.4% of Swedish women of childbearing age (20 – 39 years) purchased prescribed antidepressant at least once. While the majority of these
women are not pregnant when treatment is initiated, some will find themselves in a situation where they need to consider whether or not to continue
treatment during a planned or unplanned pregnancy. The decision to continue treatment must be individualized, and be made together with the prescribing physician, and also be based on the best available evidence and riskbenefit assessments. While we have seen an increasing number of epidemiological reports on the side-effects of selective serotonin reuptake inhibitors
(SSRI) during the past 15 years, little is known about the biological effects
of this treatment during pregnancy. Thus, the major aim of this thesis is to
expand our knowledge about the effects of antidepressant treatment on placental function and embryo development.
Antenatal depression and anxiety
Worldwide, approximately 20% of pregnant women experience depressive
symptoms and 4 - 7% suffer from major depressive disorder [1-4]. Among
women with postpartum depression, almost 40% already had depressive
symptoms during pregnancy [5]. Besides the maternal and fetal outcomes
associated with depression, discussed further below, the most severe consequence of depression is suicide. Indeed, in the United Kingdom, the Eighth
Report of the Confidential Enquiries into Maternal Deaths [6] has consistently found mental health problems to be one of the leading causes of maternal
death, with over half of these deaths being caused by suicide. In Sweden a
similar pattern is noted, although it should be emphasized that maternal suicides predominantly occur in the postpartum period [7]. Out of 103 maternal
suicides reported between 1980 and 2007 in Sweden, only three occurred
during pregnancy (Esscher et al., in press).
However, major depressive disorder is not the only reason for women to
continue SSRI treatment during pregnancy, as these drugs are often used for
treatment of anxiety disorders as well. In addition, co-morbid depressive and
anxiety disorders are common during pregnancy [8], ultimately affecting the
choice of comparison group for this thesis, and the possibility of separating
the effects of anxiety and depression on fetal outcomes.
11
Unfortunately, anxiety disorders are far less studied than depression in
pregnancy, which is also one of the major reasons why this thesis focuses on
cases of depression. The prevalence of most anxiety disorders in pregnancy
appear similar to what is found at other times in women’s lives [8]; a population-based U.S. study reported a 13% one-year prevalence of any anxiety
disorder in pregnancy, which was comparable to non-pregnant women [9].
By comparison, the point prevalence of any anxiety disorder in a populationbased sample of Swedish pregnant women was 6.6% [2]. As to the prevalence rates of specific anxiety disorders, a recent systematic review reported
on wide variations across studies. For instance, generalized anxiety disorder
(GAD) was found in 0 – 10.5%, panic disorder in 0.2 – 5.7%, specific phobia in 3.2 – 19.9%, social phobia in 0.4 – 6.4% and posttraumatic stress disorder (PTSD) in 0 – 7.9% of pregnant women [8]. Generally, European and
U.S. studies contributed with prevalence rates in the lower range.
The gold standard for depression and anxiety diagnoses is the structured
psychiatric interview, with the Structured Clinical Interview of Diagnostics
and Statistical Manual of Mental (DSM) disorders (SCID) being the most
commonly used in research studies. However, for screening purposes during
pregnancy and in the postpartum period, most researchers and clinicians use
the Edinburgh Postnatal Depression Scale (EPDS) [10]. EPDS is a selfreporting questionnaire with 10 items [11], which has been translated into
Swedish and validated for use in the postpartum period [12] and during
pregnancy [13]. While the sensitivity of the instrument is not impressive,
typically ranging between 0.47 – 0.71 depending on cutoff, the specificity as
regards depressive disorders is excellent (pooled specificity 0.94 – 0.98)
[14]. The EPDS is implemented in clinical routine in Sweden and is typically
administrated at the first postnatal follow-up.
Untreated depression is strongly associated with poor quality of life, for
patients and their relatives, and increased mortality [15-18]. The World
Health Organisation predicts that major depressive disorder will be the most
serious disease worldwide in this century [19]. Depressive disorders are associated with heavy social and financial burdens [20, 21] and results in a
substantial public health disease burden [22]. Therefore, it is important to
identify the symptoms of depression, to be able to treat the disease and increase quality of life for the patient.
Typical symptoms of depression are fatigue, trouble sleeping, reduced
appetite, pessimistic thinking, anhedonia, reduced concentration and attention, as well as low self-esteem [23]. Important symptoms to notice are
thoughts of self-harm or life not being worth living, since these thoughts
increase the risk of suicide. In patients with more severe depression, psychomotor changes are noted. Both slowed psychomotor functioning, as well
as agitated psychomotor activity, can be seen [23].
“Biological” symptoms can sometimes be seen in depressed patients.
These symptoms, sometimes called melancholic, are e.g. weight loss, consti12
pation, loss of appetite, early morning waking, and diurnal mood variations,
with the worst feelings in the morning and feeling better later in the evening
[24]. Melancholic depression is a validated subgroup of depression; however, melancholic symptoms can be found in most depressed patients [25].
Causes of antenatal depression
Several risk factors for antenatal depression have been identified: prior history of depression or other mental health problems, low socioeconomic status,
being a single mother or a teenage mother, domestic violence, smoking, and
obesity are among the most common [26, 27]. In addition, the genetic set-up
of the mother, in combination with environmental factors like stress, life
experiences and previous reproductive/hormonal history, may also influence
the individual risk of developing depression during pregnancy [28].
Depression is associated with altered levels of monoamine neurotransmitters, especially noradrenalin and serotonin, in the brain. These altered levels
can be caused by a genetic predisposition, tissue damage, or, as in the case of
pregnancy, by altered hormone levels [29-31]. Clinical studies on neurotransmitter adaptations in the central nervous system during pregnancy are
rare, primarily due to a precautionary approach to the effects of imaging
techniques on the fetus. However, positron emission tomography studies
have revealed significantly increased monoamine oxidase A (MAO-A) activity (i.e. increased breakdown of serotonin, norepinephrine, and dopamine),
throughout all analyzed brain regions in the early postpartum period of
healthy women [32]. Likewise, studies on serum indicators of serotonergic
activity in healthy women a few days after delivery strengthen the idea that
the early postpartum is a state of serotonin deficiency [33-35]. Indirect
evidence of central nervous changes also include markedly decreased maternal serum levels of brain-derived neurotrophic factor (BDNF), both before
and after delivery, and at both time-points BDNF was correlated with decreased serotonin levels [35].
Both increased and decreased levels of cortisol have been associated with
depression [36, 37]. Stressful life events (e.g. parental loss, major childhood
stress or abuse, deteriorating physical health) can increase cortisol levels,
which lead to an increased vulnerability to depression [38]. At the end of the
pregnancy, the cortisol levels have increased three fold compared to those of
non-pregnant women. At the same time, the hypothalamus-pituitary-adrenal
(HPA) axis reactivity to stressful stimuli is dampened [39]. In addition, by
positive feedback from cortisol, the placenta also secretes large amounts of
corticotropin releasing hormone (CRH), resulting in 100 - 1,000-fold increased CRH levels in maternal serum during pregnancy [40]. After birth,
the cortisol and CRH levels drop dramatically, but the responsiveness of the
HPA axis will not return to normal until approximately 6 - 8 weeks postpartum [41, 42]. These perturbations in the HPA axis across pregnancy and
13
postpartum have repeatedly been held responsible for the mood symptoms
that may develop, but findings in pregnant (and postpartum) women are thus
far mixed [43]. For instance, previous research from our department has
indicated that women with antenatal depression have similar cortisol awakening response [44] and similar CRH levels (Hannerfors et al., submitted
manuscript) in comparison with healthy pregnant women.
The effect of antenatal depression on pregnancy outcomes, fetal
development and offspring
A number of negative effects of antenatal depression have been described in
the literature. However, when reading through the list of possible outcomes,
it should be emphasized that the association between exposure (antenatal
depression) and outcome may not always be as straightforward as individual
reports may claim. It is quite possible that risk factors such as smoking, obesity, drug or alcohol use, or domestic violence and abuse, which are common
in women with mental health problems, explain some of the effects that have
been attributed to the antenatal depression, as these comorbidities are also
risk factors for adverse obstetric and child outcomes. Thus, to separate the
effects of mental health problems in pregnancy from these confounders,
large sample sizes are needed. Furthermore, there may be additional factors
that researchers thus far have not paid attention to. For instance, Conroy and
colleagues [45] showed that children with behavioral disorders are predominantly found when the mother is suffering from comorbid depression and
personality disorder, whereas these problems were not found in children of
women with only depression. Also, poor outcomes in children occur predominantly in families living with socioeconomic difficulties, and high educational level in the depressed mother is, in fact, protective [46, 47]. Taken
together, these findings suggest that not only the depression per se, but also
other factors influence the outcome of a child.
Nevertheless, antenatal depression has been associated with an increased
risk of preeclampsia, preterm birth, decreased fetal head growth, impaired
fetoplacental function, and increased risk of neonatal complications [48-50].
Maternal depression has also been associated with low birth weight; however, this effect was predominantly found in low-income countries and in
countries with marked health inequalities [51]. Several behavioral problems
and developmental delays have been described in children exposed to antenatal depression (in utero). Developmental delays were found in 18-monthold children [49, 50]. Behavioral problems were found in both young and old
children, where children of 3 and 4 years of age had increased behavioral,
emotional and attention problems. Children aged 6 to 9 years had increased
levels of anxiety, and in adolescence, children exposed to antenatal depression showed emotional disorders like violent behavior [49, 50, 52]. Howev14
er, this association was only significant in girls, suggesting that there might
be a gender difference in how antenatal depression affects the offspring.
The obstetric and fetal outcomes in relation to anxiety disorder are sparsely studied, and, thus far, findings are mixed. A recent systematic review reported on six studies where premature birth or infant weight had been assessed in relation to anxiety disorders [8]. While no meta-analytic approach
was possible, the review concluded that half of studies found no influence on
these outcomes, whereas remaining studies did show some association with
premature birth and/or birth weight [8]. Even more preliminary, two small
studies, possibly on the same material, suggested lower BDNF [53] and
higher circulating tumor necrosis factor-alpha (TNFα) [54] levels in the cord
blood of newborns to mothers with GAD. Yet another study reported on
higher stress-induced cortisol levels (but similar behavioral response) in 7month old infants of prenatally anxious mothers [55]. Thus, given the commonly encountered comorbidity between depressive and anxiety disorders,
the long-term effects on offspring to depressed mothers may equally well be
a result of peripartum anxiety.
Selective serotonin reuptake inhibitors (SSRIs)
In the 1980s selective serotonin reuptake inhibitors (SSRIs) (Table 1) were
introduced and soon became the primary antidepressants used during pregnancy due to their good efficacy and relatively few side-effects [56]. With
time, SSRIs were found to be associated with rare but significant side effects, discussed below. As of today, 1 - 3% of European women use antidepressants during pregnancy [48, 57] and in the U.S. as many as 4 - 13% of
pregnant women are using antidepressants [58, 59]. In addition, Alwan and
colleagues [60] have shown that these numbers are increasing. In 1998, 2.5%
of pregnant women reported using antidepressants at some time during pregnancy. In 2005, this number had increased to 8.1% and the numbers are still
increasing. However, studies have shown that 75% of SSRI-treated women
in the U.S. discontinue antidepressant treatment either before pregnancy, at
discovery of pregnancy, or during the first trimester [59].
Serotonin (5-HT) is one of the neurotransmitters that regulate mood and
emotions, depending on how much of the protein is released into the synaptic cleft (Fig. 1). To regulate the levels of serotonin in the extracellular cleft,
the released serotonin is reuptaken to the presynaptic neuron by serotonin
transporters [61]. SSRIs inhibit the reuptake of serotonin, thereby increasing
the levels of serotonin in the synaptic cleft between two neurons (Fig. 1).
SSRIs cross the placenta and have been found in cord blood and amniotic
fluid [62-65], which means that the fetus is exposed to SSRIs during its development, either via the blood or by swallowing and inhaling the amniotic
fluid.
15
Table 1. SSRI substances and medical products available for prescription in Sweden
according to FASS (Farmaceutiska Specialiteter i Sverige). Number of prescriptions
of each substance to Swedish women in 2014 according to The National Board of
Health and Welfare in Sweden (shown as % of total SSRI prescriptions in Sweden)
Substance
Medical product
Citalopram
Cipramil, Citalopram
Oraline, Sertraline, Sertrone,
Zoloft
Cipralex, Premalex
Fluoxetine, Fontex
Paroxetine, Paroxiflex, Seroxat
Fevarin
Sertraline
Escitalopram
Fluoxetine
Paroxetine
Fluvoxamine
Prescriptions to women in
Sweden (%)
48.32
28.88
11.31
7.35
4.09
0.05
Figure 1. The function of SSRI on serotonin reuptake in the extracellular synaptic
cleft (Olivier et al. (2013) Front Cell Neurosci vol. 7:73, p. 1-15).
16
Serotonin and its role during embryo development
Embryo development
The development of a child is a nine month long process that begins with the
fertilization of an egg and ends at the delivery of the child. During this development, a number of processes and events have to occur in a structured
and chronological order. The development of an embryo, from the first cell
division to the formation of a blastocyst, has to follow certain critical steps.
Even if these initial events proceed according to plan, nine months remain
during which the embryo/fetus continues to develop, and is affected by the
surrounding environment in utero. The embryo development can be divided
into four periods: pre-embryo period (from fertilization until blastocyst formation); implantation period (day 6 until 7); post-implantation period (day 9
until week 4) when the organs and organic systems are developed; and fetal
period (from week 4 until birth) when the organic systems grow and mature
[66, 67].
After fertilization of the oocyte, the single cell divides into two cells and
continues to divide until it reaches compaction and forms the morula (Fig.
2). In the morula, a cavitation is formed and the embryo starts to expand into
a blastocyst with an outer layer of trophoblastic cells, the trophectoderm, and
an inner cell mass, the embryoblast (Fig. 2). The blastocyst is surrounded by
the zona pellucida, but at day 6 the blastocyst breaks out from the zona, an
event called hatching (Fig.2). By this time, the blastocyst has travelled
through the fallopian tubes and has reached the uterine cavity. The trophoblasts invade the outer cell layers of the endometrium to implant. Later on,
the trophectoderm develops into the placenta and the inner cell mass forms
the fetus and the amniotic cavity [68-70].
Scoring of embryo development
Endogenous embryonic gene expression first occurs between the four- to
eight-cell stages [71]. Based on studies within the in vitro fertilization (IVF)
field, the timing of the different embryo developmental stages has been
shown to be of importance. If the timing between embryo developmental
stages is too short, the quality of the embryo is negatively affected [72-76].
Neither is it beneficial for a proper embryo development, if the timing is too
long [73, 77]. For IVF, it is important to carefully select which embryo to
transfer back to the woman after embryo culturing. Therefore, the timing of
developmental stages has been carefully studied and a scoring system has
been developed [76, 78, 79].
To monitor development of an embryo, embryologists have started to use
a closed system consisting of an incubator with a time-lapse camera, e.g.
EmbryoScope® (Unisense FertiliTech, Denmark). Timing between cell divisions, compaction, cavitation and formation of blastocyst is analyzed, together with recording whether the embryo contains fragments reflecting cell
17
deaths or improper cell division. All these events are scored and a total final
score is set for each embryo. The inner cell mass and the trophectoderm are
scored from A (the best) to D (the worst), and the developmental stage is
scored from 1 to 6, where 6 refers to a hatched blastocyst [70, 76, 79]. The
best total score for an embryo is 6AA. Gardner and colleagues [80] found
that embryos with a score of 3AA had an implantation rate of 70%.
Figure 2. Pictures of embryo development monitored in an EmbryoScope®. A)
embryo at 4 cell stage, B) embryo at 8 cell stage, C) morula, D) beginning of cavitation (cav), E) blastocyst with trophectroderm (t) and inner cell mass (i), and F)
hatching.
Implantation window
One major event with high impact on a successful pregnancy rate is the implantation of the embryo, which occurs in the luteal phase in the menstrual
cycle in humans. This well-orchestrated cross-talk between the blastocyst
and the uterine endometrium is referred to as the window of implantation
[81]. The window of implantation consists of four major events, i.e. the blastocyst activation, uterine receptivity, blastocyst attachment and uterine decidualization, the latter meaning that the endometrial stroma undergoes cellular transformation to facilitate embryonic growth and invasion [81]. A
18
prerequisite for attachment to the uterine endometrium is hatching of the
blastocyst, in which the blastocyst bursts out of the zona pellucida. The
presence of an embryo in the uterus stimulates the endometrium to become
more receptive for embryo attachment [82]. During the window of implantation, estrogen and progesterone induce cells in the endometrium to proliferate and differentiate and to secrete proteins that affect trophoblast development. Also, Altmäe and colleagues [83] confirmed in their study that many
genes and signaling pathways are involved in human endometrium receptivity and the embryo-endometrium interaction. Some of the genes and pathways included are JAK-STAT signaling pathways, inflammatory pathways,
leukemia inhibitory factor (LIF), leptin (LEP), fibroblast growth factor 7
(FGF7), and heparin-binding EGF-like growth factor (HB-EGF) [82]. Once
the endometrium has become receptive to the embryo, the embryo attaches
to pinopodes in the endometrium [84] and the decidualization of the endometrium is initiated. Also, serotonin may have a role in the implantation
process. Mitchell and colleagues [85] showed that rats subcutaneously injected with serotonin had an impaired implantation due to suppression of
decidualization, indicating that serotonin has an impact on embryo implantation. After decidualization, the trophoblasts can invade the endometrium, the
embryo can become embedded, and the placenta is eventually formed [69].
Embryo susceptibility to teratogens
During the post-implantation period in the embryo development, when the
majority of the organs are developed, the embryo is most susceptible to teratogens [86]. Teratogens are substances, drugs or infectious agents, which
cause congenital malformations in the fetus. The timing of drug administration during pregnancy is crucial: use of drugs early in pregnancy can have
severe effects on the offspring. Administration later in pregnancy reduces the
risk of congenital malformations, but there is a risk of toxicological or pharmacological effects on the fetus. Sometimes, the combination of two drugs
can increase the teratogenic effects and there may also be a correlation between drug dose and severity of malformations [86].
There is evidence that the effects of teratogens on individual humans vary
considerably [87] and there have been attempts to elucidate the mechanisms
behind the individual differences. Genetic predisposition for increased susceptibility has been suggested [88] as well as genetic polymorphisms of placental transporter proteins [89]. Unfortunately, the available data are often
conflicting and methodological tools need further development.
Serotonin and its role for embryo development
Serotonin is found in follicular and fallopian tube fluid (at least in female
rats) [90, 91]. Serotonin transporters and tryptophan hydroxylase 2, the enzyme responsible for serotonin synthesis, are expressed as early as in twocell mice embryos [92, 93]. In addition, expression of some serotonin recep19
tors has been demonstrated in mouse preimplantation embryos, such as 5HT1D and 5-HT7, but thus far, none of the 5-HT2 receptors have been identified [94]. The exact role of serotonin at this stage is not known; it has merely
been speculated that the serotonin transporter expressed in embryos is important for transport of serotonin from surrounding fluid. During embryogenesis, high and low levels of serotonin can have both positive and negative
effects on embryo development, depending on the stage of development.
Mouse embryos exposed to serotonin, from 4-cell stage until blastocyst,
showed significantly reduced number of cells and there was an increased
incidence of dead cells [95]. However, Khozhai and colleagues [96] showed
that, by reducing the levels of maternal serotonin by administration of a tryptophan hydroxylase inhibitor to pregnant mice, the development of mouse
preimplantation embryos was impaired. Altered expression of different Gprotein coupled serotonin receptors during the preimplantation period could
explain these differences in response to serotonin, since the different serotonin receptors could activate different signaling pathways at different developmental stages [94]. Kim and colleagues [97] treated mouse 2-cell stage
embryos with fluoxetine, a selective serotonin reuptake inhibitor, and found
that embryos exposed for a short period to low-dose fluoxetine had an increased percentage reaching blastocyst stage compared with controls. This
was probably mediated through activation of Ca2+/calmodulin-dependent
protein kinase II (CaMKII)-dependent signaling pathways. Also, long-term
exposure decreased mouse early embryo development through inhibition of
the potassium channels TREK-1 and TREK-2. This indicates that both the
exposure time and the developmental stage of the embryo at exposure, has
an influence on mouse embryo development. However, studies in human
embryos are still lacking.
Serotonin and its role during placental development
The placenta is an organ that develops during pregnancy. It protects and
provides the fetus with nutrients, proteins and hormones to secure normal
fetal development. The placenta serves as an endocrine organ throughout
fetal development and synthesizes and secretes a variety of steroid and protein hormones, including estrogen, progesterone, human chorionic gonadotropin, and placental lactogen. In addition, it also produces and secretes a
number of substances important for the development of the fetal brain [98].
During placentation, the trophoblasts of the blastocyst invade the epithelial cells of the uterus [68]. This step is of great importance since dysregulation and improper implantation can lead to miscarriage [99]. From the myometrium, spiral arteries stretch towards the intervilluos space of the placenta [68]. Early in pregnancy, the spiral arteries are composed of smooth muscle cells. Later on in pregnancy, cytotrophoblasts from the fetal part of the
placenta invade the spiral arteries, making them more dilated and less sensi20
tive to changes in blood pressure. Improper invasions of trophoblasts are
involved in e.g. preeclampsia, where the spiral arteries have kept some of
their ability to respond to changes in blood pressure [100].
Serotonin is important for placentation [101] and acts as a vasoconstrictor
in the placenta [102]. Oufkir and colleagues [101] demonstrated that activation of the serotonin receptor 5-HT2A in trophoblast cells activates intracellular signals that are known to regulate survival, differentiation, migration and
invasion, suggesting a role for serotonin in placentation. This fits well with
the suggestions that serotonin plays a role in preeclampsia [103] and in the
pathophysiology of gestational diabetes [104].
During brain development, serotonin acts as a neurotrophic factor regulating e.g. cell division, differentiation and synaptogenesis [105]. The placenta
has been shown to serve as a serotonin source and serotonin produced by the
placenta is accumulated in the fetal forebrain where it is involved in fetal
brain development [106].
The effects of SSRIs on pregnancy and fetal development
As for antenatal depression, the reported risks associated with SSRI use during pregnancy need to be interpreted with caution. Most epidemiological
studies rely on registers, but these registers rarely include information on
health behaviors in pregnancy, and thus, important confounders may be lacking in the analyses. Furthermore, pharmaco-epidemiological studies need to
consider confounding by indication (i.e. the underlying effects of the psychiatric disorder) when examining adverse effects of SSRIs. This is typically
accomplished either by defining a comparison group who have discontinued
SSRI use prior to pregnancy or by using women with untreated psychiatric
disorders for comparison. In addition, in most epidemiological research
SSRI exposure relies on filed prescriptions, but women may not always use
their medication as prescribed, ultimately resulting in misclassification bias.
These problems were elegantly demonstrated by a recent Danish study on
the risk of miscarriage in SSRI-treated women. With previous reports suggesting an increased risk for miscarriage in SSRI users [107, 108], the Danish study came to quite different conclusions. Being able to adjust for underlying psychiatric diagnoses, the study found that women who discontinued
SSRI treatment had higher risk of miscarriage than those who continued
treatment and non-users. While the SSRI users also had an increased risk of
miscarriage, this finding no longer remained when psychiatric diagnoses
were adjusted for. In fact, women on SSRI who had no psychiatric diagnosis
(suggesting that they were treated in primary health care rather than in psychiatric care) had lower risk of miscarriage in comparison with untreated
women who had a psychiatric diagnosis [109]. Furthermore, the risk for
second trimester miscarriages were the same regardless of whether the depressed women were using SSRI or not [109]. Similarly, no risk for still21
birth, neonatal mortality or postneonatal mortality has been noted in SSRI
users [110].
SSRI treatment during pregnancy has been associated with preterm birth
[59, 111-113], decreased birth weight (or small for gestational age), reduced
fetal head growth, and impaired fetoplacental function [48, 114-118]. However, there are also several studies that show no effect of SSRI on preterm
birth and birth weight [113, 119, 120], suggesting that it is the depression
per se and/or other associated behaviors or conditions, such as smoking,
obesity or drug abuse, that causes these outcomes. Of specific relevance for
this thesis, an association with preeclampsia has also been described [116,
121-123], which appears relevant in relation to the pathophysiology of the
disorder, i.e. increased risk if treatment is continued into the second trimester
in comparison with those who discontinued during the first trimester. However, it should be emphasized that risk estimates for development of
preeclampsia are higher for serotonin and noradrenaline reuptake inhibitors
(SNRI) and tricyclic antidepressants than for SSRI monotherapy [122].
The effects of SSRIs on the offspring
Infants exposed to SSRIs during pregnancy run the risk of poor neonatal
adaption, such as jitteriness, convulsions, respiratory distress, hypoglycaemia and feeding difficulties, also referred to as the neonatal adaptation syndrome (reviewed in [49, 124]). There are several studies showing that infants
antenatally exposed to SSRIs more often need neonatal intensive care and
have a higher rate of neonatal seizures [115, 117, 125].
Other, less frequent, effects of antenatal SSRI exposure are physiological
malformations. There have been several reports on the association between
SSRI exposure and congenital malformations like omphalocele, anencephaly
and craniosynostosis [126-129]. Also, cardiac malformations have been
found in infants exposed to SSRIs in utero [120, 130]. However, risks of
congenital anomalies in association with the use of SSRIs (or other drugs)
might be underestimated if elective terminations of pregnancies are not taken
into account. Kieler and colleagues [131] recently addressed this concern,
but found no association between the use of SSRIs and elective termination
of pregnancy for fetal anomalies. As expected, induced abortion due to maternal ill health or socio-economic disadvantage was more common in women using antidepressants.
Another rare condition found in infants prenatally exposed to SSRI, is
persistent pulmonary hypertension (PPHN) [57, 114, 127, 132]. In PPHN,
the pulmonary vasculature does not relax after birth, which leads to hypoxemia. There is significant evidence of an increased risk of PPHN due to
SSRI exposure, but the actual risk difference between exposure and nonexposure is very small (an increase of 2 per 1000) [14].
22
Later in life, children exposed to SSRIs during pregnancy are at greater
risk of developing behavioral disorders; like autism spectrum disorders [133,
134]. Externalizing behavior has been found in 4-year-old children after
antenatal exposure to SSRI [135], but Oberlander and colleagues later also
reported on internalizing behaviors in 3- and 4-year-old children following
SSRI exposure [136]. Again, there are also studies suggesting no association
between prenatal SSRI exposure and internalizing behaviors [137].
Treatment of mental health problems during pregnancy
International guidelines for treatment of antenatal health problems are available. The American College of Obstetricians and Gynecologists (ACOG)
published their recommendations in 2008 [138], and The National Institute
for Health and Care Excellence (NICE) recently published their updated
recommendations on clinical management and service guidance for women
with antenatal and postnatal mental health problems [14]. Both guidelines
stress the importance of preconception counselling, and the importance of
integrated care plans for women who present in antenatal care with mental
health problems. In addition, both guidelines emphasize that the potential
risk of SSRI use in pregnancy must be considered in the context of the risks
associated with untreated mental health problems and the risk of relapse of
depression if treatment is discontinued in pregnancy. Treatment with SSRIs
or other antidepressants during pregnancy should be individualized. However, it is interesting that the ACOG guidelines strongly recommend that paroxetine use in pregnant women and women planning pregnancy should be
avoided.
The NICE guidelines, being issued by psychiatrists, are more detailed in
their treatment recommendations and emphasize the role of high-intensity
psychological interventions. For women with mild to moderate depression,
psychological intervention is the first line of treatment. Pharmacological
treatment is only recommended for women with moderate or severe depression if the woman has expressed a preference for medical treatment, or declines psychological treatment, or has not responded to psychological treatment. However, the NICE guidelines suggest that in women with a history of
severe depression who present even with mild symptoms of depression in
pregnancy, pharmacological treatment should be considered. As to choice of
antidepressant treatment, NICE is less specific than ACOG but stresses that
the drug with lowest risk profile and at the lowest effective dose should be
used (taking into account that dosages may need to be adjusted during pregnancy). In line with ACOG, single drug treatment is advocated. NICE guidelines also mention the risk of neonatal adaptation syndrome in babies exposed to paroxetine and venlafaxine.
23
The role of NGF in neuronal and non-neuronal
development
In the 1950s, Levi-Montalcini discovered a protein later called the nerve
growth factor (NGF), reflecting its ability to stimulate growth and differentiation of neuronal cells [139].
The most obvious role of NGF is in neurogenesis and neuritogenesis (reviewed in [140]), but NGF is also found in a number of other tissues, suggesting a role in other non-neuronal processes like angiogenesis [141] and
immune response [142].
Ovarian production of NGF is involved in folliculogenesis, where it
seems to be important for a proper ovulation to occur [143-145]. But, an
excessive production of NGF has been shown to have an opposite effect,
where mice with an overproduction of NGF are sub-fertile [146].
During pregnancy, NGF is expressed in the placenta and plays a crucial
role in placentation and pregnancy maintenance [147]. NGF expression was
also found in the maternal decidua and the decidual vessels, indicating a role
for NGF in implantation and formation of vessels in the decidua [147]. Also,
there are studies showing that NGF is involved in stress-induced abortion
and preterm birth [148, 149].
Signaling pathways downstream of the NGF receptor
NGF binds to two different receptors, tropomyosin receptor kinase A (TrkA)
and p75 [150-152]. Upon binding to TrkA, the receptor becomes autophosphorylated on several tyrosine residues, initiating a number of signaling cascades. In this thesis, focus is on the rat sarcoma (Ras)-rapidly accelerated
fibrosarcoma (Raf)-mitogen-activated protein kinase (MAPK) and Rasrelated C3 botulinum toxin substrate (Rac)-Ras homolog gene family member A (RhoA) pathways (Fig. 3).
When TrkA is phosphorylated on tyrosine residue 490, Src homology 2
containing (SHC) protein is bound to the receptor and forms a complex with
son of sevenless (SOS) and growth factor receptor-bound protein 2 (Grb2)
(Fig. 3) [153]. This initiates a signaling cascade that involves Ras, Raf, mitogen/extracellular signal-regulated kinase 1/2 (MEK1/2) and extra-cellular
signal-regulated kinase 1/2 (ERK1/2), which leads to differentiation and
survival of a cell [154-156].
Another pathway down-stream of TrkA is the Rac-RhoA signaling pathway [157, 158], where RhoA further activates Rho-associated, coiled-coilcontaining protein kinase 1/2 (ROCK1/2) [159, 160].
ROCK1/2 can also be activated via Ras-Raf-MAPK [161], which is then
mediated via ribosomal S6 kinase (RSK) [162, 163].
NGF also cross-talks with other signaling pathways. Both vascular endothelial growth factor (VEGF) and its receptor (VEGFR) are up-regulated by
24
Figure 3. Schematic figure on NGF signaling pathways with focus on Ras-RafMAPK and Rac-Rho pathways.
25
over-expression of NGF in brown adipose tissue in mice and this upregulation induces angiogenesis [164].
Takei and Laskey [165] found that NGF induces expression of tumor necrosis factor-α (TNFα) in the undifferentiated SH-SY5Y neuroblastoma cell
line. The increased TNFα expression suppresses the NGF-induced ERK
activation, thereby blocking NGF-induced differentiation of the cells. Also,
mouse macrophages treated with NGF induces TNFα expression and this
increased production was mediated via the MAPK-ERK pathway [166].
In human monocytes, the NGF-receptor TrkA, the epidermal growth factor receptor (EGFR) and the formyl peptide receptor (FPR) cross-talks via
e.g. ERK when modulating pro-inflammatory mediators [167].
Raf-1 can also be activated by the phorbolester 12-O-tetradecanoylphorbol-13-acetate (TPA) in a protein kinase C (PKC)-dependent manner
[168, 169] and NGF-induced neurite outgrowth has been shown to involve
PKCδ [170]. PKC can also increase the activation of Ras, which causes an
activation of Raf-1[171].
One of PKC’s substrates is myristoylated alanine-rich C-kinase substrate
(MARCKS) [172, 173], which is a protein important for normal cerebral
development in mice [174]. Since this protein is endogenously expressed in
cells, measurement of phosphorylation of MARCKS in vivo is a commonly
used method to measure the activation of PKC.
Neuroblastoma cell line SH-SY5Y as a model system
Neuroblastoma is a tumor primarily found in children. It consists of immature neuronal cells that have stopped differentiating and started to proliferate
instead [175]. The neuroblastoma cells can be arrested at different stages
during development, which is the reason why there are different stages of
neuroblastoma found in children and some which spontaneously regress
without treatment [176]. Several neuroblastoma cell lines have been established over the years and they make perfect models for studying neuronal
development, since they can be stimulated to differentiate by different treatments [175].
In study IV, the neuroblastoma cell line SH-SY5Y was used. These cells
can be induced to differentiate by treatment with the phorbolester TPA
[177]. Even though SH-SY5Y cells are of a neuronal phenotype, they do not
differentiate when treated with NGF, possibly due to the low expression of
the NGF receptor TrkA [178]. When transfecting SH-SY5Y cells with human exogenous TrkA, the cells respond to NGF treatment by formation of
neurites [179].
26
Aims
The main aim of this thesis was to investigate whether SSRI treatment during pregnancy has an effect on human placenta and early embryo development. The specific aims in each study were:
I.
To investigate the impact of antenatal depression and antidepressant
treatment during pregnancy on gene expression in the fetal placenta.
II.
To investigate the independent influence of SSRI treatment and antenatal depression on placental proteins in the NGF signaling pathway.
III.
To evaluate whether fluoxetine exposure influences the timing of
different embryo developmental stages, and furthermore, to analyze
what protein networks are affected by fluoxetine in the early development of an embryo.
IV.
To investigate whether treatment of SH-SY5Y/TrkA cells with TPA
and NGF leads to activation of Raf-1 and, if so, to investigate the involvement of the upstream signaling molecules Ras and PKC.
27
Materials and methods
Material
Women and placentas (study I and II)
Placental tissue was collected from women participating in an ongoing longitudinal study on antenatal and postpartum depression called the BASIC
(the Biology, Affect, Stress, Imaging and Cognition in Pregnancy and the
Puerperium) project. All women attending the routine ultrasound at gestational week 16-18 at Uppsala University Hospital are asked to participate in
the BASIC study, which results in a population-based cohort. Oral and written information was given and an informed written consent was obtained.
The women filled out web-based questionnaires at gestational age 17 and 32
weeks, including questions on physical and socio-demographic characteristics, medical, psychiatric, gynecologic and obstetric history variables, lifestyle, medication parameters, and the Swedish version of the Edinburgh
Postnatal Depression Scale (EPDS). Information concerning maternal depression, SSRI use, delivery and neonatal outcomes were retrieved from the
medical records.
For the specific aim of these sub-studies, the inclusion criteria were women of Western European descent, normal pregnancies, normal deliveries and
healthy offspring (no diagnoses and no admittance to neonatal care). Exclusion criteria were smoking or alcohol use during pregnancy, any daily use of
prescribed drugs during pregnancy, any other chronic conditions or disease,
gestational age <35 weeks, and maternal age <18 or >42 years. Depressed
women had EPDS scores corresponding to major depression, in combination
with medical records confirming depression and ongoing treatment for their
depression in terms of psychotherapy. Women on SSRI treatment used their
treatment during the pregnancy in clinically relevant doses, i.e. low-dose use
was excluded.
In study I, five women with ongoing depression, five women on SSRI
treatment and ten women with normal pregnancies were included for the
microarray analysis. The control group was divided into two groups and was
matched with depressed or SSRI-treated women, respectively, by age, body
mass index (BMI) and gestational length on an individual level.
28
For the validation of the microarray analysis in study I, a larger number of
women was included in each group: 24 depressed women, 29 SSRI-treated
women and 31 healthy pregnant women (Table 2). All the women from the
microarray analysis were included in the study groups. The depressed women had an EPDS score of >12 in gestational week 17 and 32 (except for one
woman with EPDS >14 at one time point).
In study II, there were 12 women in the depressed, SSRI-treated and
healthy control groups, respectively (Table 3). The depressed women had
EPDS scores of 13 or higher in gestational weeks 17 and 32, together with
hospital records confirming major or minor depressive disorder and ongoing
treatment in terms of psychotherapy. The groups were matched on maternal
age.
Placental biopsies were collected at delivery from two different locations
on the placenta and all the way through the placenta, containing both the
maternal and fetal side. The biopsies were rinsed in cold sterile phosphatebuffered saline (PBS), frozen on dry ice within 60 minutes after delivery and
stored at -70 ºC until further use.
Embryo collection (study III)
Couples that had undergone IVF treatment at the Center for Reproduction in
Uppsala, Sweden, were asked to donate surplus embryos that would have
been destroyed after 5 years of cryopreservation, according to Swedish law.
A written informed consent was obtained from the couples donating the embryos.
Before freezing, the embryos had been inseminated with sperms and, after
evaluation of fertilization, zygotes with 2 pro-nuclei were selected for further
culturing in a humidified incubator.
Cell culture and treatments (study IV)
SH-SY5Y cells transfected with full-length human TrkA (SH-SY5Y/TrkA)
were cultured under ordinary conditions. For experiments on serum-deprived
cells, the cells were cultured in serum-free SHTE medium.
SH-SY5Y/TrkA cells were treated with 100 ng/mL NGF (Promega Corp.,
US) and 16 nM TPA (Sigma-Aldrich Corp., US) with or without serum.
Treatment with 4 µM GF109203X (PKC inhibitor, Boehringer Mannheim
GmbH, Germany) was performed 30 minutes before the addition of NGF or
TPA.
29
125 ± 11
79 ± 8
Systolic blood pressure at last visit, mmHg
Diastolic blood pressure at first visit, mmHg
8.7 ± 5.3bd
30
2.9 ± 1.8
EPDS score week 17
EPDS score week 32
2.9 ± 1.8
15.9 ± 3.6a
a
significantly higher than all other groups, P<0.001, ANOVA post hoc Bonferroni
b
significantly higher than healthy control group, P<0.001, ANOVA post hoc Bonferroni.
c
significantly lower than healthy control group, P<0.01, ANOVA post hoc Bonferroni.
d
significantly lower than depressed group, P<0.001, ANOVA post hoc Bonferroni
45
7.7 ± 4.9b
281 ± 7
Gestational length
15.0 ± 4.2a
54
3589 ± 400
11.0 ± 0.9c
77 ± 7
125 ± 10
70 ± 7
118 ± 12
27.2 ± 4.9
1 (0-3)
31.2 ± 4.1
SSRI-treated women
(n=29)
273 ± 8c
65
3546 ± 499
11.4 ± 0.8
76 ± 6
119 ± 11
69 ± 7
111 ± 12
24.0 ± 6.2
1 (0-2)
31.1 ± 4.3
Depressed women
(n=24)
275 ± 11
Gender offspring (% boy)
3577 ± 351
73 ± 9
Diastolic blood pressure in first trimester, mmHg
Birth weight (gram)
119 ± 13
Systolic blood pressure in first trimester, mmHg
11.7 ± 0.8
26.0 ± 4.9
BMI (kg/m )
Lowest hemoglobin level during pregnancy, g/dl
0 (0-3)
2
31.4 ± 3.9
Parity (n, median, range)
Age (years)
Healthy controls
(n=31)
Table 2. Demographic data on the depressed, SSRI-treated and healthy women included in the qRT-PCR validation of the microarray results.
Data are presented as mean ± SD or median (min – max)
3.0 (0.0 – 5.0)
11.5 (3.0 – 24.0)***, a
EPDS gestational week 17
EPDS gestational week 32
3.5 (1.0 – 6.0)
16.0 (11.0 – 23.0)***
* p < 0.05, significantly different in comparison with controls, Pearson Chi-Square test
** p < 0.01, significantly different in comparison with controls, Mann-Whitney U test
*** p < 0.001, significantly different in comparison with controls, Mann-Whitney U test
a
p < 0.05, significantly different compared with depressed, Mann-Whitney U test
3510 (3170 – 4230)
8 / 4 (66.7% / 33.3%)
272 (263 – 284)**
1 (8.3%)
2 (16.7%)
101.2 (94.0 – 113.0)
93.0 (85.0 – 102.0)
27.0 (22.7 – 35.8)a
8 (66.7%)*
29.0 (25.0 – 35.0)
SSRI-treated women
(n = 12)
10.5 (0.0 – 20.0)**, a
3730 (2890 – 4540)
5 / 7 (41.7% / 58.3%)
277 (263 – 293)
0
1 (8.3%)
99.5 (89.5 – 110.0)
85.2 (77.5 – 104.0)*
24.3 (18.2 – 45.4)
10 (83.3%)*
31.5 (26.0 – 36.0)
Depressed women
(n = 12)
17.0 (4.0 – 24.0)***
3520 (3110 – 4080)
Birth weight, grams
282 (272 – 289)
5 / 7 (41.7% / 58.3%)
Sex of child, n of girls/boys (%)
Gestational length, days
0
IVF treatment
103.8 (87.5 – 120.0)
MAP partus
0
97.5 (82.5 - 112.0)
MAP first trimester
Smokers, n (%)
24.2 (20.1 – 31.7)
3 (25.0%)
28.5 (25.0 – 33.0)
BMI
Parous women, n (%)
Age, year
Healthy controls
(n = 12)
31
Table 3. Demographic data on the depressed, SSRI-treated and healthy control women included in study II. Blood pressure is shown as mean
arterial pressure (MAP). Data are presented as median (min – max)
Methods
Study I
RNA isolations from placenta
A 3 mm3 cube was taken from the fetal side of the placenta biopsies.
For isolation of total ribonucleic acid (RNA) for the microarray analysis,
miRNeasy mini kit (Qiagen, Germany) was used. The tissue was lysed with
QIAzol (Qiagen, Germany) using a rotor-stator homogenizer and chloroform
was added for phase-separation.
Total RNA isolation for the quantitative reverse transcriptase-polymerase
chain reaction (qRT-PCR) analysis was done similarly, except that RNeasy
mini kit (Qiagen, Germany) was used and tissue homogenization was performed by using stainless steel beads in a TissueLyser (Qiagen, Germany).
After phase-separation, the protocol in the miRNeasy mini kit and RNeasy mini kit handbooks was followed. The RNA quality was determined
using the Agilent 2100 Bioanalyzer system (Agilent Technologies Inc, US).
Microarray analysis
250 ng of total RNA was used to generate amplified and biotinylated sensestrand complementary DNA (cDNA) from the entire expressed genome using Ambion WT Expression kit and Affymetrix GeneChip® WT Terminal
Labeling and Hybridizaton User Manual (Affymetrix Inc., US). GeneChip®
ST Arrays were hybridized and, after the incubation, the arrays were
washed, stained and scanned using the GeneChip® Scanner 3000 7G.
cDNA synthesis and qRT-PCR analysis
From 250 ng of total RNA, 20 µL of cDNA was synthesized using random
primer and SuperScriptIII reverse transcriptase (Invitrogen, US) according to
the manufacturer’s protocol.
qRT-PCR was performed using the StepOne Plus qPCR machine (Applied Biosystems, Life Technologies, US). For normalization, GAPDH and
β-actin were selected as reference genes. TaqMan Gene Expression Assay
primers, probes and master mix (all Applied Biosystems, Life technologies,
US) were used and the qRT-PCR was run according to the manufacturer´s
protocol.
Study II
Protein isolation
Pieces were taken from the fetal side of the placenta biopsies and total protein lysates were prepared using RIPA lysis buffer (Sigma-Aldrich Corp.,
32
US) containing Protease inhibitor cocktail (Sigma-Aldrich Corp., US),
PMSF and ortovanadate. The biopsies were homogenized and, after centrifugation, the supernatants containing the total protein lysates were collected.
The proteins were separated by NuPage Novex 4-12% Bis-Tris gels (Invitrogen, Life Technologies, US).
Western blot (WB)
After separation, the proteins were transferred to Immobilon-FL PVDF
membrane (Merck Millipore, US). The proteins were detected by protein
specific primary antibodies (Table 4). Protein levels were normalized against
β-actin. Secondary antibodies were labelled with IRDye 680RD and IRDye
800CW, which enables detection of two proteins at the same time using Odyssey Image Scanner (LI-COR Biosciences Inc., US).
Immunohistochemical staining (IHC)
Paraffin-embedded placental biopsies were sectioned into 5 µm slices and
mounted on slides. After deparaffinization and rehydration, antigenic retrieval was performed by heating the slides in citric acid. The sections were
fixated using an ice-cold acetone/methanol mix and incubated against endogenous peroxidase activity. Non-specific binding was blocked by incubating the sections in PBS containing horse or goat serum. Proteins of interest
were detected using primary antibodies (Table 4). After incubation with
biotinylated secondary antibodies, the sections were incubated with avidinD
conjugated horseradish peroxidase, followed by staining/developing using
liquid 3,3’-diaminobenzidine tetrahydrochloride (DAB) + substrate chromogen system (Dako, Denmark), and counterstaining with Mayer hematoxylin
(HistoLab Products, Sweden). The sections were dehydrated and mounted
under cover glass.
Scoring of the staining intensity was done visually in a light microscope
(Axio Observer.Z1) (Carl Zeiss AG Corp., Germany) using a scale from 1 to
4, where 1 corresponds to the lowest intensity and 4 the highest. A doubleblinded validation of the scoring was performed: a second person scored the
slides in the same way as the first scorer, without knowing the initial results.
33
Table 4. Primary antibodies used in Western blot and immunohistochemistry on
placenta material
Target
Catalogue no
Commercial supplier
NGF
sc-548
SantaCruz Biotechnology Inc., US
Used for WB
and/or IHC
WB and IHC
TrkA
06-574
Upstate, Merck Millipore, US
WB and IHC
pTrkA
(pTyr490)
pTrkA
(pTyr496)
Raf-1
ab1445
Abcam plc, England
WB
sc-7987-R
SantaCruz Biotechnology Inc., US
WB
sc-133
SantaCruz Biotechnology Inc., US
WB and IHC
pRaf-1
(pTyr340/341)
RhoA
sc-16806
SantaCruz Biotechnology Inc., US
IHC
sc-418
SantaCruz Biotechnology Inc., US
WB and IHC
ROCK1
sc-5560
SantaCruz Biotechnology Inc., US
WB and IHC
ROCK2
sc-1851
SantaCruz Biotechnology Inc., US
WB and IHC
pROCK2
(pSer1366)
β-Actin
PA5-34895
Thermo Scientific Inc., US
IHC
sc-47778
SantaCruz Biotechnology Inc., US
WB
Study III
Embryo culturing and developmental scoring
Cryopreserved two-day embryos were thawed and transferred to equilibrated
culture medium used for human cleavage stage embryos (CCM) (Vitrolife,
Sweden), which were the culturing conditions for control embryos. For
fluoxetine (FLX) treatment of the embryos, FLX (Sigma-Aldrich Corp., US)
was added to the culture medium to reach a final concentration of 0.5 µM.
The FLX dose was chosen based on levels found in cord blood and amniotic
fluid of FLX-treated pregnant women.
The embryos were cultured in an EmbryoScope® (Unisense FertiliTech,
Denmark) with a time-lapse camera taking images every 15 minutes. The
embryo development and quality was evaluated and scored retrospectively
using standard morphological criteria developed by several research groups
[70, 76, 78, 79].
34
Bottom-up shotgun proteomics of embryos
Embryos were snap frozen after culturing and dissolved in 2 M thiourea/6 M
urea in ammonium bicarbonate for lysis and protein extraction. Proteins
were reduced with dithiothreitol (DTT) and alkylated with idoacetamide
(IAA) before enzymatic digestion, first by Lys-C (Wako Chemicals GmbH,
Germany) and then by trypsin (Promega, US). The peptides were solid phase
extraction purified by ZipTips® (Merck Millipore, US), dried in a
SpeedVac® system, and analyzed by high resolution tandem mass spectrometry.
The analysis were performed using a QExactive Plus Orbitrap mass spectrometer (Thermo Fisher Scientific, Bremen, Germany) after on-line separation of the digested peptides by reversed phase liquid chromatography. A
150 min. linear gradient from 4% to 100% acetonitrile at 250 nL/min was
used to elute the peptides.
Proseek Multiplex Immunoassay analysis on embryo culture medium
For the detection of proteins secreted into the culture medium, 20 µL of the
culture medium was snap frozen after embryo culturing. The protein analysis
was performed using the Proseek Multiplex Immunoassay analysis with the
Inflammation I96x96 panel. The analysis method is based on the Proximity
Extension Assay technology and was run at Olink Bioscience facilities in
Uppsala, Sweden.
Immunohistochemical staining
After culturing, the embryos were fixed in 2.5% paraformaldehyde (PFA) in
PBS and stored in PBS/polyvinylpyrrolidon (PVP) until later. The embryos
were permeabilized in 0.25% Triton X-100 in PBS/PVP, blocked in blocking
solution containing bovine serum albumin and Tween20 and incubated with
primary antibodies against urokinase-type plasminogen activator (uPA) and
NGF (both from SantaCruz Biotechnology Inc., US). The embryos were
incubated with secondary antibodies labeled with AlexaFluor488 or
AlexaFluor594 (Life Technologies, Thermo Scientific Inc., US), which
makes it possible to detect two proteins at the same time. After incubation,
the embryos were mounted on a slide in 5 µL VectaShield (Vector Laboratories Inc., US), surrounded by Vaseline, covered by a cover slip and sealed
with nail varnish.
The stainings were evaluated using a fluorescence microscope (Axio Observer.Z1, Carl Zeiss Ag Corp., Germany).
35
Study IV
Mobility shift assay of Raf-1
Total protein lysates from SH-SY5Y/TrkA cells treated with 100 ng/mL
NGF or 16 nM TPA were separated by 7% sodium dodecyl sulphatepolyacrylamide gel electrophoresis (SDS-PAGE). Raf-1 was detected by
Western blot using primary antibody specific for Raf-1 (C-12, Santa Cruz
Biotechnology Inc., US). The protein was visualized using enhanced chemiluminescence (ECL) (Amersham, UK).
Immunoprecipitation of Raf-1 and in vitro kinase assay
Total protein lysate from NGF- or TPA-treated SH-SY5Y/TrkA cells was
incubated with Raf-1 antibody (C-12, SantaCruz Biotechnology Inc., US)
and, after incubation, the antibodies were bound to protein A Sepharose
(Pharmacia, Sweden). The immunocomplexes were washed and incubated in
a kinase buffer containing glutathione S-transferase (GST)-MEK1 substrate
(Upstate Biotechnology Inc., US, and Dr Stephen Pelech, Kinetek Pharmaceuticals, US) and 5 µCi [γ-32P] ATP (Amersham, UK). The samples were
separated by 7% SDS-PAGE and phosphorylated GST-MEK1 was visualized by autoradiography and quantified using phosphorimage analysis (Fuji
Bas 2000).
Ras activity measurement
SH-SY5Y/TrkA cells were treated with NGF or TPA and total protein lysates were prepared. The Ras-binding domain (RBD) of Raf-1 (kindly provided by Dr Julian Downward, Imperial Cancer Research Fund Lab. London
UK), expressed as a GST fusion protein, was used to bind active Ras (RasGTP). The precipitates were separated by 12.5% SDS-PAGE and Ras was
detected using Western blot and anti-Ras antibody (Ro2120, BD Transduction Laboratories™, BD, US). Total amount of Ras was analyzed in total
lysates from the same samples and detected using the same Ras antibody.
Phosphorylation of the PKC substrate MARCKS
Serum-starved SH-SY5Y/TrkA cells were incubated with [32Pi] in phosphate-free Eagle’s medium containing bovine serum albumin for 4 h before
treatment with NGF or TPA. The incubation was terminated by washing the
cell cultures with ice-cold TRIS-buffered saline (TBS). Protein lysates were
boiled in Triton-X100 and separated by 8% SDS-PAGE. The phosphorylation of MARCKS was measured and quantified by phosphorimage analysis.
Subcellular fractionation of cells and Western blot for PKC isoforms
SH-SY5Y/TrkA cells treated with NGF or TPA were incubated in digitonin
buffer and homogenized using a Dounce homogenizer with a glass pestle.
36
One part of the homogenate was incubated with Triton-X100, followed by
centrifugation. The supernatant was collected and denoted total lysate (T).
The rest of the homogenate was separated into a cytosol (S) and particulate fraction (P) by ultracentrifugation. Proteins in the cytosolic fraction were
precipitated with trichloroacetic acid (TCA) and the pellet was dissolved in
sodium hydroxide (NaOH)/hydrochloric acid (HCl). The particulate fraction
was incubated with digitonin/Triton-X100 buffer and centrifuged to remove
the insoluble cytoskeletal fraction.
Equal amounts of proteins from each fraction were separated by 8% SDSPAGE and the PKC isoforms were detected using Western blot analysis.
Primary antibodies used were anti-PKC-α (Gibco BRL, Life Technologies,
US), anti-PKC-δ and anti-PKC-ε (SantaCruz Biiotechnology Inc., US), and
the immunoreactivity was detected using ECL (Amersham, UK).
[32Pi]-orthophosphate labelling of cells, in situ trypsin digestion and 2D
phosphopeptide mapping of Raf-1
Serum-starved SH-SY5Y/TrkA cells were pre-incubated with 3 mCi [32Pi]orthophosphate (PBS.43) in phosphate-free Eagle’s medium, before treating
the cells with 100 ng/mL NGF or 16 nM TPA. After treatment, total protein
lysates were prepared. RNase A was added to the lysates and Raf-1 was immunoprecipitated using Raf-1 antibody (C-12, SantaCruz Biotechnology
Inc., US).
The immunoprecipitates were separated on SDS-PAGE, transferred to
Hybond C-extra membranes, trypsin digested and prepared for twodimensional phosphopeptide mapping [180]. The samples were separated
using a cellulose thin-layer chromatography plate, first in one dimension in
electrophoresis buffer and then in a second dimension by ascending chromatography in isobutyric acid buffer. The radiolabelled peptides were visualized using phosphorimage analysis.
Statistics and bioinformatics
All statistical analyses were performed by the Statistical Package for the
Social Sciences 20.0 (SPSS Inc., US) for Windows software package.
All significance tests were two-tailed and p-value < 0.05 was considered
statistically significant. For normally distributed data, ANOVA was used
(study I) and for non-normally distributed data, Mann-Whitney U test was
used (study II and III). Categorical data were compared using Pearson ChiSquare test (study II).
Differentially expressed genes between the groups (study I) was defined
by using an empirical Bayes moderated t-test with robust regression (Smyth,
2004). To visualize differentially expressed genes, the Genesis software,
version 1.7.1 (http://genome.tugraz.at/) was used, also to produce hierar37
chical clustering. The expression data was analyzed using Ingenuity pathway
analysis (IPA), which calculates the top genes that are the most deregulated
and identifies pathways and networks in which the focus genes are involved.
Results from the mass spectrometry analysis of embryos (study III) were
analyzed using the Mascot™ search algorithm (Matrix Science, UK) embedded in Proteome Discoverer software (ThermoFisher Scientific, Germany)
and searched against human proteins in the UniProtKB/Swiss-Prot database.
At least two matching peptides of 95% confidence per protein counted as
protein identification.
A Venn diagram (study III) was made using software freely available on
the internet (Oliveros, J.C. (2007). http://bioinfogp.cnb.csic.es/tools/venny/),
to detect and illustrate which proteins are expressed in SSRI-treated women,
healthy controls or in both groups. IPA analysis was performed to discover
which pathways and networks are associated with the proteins of interest.
Ethical approvals (study I to III)
The BASIC study, including these sub-studies, and the embryo study were
approved by the Regional Ethics Review Board in Uppsala, Sweden (approval numbers 2009/171 and 2014/298, respectively).
38
Summary of Results
Study I
The gene expression in human placenta from SSRI-treated women, untreated
depressed women and healthy controls were studied.
A microarray analysis was performed on biopsies collected from the fetal
side of the placenta from depressed women, SSRI-treated women and
healthy controls. Demographic data of the women participating in the study
is presented in Table 2. The analysis revealed 109 genes that were differentially expressed between SSRI-treated women and controls, 82 genes were
down-regulated and 27 were up-regulated. In depressed women, 108 genes
had an altered expression compared with healthy controls, 100 genes were
down-regulated and eight genes were up-regulated. IPA analysis revealed
that 17 genes in the depressed group and 20 genes in the SSRI-treated group
had the most significant alterations compared with healthy controls (Table 5
and 6). Of all genes that had an altered expression in depressed women and
SSRI-treated women compared with controls, 20 genes were overlapping
(Table 7).
The IPA analysis also detected significant canonical pathways of interest
when comparing depressed women with healthy controls, as well as SSRItreated women with healthy controls (Table 8 and 9).
For further validation of the microarray results, we selected seven genes
that were more prominently altered in their expression between groups, or
found in the pathway or canonical pathway analysis in placenta from depressed and SSRI-treated women. We validated the expression by performing a qRT-PCR analysis on a larger number of women in each study group
(Fig. 4). In placenta from SSRI-treated women, ROCK1 and ROCK2 were
significantly down-regulated when compared with healthy women (Fig. 4B).
Also, GCC2, KTN1 and DNM1L were significantly down-regulated in women treated with SSRI compared with controls. For C12orf39, there was only
a tendency for down-regulation in SSRI-treated women compared with
healthy women. The gene expression of NEXN did not differ between SSRItreated and healthy women.
In depressed women, there was a tendency for down-regulation of
ROCK2 compared with controls (Fig. 4A). There was also a significant
down-regulation of placental gene expression of C12orf39 in depressed
39
vault RNA 1-2
placenta growth factor
ribonuclease/angiogenin inhibitor 1
inositol 1,3,4-triphosphate 5/6 kinase
microRNA 503
RAD23 homolog A (S. cerevisiae)
apolipoprotein C-I
ubiquitin specific peptidase 15
protein phosphatase 1. regulatory (inhibitor) subunit 12A
formin binding protein 1-like
structural maintenance of chromosomes 6
structural maintenance of chromosomes 4
Rho-associated. coiled-coil containing protein kinase 2
A-kinase (PRKA) anchor protein (yotiao) 9
COP9 constitutive photomorphogenic homolog subunit 2 (Arabidopsis)
GRIP and coiled-coil domain containing 2
Rho-associated. coiled-coil containing protein kinase 1
Gene title
40
* “-“ = reduction in expression levels
VTRNA1-2
PGF
RNH1
ITPK1
Mir-503
RAD23A
APOC1
USP15
PPP1R12A
FNBP1L
SMC6
SMC4
ROCK2
AKAP9
COPS2
GCC2
ROCK1
Gene symbol
Table 5. Significantly up- and down-regulated top molecules in the control vs depressed group
2.28
0.70
0.57
0.55
0.52
0.52
0.50
-0.87
-0.88
-0.89
-0.92
-0.92
-0.92
-0.94
-0.98
-1.02
-1.07
log2 fold change*
0.026
0.013
0.036
0.037
0.047
0.013
0.037
0.026
0.021
0.046
0.026
0.024
0.021
0.028
0.008
0.019
0.028
P value
chromosome 12 open reading frame 39
RNA, U4 small nuclear 1
keratin 81
RNA, U4 small nuclear 2
serine incorporator 2
apelin
angiopoietin-like 4
tubulin, alpha 1c
S100 calcium binding protein A3
trans-2,3-enoyl-CoA reductase
ankyrin repeat domain 12
zinc finger protein 721
Rho-associated, coiled-coil containing protein kinase 1
A-kinase (PRKA) anchor protein (yotiao) 9
synaptonemal complex protein 2
early endosome antigen 1
ankyrin repeat domain 36B
COP9 constitutive photomorphogenic homolog subunit 2 (Arabidopsis)
GRIP and coiled-coil domain containing 2
nexilin (F actin binding protein)
Gene title
* ”-“ = reduction in expression levels
C12orf39
RNU4-1
KRT81
RNU4-2
SERINC2
APLN
ANGPTL4
TUBA1C
S100A3
TECR
ANKRD12
ZNF721
ROCK1
AKAP9
SYCP2
EEA1
ANKRD36B
COPS2
GCC2
NEXN
Gene symbol
Table 6. Significantly up- and down-regulated top molecules in the control vs SSRI group
1.28
0.91
0.78
0.71
0.64
0.63
0.62
0.56
0.54
0.54
-0.78
-0.78
-0.80
-0.84
-0.87
-0.88
-0.88
-0.88
-0.92
-1.00
log2 fold change*
0.009
0.026
0.007
0.043
0.038
0.014
0.047
0.027
0.042
0.048
0.042
0.033
0.041
0.017
0.042
0.015
0.041
0.007
0.032
0.044
P value
41
JMJD1C
DNM1L
ERGIC2
KTN1
ARID4A
SDCCAG1
COPS2
ROCK1
ZNF146
ZNF100
GCC2
ROCK2
PRPF40A
NKTR
--POLK
RAD50
LYSMD3
AKAP9
PHF20L1
jumonji domain containing 1C
dynamin 1-like
ERGIC and golgi 2
kinectin 1 (kinesin receptor)
AT rich interactive domain 4A (RBP1-like)
serologically defined colon cancer antigen 1
COP9 constitutive photomorphogenic homolog subunit 2 (Arabidopsis)
Rho-associated, coiled-coil containing protein kinase 1
zinc finger protein 146
zinc finger protein 100
GRIP and coiled-coil domain containing 2
Rho-associated, coiled-coil containing protein kinase 2
PRP40 pre-mRNA processing factor 40 homolog A (S. cerevisiae)
natural killer-tumor recognition sequence
--polymerase (DNA directed) kappa
RAD50 homolog (S. cerevisiae)
LysM, putative peptidoglycan-binding, domain containing 3
A-kinase (PRKA) anchor protein (yotiao) 9
PHD finger protein 20-like 1
42
Gene Symbol
Gene Name
7933877
7954752
7962013
7974483
7974621
7978866
7988605
8022441
8028186
8035808
8044236
8050302
8055913
8079079
8098287
8106303
8107942
8113064
8134122
8148358
Probe Set ID
Table 7. Genes with altered expression in both depressed and SSRI-treated women compared with controls
Log2 Fold change
SSRI
Depressed
-0.612753894
-0.86686842
-0.713859601
-0.716232722
-0.65241833
-0.820896474
-0.712691264
-0.866627304
-0.602522967
-0.661691075
-0.717656644
-0.776418378
-0.883531246
-0.975928968
-0.804644966
-1.074633032
-0.679184287
-0.584609444
-0.534310441
-0.741413733
-0.919095326
-1.024026926
-0.73784498
-0.924567683
-0.641694474
-0.652162751
-0.602491778
-0.63772807
-1.149966378
-0.658215787
-0.576549694
-0.814486061
-0.732319601
-0.827079801
-0.675591155
-0.780972234
-0.83694096
-0.935693684
-0.514646483
-0.551986952
Table 8. Canonical pathway analysis of the depressed group
Canonical pathway
Genes
P
value
Actin Nucleation by ARP-WASP Complex
PPP1R12A, ROCK1, ROCK2
0.003
RhoA Signaling
KTN1, PPP1R12A, ROCK1,
ROCK 2
0.029
VEGF Signaling
PGF, ROCK1, ROCK2
0.011
Protein Kinase A Signaling
AKAP9, ANAPC4, PDE3B,
PPP1R12A, ROCK1, ROCK2
0.011
1D-myo-inositol Hexakisphosphate
Biosynthesis V (from Ins(1,3,4)P3)
ITPK1
0.015
Table 9. Canonical pathway analysis of the SSRI-treated group
Canonical pathway
Genes
P
value
Ephrin A Signaling
EFNA5, ROCK1, ROCK2
0.001
RhoA Signaling
CDC42EP1, KTN1,ROCK1,
ROCK2
0.002
PEDF Signaling
FAS, ROCK1, ROCK2
0.004
Breast Cancer Regulation by Stathmin 1
ARHGEF5, ROCK1, ROCK2,
TUBA1C
0.012
Signaling by Rho Family GTPases
ARHGEF5,CDC42EP1, ROCK1, 0.021
ROCK2
43
44
Figure 4. Gene expression detected by qRT-PCR in placenta comparing depressed (A) or SSRI-treated women (B) with healthy women.
A) *P = 0.05, #P = 0.08, B) *P ≤ 0.05, **P < 0.01, #P = 0.07.
women compared with healthy controls. No other genes were differentially
expressed in placentas between depressed and healthy women.
Our results indicate that SSRI treatment and depression per se alters the
expression of many genes in placenta, although not in the same way.
Study II
To further investigate whether the altered gene expression of ROCK1 and
ROCK2 found in study I could also be detected in placenta on a protein level, SSRI-treated, untreated depressed and healthy pregnant women were
compared. The demographic data is shown in Table 3. ROCK1 and ROCK2
are involved in RhoA signaling, which is also one of the signaling pathways
downstream of the NGF receptor TrkA. We therefore studied the NGF signaling pathways and also investigated which pathway from the receptor
could lead to altered ROCK levels (Fig. 3).
By performing Western blot analysis on total protein lysates from placenta, we did not find any significant changes in protein levels of the proteins
investigated.
Immunohistochemical (IHC) staining of placental sections showed that
the staining pattern was almost the same for all the proteins investigated,
with the strongest staining in the trophoblasts, weaker in the endothelial
cells, and the weakest staining in the stromal cells (Fig. 5). Only ROCK1
had a different staining pattern, with the strongest staining in the endothelial
cells, weaker in the trophoblasts, and the weakest in the stromal cells. As an
indication of active signaling, the phosphorylated forms of Raf-1 and
ROCK2 were accumulated in the nucleus of the cells, whereas the staining
for total Raf-1 and ROCK2 was found both in the nucleus and the cytoplasm
(Fig. 5).
When performing IHC staining on placental sections, NGF levels were
increased in trophoblasts and endothelial cells in SSRI-treated women compared with depressed women and healthy controls (Fig. 6A and B). The NGF
staining intensity in endothelial cells was decreased in depressed women
compared with controls (Fig. 6B).
Phosphorylated Raf-1 was increased in stromal cells in both SSRI-treated
and depressed women compared with controls (Fig. 6C). No other differences in levels of phosphorylated Raf-1 were found between groups in
trophoblasts or endothelial cells.
There was a tendency towards increased ROCK2 levels in placental
trophoblasts and endothelial cells in SSRI-treated women compared with
healthy controls (Fig. 6A and B). In stromal cells, ROCK2 levels were significantly higher in SSRI-treated women than in controls (Fig. 6C). However, ROCK2 levels in endothelial cells were significantly lower in women
treated with SSRI compared with depressed women (Fig. 6B). Also,
45
Figure 5. Immunohistochemical stainings for NGF, TrkA, RhoA, ROCK1, Raf-1,
pRaf-1, ROCK2 and pROCK2 in placenta. T = trophoblasts, E = endothelial cells
and S = stromal cells.
depressed women had increased levels of ROCK2 in all cell types compared
with controls (Fig. 6A-C).
The staining intensity of phosphorylated ROCK2 was significantly increased in all three cell types when comparing SSRI-treated women with
depressed women and healthy controls (Fig. 6A-C). Also, the placental staining intensity of phosphorylated ROCK2 was significantly higher in endothelial and stromal cells of depressed women than in healthy controls (Fig. 6B
and C).
No significant differences in staining intensities of TrkA, Raf-1, RhoA
and ROCK1 were detected between SSRI-treated, depressed and healthy
women in trophoblasts, endothelial or stromal cells.
46
pROCK2
3
3
3
3
2
2
2
2
1
1
1
1
0
0
0
0
-1
-1
-1
-1
*
3
3
2
2
2
2
1
1
1
1
0
0
0
0
-1
-1
-1
-1
C
I
SS
R
ROCK2
3
3
3
3
2
2
2
2
1
1
1
1
0
0
0
0
-1
-1
-1
-1
***
5
*
SS
R
I
4
ep
D
C
I
*
r
**
4
SS
R
r
C
ep
D
SS
R
ep
D
I
4
r
4
C
5
***
5
**
pROCK2
5
***
***
*
I
r
D
ep
C
pRaf-1
*
4
SS
R
NGF
SS
R
D
ep
C
I
SS
R
r
ep
C
D
C)
*
I
3
**
5
SS
R
3
I
4
***
r
4
*
r
pROCK2
5
4
ep
C
I
ROCK2
#2
5
5
***
D
D
pRaf-1
SS
R
r
4
ep
C
SS
R
D
NGF
***
ep
C
I
r
SS
R
ep
C
D
B)
***
5
I
4
*
I
4
4
r
5
SS
R
ROCK2
#1
5
D
ep
r
pRaf-1
***
D
ep
r
NGF
5
C
A)
Figure 6. Protein levels in different cell types of placenta detected by IHC. A)
Trophoblasts, B) Endothelial cells and C) Stromal cells. *p ≤ 0.05, **p ≤ 0.01, ***p
≤ 0.001, #1 = 0.065, #2 = 0.056, Mann-Whitney U test.
Study III
Embryos treated with 0.5 µM FLX needed a significantly shorter time to
reach formation of a morula/compaction compared with controls (Table 10).
Also, the time to reach other developmental stages tended to be shorter in
FLX-treated embryos compared with controls. FLX treatment did not significantly alter the number of embryos developing into blastocysts or the quality of the embryos.
47
Table 10. Timing of developmental stages in human embryos treated with or without 0.5 µM FLX
Control
na = 10
0.5 µM FLX
na = 10
Thawing - first cell division,
median (min – max)
8.60 (2.20 – 38.90)
13.55 (1.00 – 19.30)
Thawing - fourth cell division,
median (min – max)
29.20 (2.90 – 58.20)
20.15 (5.20 – 37.40)
Thawing - compaction, median
(min – max)
56.75 (29.10 – 72.00)
44.05 (34.90 – 51.60)*
Thawing - cavitation, median
(min – max)
57.55 (40.90 – 84.90)
47.50 (41.90 – 71.40)
Thawing - start of expansion,
median (min – max)
71.90 (56.90 – 90.20)
67.90 (53.40 – 93.20)
Time difference (hours)
*p < 0.05 compared with control, Mann-Whitney U test
a
n; number of embryos from start of culture
By performing a bottom-up shotgun proteomics analysis on whole embryo, we investigated whether FLX induces unique protein expression patterns in the embryo. We discovered that 36 proteins were uniquely expressed
in the FLX-treated embryos and 48 proteins were detected in both groups
(Fig. 7). In order to determine the biological relevance of the 36 proteins
expressed in FLX-treated embryos, an IPA analysis was performed. Three
networks of relevance were detected in embryos treated with FLX (Table
11).
Figure 7. Venn diagram illustrating proteins detected in FLX-treated embryos and
controls by mass spectrometry.
48
Table 11. Networks identified by IPA, focusing on the proteins detected by mass
spectrometry in FLX-treated embryos compared with controls
IPA
IPA networks top 3
Proteins
score
Cell Death and Survival, Cellular
Growth and Proliferation, Hematological Disease
ENO1, HBB, HNRNPA2B1,
HNRNPC, NCL, NPM1, PEBP1,
PFN1, PPIA,RPL8, RPS16, YBX1
23
Cancer, Organismal Injury and
Abnormalities, Reproductive System Disease
ALDOA, CLTC, P4HB, PARK7,
PGK1, PRDX1, RPLP2, STMN1,
TPI1, TUBB4A
18
Inflammatory Response, Cell Death
and Survival, Digestive System
Development and Function
ATP1A1, C1QBP, HP, HPX,
KRT8, NSF, PPIA, SOD1
14
By using Proseek Multiplex Inflammation I96x96 Immunoassay from
Olink, protein secretion from embryos with or without FLX treatment was
investigated. Culture medium in which the embryos had been cultured was
analyzed. Several proteins, after normalization, were detected above the
limit of detection. Levels of urokinase-type plasminogen activator (uPA)
tended to be higher in FLX-treated embryos compared with controls (Table
12). For other proteins, there were either no significant differences between
the groups or the number of embryos where the protein could be detected
was too limited for statistical analyses.
Table 12. Top 10 proteins detected by Multiplex Immunoassay analysis and above
levels of detection in medium from 0.5 µM FLX-treated and control embryos
Control
0.5 µM FLX
n = 10
n = 10
Protein
UniProtKB
n median (min – max) n median (min – max)
uPA
P00749
9
1.30 (0.40 – 3.20)
10
2.15 (0.90 – 3.40)
IL-6
P05231
8
1.35 (0.80 – 3.20)
10
2.05 (0.50 – 4.90)
STAMPB
O95630
7
0.50 (0.40 – 1.10)
6
0.55 (0.40 – 1.00)
ADA
P00813
7
1.70 (1.10 – 2.30)
5
1.50 (1.10 – 1.80)
CST5
P28325
6
0.10 (0.10 – 0.20)
1
0.10 (N/A)
Beta-NGF
P01138
5
0.60 (0.40 – 1.50)
2
0.25 (0.20 – 0.30)
FGF-23
Q9GZV9
5
0.40 (0.30 – 0.40)
1
0.30 (N/A)
IL-8
P10145
4
1.15 (0.10 – 5.20)
5
1.30 (0.20 – 3.20)
IL-10
P22301
4
0.40 (0.30 – 0.40)
1
0.10 (N/A)
VEGF-A
P15692
3
0.60 (0.10 – 1.80)
8
0.85 (0.20 – 1.70)
49
Immunohistochemical staining of the embryos was performed to visualize
the true presence of some of the proteins of interest. uPA and NGF were
chosen because of their high ranking in the Multiplex Immunoassay analysis.
Also, NGF protein levels in placenta had been shown to be altered in SSRItreated women compared to healthy controls (study II). uPA and NGF staining were found in the cytosol and cellular membrane of the trophectoderm
(Fig. 8). NGF was furthermore found in the inner cell mass, whereas uPA
had only a weak staining in the inner cell mass of the embryos (Fig. 8).
Figure 8. IHC staining of human embryos. A-D are embryos cultured in control
medium, E-F are embryos treated with 0.5µM FLX. DAPI staining (blue) shows cell
nuclei. Staining for NGF is shown in green and uPA in red. D and H are overlay
pictures.
Study IV
Signal transduction down-stream of the NGF receptor TrkA in human neuroblastoma cell line SH-SY5Y transfected with TrkA was studied.
In SH-SY5Y/TrkA cells treated with TPA or NGF for 5 minutes, 30
minutes, 1 hour and 2 hours, both TPA and NGF caused a rapid activation of
Raf-1. Raf-1 had an almost threefold increase in activation by NGF (Fig. 9).
TPA also induced an activation of Raf-1, although this was weaker.
Raf-1 became phosphorylated to a higher extent after TPA treatment than
after NGF treatment, shown by a mobility shift assay (Fig. 10A). The NGFinduced phosphorylation and activation of Raf-1 was not inhibited by the
PKC-inhibitor GF109203X (Fig. 10B and 11).
50
Figure 9. Activation of Raf-1 in SH-SY5Y/TrkA cells after treatment with 100
ng/mL NGF (black bars) or 16 nM TPA (grey bars). The cells were treated for 5
minutes, 30 minutes, one hour and two hours. The values are mean ± SEM.
Figure 10. Phosphorylation-dependent decelerated mobility of Raf-1in SYSY5Y/TrkA cells. Phosphorylated proteins have a higher molecular weight, resulting in a band detected higher up in the Western blot. A) Raf-1 phosphorylation after
treatment with 100 ng/mL NGF or 16 nM TPA. B) The effect of PKC inhibitor
GF109203X on NGF-induced phosphorylation of Raf-1.
51
Figure 11. Effect of the PKC inhibitor GF109203X on Raf-1 activity induced by
treatment with 100 ng/mL NGF. Black bars represent NGF alone and grey bars NGF
treatment in the presence of GF109203X. Data are based on two independent experiments.
NGF treatment also induced an activation of Ras, whereas TPA had no effect on Ras activity (Fig. 12). Also, NGF did not notably activate different
PKC isoforms, whereas TPA induced a strong activation of PKC, detected as
phosphorylation of MARCKS (Fig. 13).
In conclusion, the results indicate that Raf-1 can be activated and phosphorylated by both NGF and TPA in SH-SY5Y/TrkA cells, but via different
pathways. NGF-induced activation of Raf-1 is mediated via Ras, whereas the
TPA-induced activation is mediated via PKC. Our results also show that,
even though wild-type SH-SY5Y cells with a very low amount of endogenous TrkA do not respond to NGF treatment, the signaling pathways downstream of the TrkA receptor are still intact.
52
Figure 12. Ras activity in SH-SY5Y/TrkA cells treated with 16 nM TPA or 100
ng/mL NGF. Immunoprecipitation (Ip) of active Ras was performed using the Ras
binding domain (RBD) as bait and Ras was detected by immunoblotting (Ib) for
Ras.
Figure 13. PKC activation in SH-SY5Y/TrkA cells after treatment with 16 nM TPA
or 100 ng/mL NGF. PKC activation was measured by phosphorylation of
MARCKS, where the lane to the very right is a positive control. The average level of
MARCKS phosphorylation (measured as number of pixels) is shown by numerical
values beneath each condition.
53
Discussion
Methodological considerations
Choice of control group
In studies on SSRI, the effect of the underlying condition that is being treated must be isolated. In epidemiological studies this is typically done by including control groups that either have discontinued SSRI use prior to pregnancy, or who have untreated mental health conditions. Either choice is associated with its own pitfalls. In this thesis, we have chosen women with
untreated depression (in the sense that women were not medically treated several women we’re receiving counseling or therapy) as controls. It could
be argued that women who had discontinued SSRI would have made a
stronger control group, but information on pre-pregnancy drug treatment was
not available in the BASIC study questionnaires, and information on already
discontinued treatment is rarely disclosed to the mid-wife at first antenatal
booking. Furthermore, timing of discontinuation would be critical for the
experiments of this thesis. While women may state at a first antenatal booking that they have discontinued treatment, they may not have done so until
they discovered the pregnancy, ultimately exposing the early placentation
process to SSRI. In addition, 75% of women who are on SSRI treatment
when they start planning a pregnancy or realize they are pregnant will discontinue [59]. At present, there is no information on why some women decide to quit their medication while others continue. It can be assumed that
women who chose to continue SSRI during pregnancy are the ones in greatest need of treatment. Possibly, they had a severe episode when treatment
was initiated, possibly they had been on treatment for a longer time, possibly
prior attempts to terminate treatment had failed, and possibly a co-morbid
anxiety disorder made it impossible to stop treatment during pregnancy.
Whatever reason they may have had for continuing treatment, we are left
with the possibility that their depression is more severe than the depression
of untreated depressed women.
However, SSRIs are not only used in depression but also in anxiety disorders. We chose untreated depression as control, but it may be argued that
untreated anxiety could have been a relevant alternative. However, our
choice was merely based on the fact that depression is far more studied than
54
anxiety in pregnancy. Furthermore, anxiety disorders are relatively heterogeneous, with some diagnoses stressing situational anxiety (specific phobias,
panic disorder) whereas other disorders may be characterized by more generalized anxiety symptoms.
In choosing untreated depression, it should also be acknowledged that depression is not a simple and straightforward phenotype. The diagnosis of
depression is based on the presence of a group of symptoms, but the underlying causes of the depressive episode may vary; life stress, trauma, infection,
severe medical conditions, drug or alcohol abuse, just to mention a few, all
of which may influence the placental function differently. In addition, the
depressive episode may also differ in its duration and severity, which in turn,
may affect the associated disturbances in the HPA axis [24]. All in all, the
complexity of depression is illustrated in study I, where among the genes
chosen for biological validation of the microarray, only 2 were validated
with qRT-PCR for depressed women, whereas 6 (out of 7) genes were validated among women who had been treated with antidepressants. As more
placental gene alterations were validated in a larger subset of SSRI-treated
women compared to those with antenatal depression, it was concluded that
for these subset of genes, the effects of SSRI intake during pregnancy appear
more robust. Presumably, a more homogeneous group of depressed women
could have been assembled if we had added some type of biological criteria
to the diagnosis, for instance depression with elevated baseline cortisol levels.
It may also be argued that the severity of depression actually differed between SSRI cases and women with untreated depression. Because the placental studies were performed within the BASIC study framework we had
access to self-rated depression scores for all participating women in gestational week 17 and 32. Indeed, from these scores it was apparent that women
on SSRI treatment displayed lower depression scores than the untreated controls. Had we chosen a control group with similar depression scores to the
SSRI users, they would not have been depressed. Alternatively, finding SSRI
users with high depression scores would impact greatly on the number of
cases available.
In study II and in the validation experiment of study I, untreated depression was merely defined by the EPDS scores of participating women. Preferably, a group with diagnosed major depressive disorder should have been
included in all experiments, but most women with depressive disorders go
undetected throughout pregnancy [2]. In addition, it should also be pointed
out that, while EPDS may fail to detect depression in almost half of cases, its
specificity for depressive disorders is very high at the cut-offs we were using
[14].
Depression is also associated with a number of factors that may influence
placental function. Smoking, obesity, young age, drug or alcohol use, and
domestic violence all contribute to the risk of adverse perinatal outcomes,
and smoking and obesity are specifically associated with altered placental
55
function [181-183]. We have tried to control for these associated factors by
matching controls by age (study I and II) and BMI (study I), or adjusting for
these factors in the statistical analyses. Severe medical conditions, daily
medication and obstetric complications were exclusion criteria for both papers. Smoking was not an exclusion criteria but was rare in our study population. Similarly, none of the subjects acknowledged alcohol use during
pregnancy.
Sample size
Ideally, sample size is determined by a priori power analyses. Adequate
sample sizes are important not only to avoid type two errors (failure to detect
true findings) but also to protect against false significant findings. However,
to be completely honest, the sample sizes used in the present thesis were to a
large extent based on available tissue. The BASIC study started to collect
placental tissue in 2010, and by the time the initial microarray analysis was
planned, approximately 200 placentas were available. With time, the placenta biobank has grown to include more than 50 women who have used SSRIs
during the entire pregnancy, but all cases cannot be used due to the presence
of obstetric complications. Presumably due to positive attrition, the proportion of SSRI users is greater than in the general population (approximately
5.5% in comparison with the expected 2.5%) [57]. In addition, the sample
size for study II also depended on availability of formalin-embedded placental tissue. However, while the sample sizes for the microarray analysis were
extremely small, i.e. 5 depressed and 5 SSRI-treated women, each compared
with 5 healthy controls (in total 10 controls), the microarray analysis revealed many genes to be differentially expressed between the groups. Also,
these results were validated by qRT-PCR in a larger population, where significantly altered gene expressions could be confirmed, at least in the SSRI
cases.
Obviously, the above limitation has precluded any differentiation between
different SSRIs, and for the validation of the microarray, a few cases using
other types of antidepressants were also included. This may have influenced
the outcome of the gene expression in the SSRI treatment group.
Similarly, the number of embryos used in study III is very low, but for
obvious reasons, the number of human embryos that can be used for research
is not unlimited. The relatively low number of embryos in this study could
have an impact on the statistics. However, several significant alterations in
embryos treated with FLX were discovered. Also, several changes were noted in e.g. protein expression in embryo culture medium that tended to differ
between treated and untreated embryos.
56
Human embryos
The use of human embryos for study III distinguishes us from all previous
attempts to elucidate the effects of SSRIs on embryo development. However,
with no previous studies to rely on, a number of methodological issues were
encountered. The aim was to investigate the effects of pharmacologically/therapeutically relevant levels of FLX on human early embryo development. The dose used in the study corresponded to physiological umbilical
cord and amniotic fluid levels of FLX-exposed fetuses in women with commonly used doses of FLX [64, 65, 184]. A small dose-response pilot study
was also conducted before we started the trial; with higher concentrations of
FLX (1 µM) added to the culture medium, the number of dead embryos increased. These dual effects of SSRI have been noted earlier with mice embryo, where FLX induced an enhanced embryo development into blastocysts
up to a certain dose, followed by an inhibition of blastocyst formation at
higher doses [97].
The treated embryos in culture correspond to embryos in early preimplantatory phase. In a spontaneous pregnancy this means that the placenta
has not yet been developed and there is no amniotic fluid or umbilical cord
through which FLX could reach and affect the embryo. However, during the
pre-implantation period, the embryo might be exposed to FLX via the surrounding environment (follicular fluid in ovary, fallopian tubes, endometrium and myometrium in uterus).
The analysis method used for the proteomics has been used in several
previous studies within reproductive medicine [185-187], but the complexity
of the method should be taken into consideration when interpreting and analyzing the results.
Another possible limitation of the study is that the development of an embryo in culture is different from maturation and development in an intrauterine environment. However, this also holds true for embryos that are transferred back to the utero during in vitro fertilization (IVF), meaning that these
embryos are in an environment that allows them to develop into blastocysts
and further on into a fetus.
Experimental considerations
To prepare and isolate RNA is already known to be quite difficult and to
prepare RNA from placenta (study I) needed an optimized protocol. RNA
easily degrades and degraded RNA is useless for down-stream applications.
However, the speed of degradation can be reduced by immediate freezing of
the tissue after collection. In our study, placenta was collected at birth and
frozen almost at bedside. However, our experience was that RNA from placenta seems to degrade quickly and that it is not always easy to handle sample collection in the delivery room.
57
A full-length RNA is of great importance for both microarray and qRTPCR analysis. For microarray analysis, a poor quality RNA reduces the
chances of identifying genes of importance. For qRT-PCR, the cDNA used
needs to be of full-length, to avoid any false negative findings or misinterpreted results. Since cDNA synthesis uses RNA as a template, a good quality
RNA is crucial.
Other limitations for qRT-PCR analysis are the primers and probes, which
need to be selected carefully so that they have as good hybridization capacity
as possible for the gene of interest. The primer and probes used in study I are
validated for each gene detected, but of course these validations are made by
the manufacturer and could be biased.
Performance of immunohistochemistry (IHC) (study II and III) is highly
dependent on the primary antibody and its specificity for the targeted protein. A poor specificity can give both false positive and negative results.
Also, a highly purified secondary antibody is important to avoid false binding to other compounds and materials, creating a high background or false
positive findings. The dependency on the specificity of the primary and secondary antibodies also holds true for Western blot (WB) analysis (study II
and IV).
For WB, the enhanced chemiluminescence (ECL) method using films to
identify proteins (study IV) has, since the paper was published, been further
developed. If the study had been performed today, the detection of chemiluminescence would probably have been made by sensitive screens and/or
scanners. Alternatively, fluorescently labeled antibodies detected by imagers
could have been used, making it possible to analyze Western blots more
quantitatively (as was done in study II).
When scoring IHC stained placental sections (study II), this was done by
visual inspection in a light microscope. This could be a limitation to the
study, since such scoring can be subjective. To avoid biased results, a double-blinded scoring was performed in the same way by a second person who
had no access to the initial results.
The effects of SSRI and antenatal depression on
placenta
Many studies have been conducted in order to investigate the effects of antenatal depression and SSRI treatment during pregnancy, but the underlying
biological mechanisms for these are largely unknown. The placenta is an
important organ in the development of the fetus and this thesis includes two
studies which: 1) investigated the gene expression in the fetal side of the
placenta in order to find gene expression patterns in mothers with antenatal
depression and in mothers using SSRI treatment during pregnancy (study I);
58
and 2) investigated, based on the results in study I, the NGF signaling pathway in placenta from mothers with antenatal depression and using SSRI
treatment during pregnancy (study II).
ROCK1 and ROCK2
ROCK1 and ROCK2 are part of the Rho-associated coiled-coil kinase family
[188, 189] and are downstream effectors of RhoA-GTP. Rho-ROCK signaling pathways are involved in the regulation of actin cytoskeleton, cell migration and proliferation [190]. In mice, ROCK1 is highly expressed in the lung,
liver, spleen, kidney and testis, whereas ROCK2 is most abundant in the
brain and heart [189, 191, 192].
No altered protein levels of ROCK1 was seen between the groups by
IHC, but elevated levels of phosphorylated ROCK2 were detected in all placental cell types in SSRI-treated women compared with depressed and
healthy controls. These findings might seem contradictory to the results in
study I, where ROCK1 and ROCK2 gene expressions were down-regulated
in SSRI-treated women. Also, phosphorylated ROCK2 levels were slightly
elevated in all placental cell types in depressed women compared with controls (although lower than in SSRI-treated women), whereas a tendency towards lower ROCK2 gene expression was noted in depressed women compared with controls. This discrepancy between protein levels and gene expression might be explained by the fact that in gene expression analysis,
pooled placental cells were used. However, gene expression and protein
expression do not necessarily mirror each other and the results found could
also be explained by different post-translational modification in SSRI users
and depressed women [193, 194].
Also, IHC revealed alterations in protein levels, but this was not confirmed by WB analysis. This is most likely explained by the fact that in WB
analysis all cell types in the placenta are pooled, meaning that a decrease in
levels in one cell type might mask an increase in another cell type and vice
versa, or a small effect in only one cell type might not be seen when all cell
types are mixed together.
It is interesting though, that SSRI treatment alters both gene expression
and protein levels of ROCK2 in placenta. The role of the Rho/ROCK family
in cardiovascular diseases has been extensively studied [195]. Cardiac malformations [130], including pulmonary hypertension [57, 114, 127, 132],
have also been reported in the SSRI-exposed offspring. ROCK2 has been
found in vascular smooth muscle cells and has been shown to play a role in
hypoxia-induced pulmonary hypertension in mice [196], a rare but important
complication from SSRI use during pregnancy [57].
In addition, normal ROCK1 and ROCK2 activity is required for normal
inner cell mass morphogenesis, which is of importance for successful fetal
development [197]. The findings in study I indicate that antidepressant use
59
during pregnancy may influence pregnancy complications and fetal development, and that ROCK1 and ROCK 2 may be involved with these processes.
Due to lowered ROCK2 gene expression found in the fetal placenta of
SSRI-treated women compared with depressed women, it is tempting to
speculate that a normal expression of ROCK2 in placenta is important for
normal functioning of the cardiovascular system in the fetus. Ongoing studies in the lab on SSRI-treated heterozygous serotonin transporter knock-out
rats exposed to maternal deprivation (leading to a depressive-like phenotype)
may shed light on the SSRI-induced effects on offspring cardiovascular system.
Trophoblasts, placenta and pregnancy complications
Trophoblasts are important in exchanges between the fetus and the mother
and possess endocrine activity releasing hormones that are important in the
maintenance of pregnancy [198]. In addition, trophoblasts are involved with
the secretion of placental growth hormone and are important in the development of the placenta [199].
NGF is found to be expressed in human placenta [200, 201], where it is
involved in important functions such as placentation [202] and pregnancy
maintenance [142]. In study II, elevated protein levels of NGF in placental
trophoblasts and endothelial cells in SSRI-treated women were found. Previous animal and human research has shown that NGF signaling is involved in
miscarriage and preterm birth [148, 149, 203], so our study could indicate
that NGF might have a role in SSRI-induced pregnancy outcomes. Inadequate trophoblast invasion and endothelial dysfunction are important features in the development of preeclampsia.
Also, as shown by IHC, higher ROCK1 levels were detected in endothelial cells, whereas ROCK2 was predominantly found in trophoblasts. A role
for ROCK1 has been suggested in hypertension [204-207] and increased
ROCK2 expression has been described in pre-eclamptic human placentas
[208]. This indicates that ROCK1 and ROCK2, even though both in the
NGF signaling pathway, might play different roles in the development and
function of the placenta.
Since SSRI-treated women had increased protein levels of ROCK2 in
trophoblasts, it may be speculated that NGF signaling also plays a role in
preeclampsia. Besides the role of the Rho kinase pathway in cardiovascular
diseases, a role has also been proposed for the modulation of the placental
vasculature. It has been shown that, although the gene expression of ROCK1
and ROCK2 was not different, a higher RhoA mRNA expression was found
in placentae from women who suffered from preeclampsia compared with
placentae from those that were normotensive [209]. Interestingly, the use of
antidepressants has been associated with an increased risk for preeclampsia
60
[121] and this risk also depends on duration of SSRI use during pregnancy;
i.e. women using SSRIs during the entire pregnancy have an increased risk
in comparison to those who discontinue before gestational week 20, and in
comparison with non-users [111, 113, 116].
In study I, down-regulated KTN gene expression was found in SSRItreated women compared with healthy controls. KTN (the full-length kinectin) is mainly expressed in the brain, liver, ovarian, and hematopoietic cells
and is found in the endoplasmic reticulum, where it is responsible for the
transport of vesicles along microtubules [210, 211]. Of interest is that kinectin can interact with RhoA [212], and, as mentioned above, the RhoA signaling pathway is affected in the depressed and antidepressant-treated women.
Normal RhoA signaling is important to prevent pregnancy complications,
where elevated levels of RhoA have been found in placenta from women
suffering from preeclampsia [209]. However, more research is needed to
investigate the consequences for the fetus due to altered gene expression of
KTN1 in the placenta.
The validation of the microarray analysis in study I revealed that the gene
expression of C12orf39 was down-regulated in both depressed and SSRItreated women. Also, a down-regulated expression of DNM1L was found in
women treated with antidepressants. Interestingly, C12orf29 and DNM1L are
found in trophoblasts. C12orf39 is located in the villous trophoblasts and is
mainly expressed in the placenta and brain, suggesting a function for
C12orf39 in these active secretory tissues [213]. These findings suggest that
C12orf39 is involved in the regulation of placenta development through its
role in the biological functions of the trophoblasts and that antenatal depression during pregnancy may influence this development. The fact that in antidepressant-treated women, in study I, the effect is no longer significant,
which may indicate that antidepressants may restore the effects of the depression-induced down-regulation of C12of39.
DNM1L is a GTPase regulating the mitochondrial fission and ablation of
the DNM1L gene induced defects in trophoblast giant cells and cardiomyocytes in mice [214]. Also, brain-specific DNM1L ablation caused developmental defects in the cerebellum [214]. Although these results were found in
knockout mice it is tempting to speculate that the down-regulation of the
DNM1L found in the antidepressant-treated women in study I, might have an
effect on embryo and brain development as well. However, again, this effect
remains to be established.
Study I shows that both depressions per se and SSRI treatment alter the
expression of several genes in the placenta. These results are confirmed in
study II, where both depression and SSRI treatment have an effect on the
protein levels in placental trophoblasts. Taken together, this indicates that
depression and SSRI treatment have an effect on trophoblast development
and placental function, which could ultimately affect pregnancy outcome
and fetal development.
61
Developmental programming
Adequate implantation/placentation is a prerequisite for a pregnancy to occur, and a well-functioning placenta is of major importance for the intrauterine development and growth of a child [215, 216]. Barker [217] hypothesized that diseases that might manifest later in life can be traced back to early development, and a mechanistic role of the placenta in fetal programming
has been discussed during recent years [215]. This phenomenon is usually
referred to as the Barker hypothesis and the relevance of this has also been
confirmed by others [218-221].
The two major hypotheses regarding intrauterine developmental programming are exposure to either malnutrition or stress (i.e. increased cortisol
levels). Indeed, in many mammalian species, maternal under-nutrition or
exposure to stress or glucocorticoid excess leads to reduced birth weight and
may cause persisting hypertension, hyperglycemia, affective dysfunction,
and neuroendocrine abnormalities in the offspring [222, 223]. Both placental
11β-hydroxysteroid dehydrogenase type 2 (11β-HSD-2) and the glucocorticoid receptor (NR3C1) have been implicated in the prenatal glucocorticoid
programming. While glucocorticoids are important for fetal growth and organ maturation, the enzyme 11β-HSD2, which converts cortisol to inactive
cortisol, is strategically positioned in the syncytiotrophoblast to protect the
fetus from the pregnancy-induced glucocorticoid excess in the maternal circulation [224]. Indeed, placental 11β-HSD2 inactivates the majority of maternal glucocorticoids passing to the fetus in humans, and inhibition of fetoplacental 11β-HSD-2 leads to long-standing pertubations in the HPA axis
and anxiety-like behavior in the offspring [225, 226]. Maternal depression
during the third trimester of pregnancy has been associated with increased
methylation of NR3C1, and greater methylation of placental NR3C1 or 11βHSD-2, in association with depression and anxiety, respectively, were related to hypotonia and lethargy in infants [227, 228]. Parenthetically, no differences in NR3C1 or 11β-HSD-2 placental gene expression have been noted in
SSRI users or women with untreated depression (Edvinsson et al., preliminary results).
Based on the results of this thesis, it could be speculated that SSRI exposure may also have fetal programming effects. It has relatively recently been
shown that the placenta may synthesize serotonin and that placental serotonin is accumulated in the fetal forebrain during an important growth period
[106, 229]. While it may be argued that the epidemiological evidence suggesting behavioral disturbances in infants exposed to SSRI would strengthen
such an assumption, the clinical evidence at present is extremely preliminary
[133-137]. Until further long-term follow-ups of exposed children (with the
use of relevant controls) are available, this will merely remain a speculation.
62
Additive or separate effects?
In study I, antenatal depression and antidepressant exposure during pregnancy both had influences on the gene expression of the placenta. In the microarray, 108 genes were differentially expressed in women with antenatal depression, while 109 genes were differentially expressed in antidepressanttreated women. However, only 20 genes were overlapping between depressed women and women on antidepressant treatment. Because we were
not able to match SSRI users and untreated depressed women (due to availability of tissue at that stage), we refrained from direct comparison of these
two groups. With more tissue now available, a repeated microarray experiment could be considered, using only women with untreated and treated
depression. Nevertheless, while the microarray data relied on few individuals, the gene expression pattern suggested mostly separate effects of the two
exposures.
In addition, this pattern of mixed additive and separate effects was also
repeated in study II. Additive effects by depression and SSRI use were found
on certain aspects of NGF signaling (ROCK2 and phosphorylated ROCK2).
Other parts of the signaling pathway, however, showed opposite effects of
depression per se and SSRI use, as in the case of NGF levels in endothelial
cells. Other findings from our lab are also emerging, suggesting that depression and SSRI may have separate effects; for instance, SSRI treatment is
associated with increased placental corticotropin releasing hormone (CRH),
whereas depression on the contrary seems to result in decreased CRH levels,
at least in comparison with the SSRI users (Hannerfors et al., submitted
manuscript).
An alternative interpretation could, of course, be that while we attempt to
control for depression in our experiments, we may not have fully accomplished that.
Although these studies are not able to elucidate the role of these alterations for all of these maternal and fetal complications, these findings nevertheless points towards important distinctions between how the exposure to
depression per se and SSRI treatment of depression affects placental function. However, it remains to be established how these differences influence
the development of the child, and whether these differences are found in the
fetus as well.
Fluoxetine affects embryo development
In study I and II, SSRI treatment in pregnant women altered gene expression
and protein levels in the placenta. This could affect the function of the placenta, and consequently also the developing fetus, but the fetus can also be
exposed to SSRI more directly. Based on this, and considering that serotonin
is involved in embryogenesis [230] and that the serotonin transporter is ex63
pressed in early embryos [92], the effects of fluoxetine (FLX) on early human embryo development were studied (study III).
FLX affected the timing to different embryo developmental stages, with a
significant shorter time between thawing and compaction compared with
controls. The same tendency was noted for times to other embryo developmental stages. The timing between different developmental stages of the
embryo has been shown to be crucial for embryo quality and implantation
rate after assisted reproduction [72-75]. The results in study III indicate that
FLX may alter the time between embryonic gene activation, which is initiated at the 4- to 8-cell stage of the embryo [71], and the formation of morula.
The impact of this alteration on the future development of the fetus remains
to be determined. However, effects of in vitro culturing conditions have been
shown to have an effect on fetal development in cattle and mice [231, 232].
Also, embryo culture medium used in IVF is shown to have an effect on the
perinatal outcome [233, 234]. It has been suggested that these outcomes are
due to epigenetic changes in the placenta and/or fetus [235-237].
In the mass spectrometry analysis of embryos, several important pathways, including cellular growth, proliferation and survival, seem to be affected by FLX. When analyzing proteins secreted into the culture medium
from the embryo, significantly increased levels of uPA were found in medium from FLX-treated embryos. uPA is involved in cell proliferation, differentiation and migration [238] and uPA in complex with its receptor is involved in cell signaling during neuronal migration and neuritogenesis [239].
In study III, uPA was detected by immunohistochemistry in the
trophectoderm of the embryo. Also, NGF was detected in the trophectoderm,
which is in line with the findings in study II, where the levels of NGF were
increased in trophoblasts in placenta from SSRI-treated women compared
with healthy controls. Again, the embryonic trophectoderm will develop into
the placenta and alterations in protein levels in the placenta could lead to
changes in the placental function, which could ultimately affect the development of the fetus.
In conclusion, this study shows that FLX affects embryo development as
well as the embryo protein expression. However, the impact on the developing fetus remains to be investigated.
NGF signaling pathways
In study IV, the NGF signaling pathway in the neuroblastoma cell line SHSY5Y was investigated. Since the wild-type cell line does not respond to
NGF, due to low expression levels of the receptor TrkA, SH-SY5Y was
transfected with exogenous TrkA. In contrast to TPA treatment of SHSY5Y/TrkA, the NGF-induced activation of Raf-1 in SH-SY5Y/TrkA cells
was not mediated via PKC. SH-SY5Y/TrkA cells do respond to NGF treat64
ment by neurite outgrowth and this response was mediated via the Ras-Raf-1
pathway. This is in line with the fact that SH-SY5Y cells treated with NGF
in combination with the growth inhibiting agent aphidicholin, induced an upregulation of the receptor TrkA after 3 days of treatment [240]. After 3 to 4
days of treatment, the cells begun to differentiate in response to NGF.
It was concluded that, since SH-SY5Y/TrkA cells respond to NGF treatment by neuronal differentiation, the Ras-Raf-MAPK pathway was intact in
these cells. However, one could speculate about how accurate it is to use
tumor cell lines to investigate certain aspects of signaling cascades. The neuroblastoma cell line SH-SY5Y is developmentally arrested and defects causing this arrest may interfere with pathways investigated. Even though it was
elucidated that Raf-1 activation was mediated by Ras, other signaling proteins might also be involved. However, the aim of study I was to investigate
whether Ras and/or PKC were involved in NGF-mediated Raf-1 activation
and to conclude that the developmental arrest of these neuroblastoma cells
are not due to a defective signaling cascade down-stream of the TrkA receptor.
The focus of study II was the effects of antenatal depression and SSRI use
during pregnancy on proteins in the NGF signaling pathway. In study I, altered ROCK1 and ROCK2 gene expression in depressed and SSRI-treated
women had been detected. Study IV had revealed that NGF-induced differentiation of neuroblastoma cells was mediated via activated Raf-1 and the
most well-known down-stream mediators for Raf-1 signaling are the MAP
kinases. But Raf-1 can also activate ROCK1 and ROCK2 via ribosomal s6
kinase (RSK) [241]. Therefore, study II also investigated whether the effect
of antenatal depression and SSRI on ROCK1 or ROCK2 were mediated via
RhoA or Raf-1. Immunohistochemical staining of placental sections showed
increased levels of ROCK2 and phosphorylated ROCK2 in SSRI-treated
women compared with healthy controls. SSRI use or depression per se did
not alter the RhoA levels; however, increased levels of phosphorylated Raf-1
were found in stromal cells in SSRI-treated women compared with controls,
indicating that the altered levels of ROCK2 and phosphorylated ROCK2 are
mediated via Raf-1.
NGF can also cross-talk with other signaling pathways, i.e. vascular endothelial growth factor (VEGF), tumor necrosis factor-α (TNFα) and epidermal
growth factor (EGF) [164, 165, 167]. This means that alterations in the NGF
signaling pathway induced by SSRI treatment or depression, could also affect other signaling pathways that may contribute to the associated pregnancy outcomes and fetal development. And vice versa, signaling pathways not
investigated in this thesis could be differentially regulated by SSRI-treatment
or depression per se, and add to the altered gene expressions or protein levels found in this thesis.
65
Conclusions and remarks
Even though significant alterations in placental gene expression and protein
levels due to SSRI treatment and depression were found in this thesis, it is
still difficult to conclude what clinical outcomes these alterations might lead
to. But, the fact that differences in gene expression could also be seen at
protein level further strengthens the hypothesis that depression and SSRI
treatment during pregnancy have an effect. Also, some of the findings in this
thesis could be important for the understanding of the clinical effects that
have been associated with SSRI use.
Also, FLX clearly has an effect on embryo development and protein expression, but it is hard to be certain whether these effects can be directly
correlated to pregnancy outcomes of increased risk of miscarriage, and malformation and behavioral defect in infants. Based on findings from study II,
it seems appropriate for the NICE guidelines to recommend the lowest possible dose of SSRI to women who consider continuing treatment during
pregnancy.
In addition, this thesis has also pointed out that the choice of appropriate
control groups for epidemiological research should be reconsidered, or at
least be interpreted with caution. We chose untreated depression, but instead
of the additive effects we had expected, we ended up having more findings
that separated the two groups. Additional experiments with women who
have discontinued SSRI use prior to their pregnancy might shed light on
what would be the best controls for epidemiological research.
However, taken together, the results in this thesis show that both depression per se and SSRI has an effect on the placenta and that FLX affects embryo development. These findings contribute to the overall understanding
about the effects of SSRI and could be an important part of the knowledge
base used when deciding about whether or not to treat a depressed woman
with SSRI during pregnancy.
66
Summary of conclusions

SSRI treatment of pregnant women alters gene expression in the placenta compared with depressed and healthy women. Also, proteins in the
NGF signaling pathway have altered levels in placenta from SSRItreated women. In placenta from depressed women, gene expression and
NGF signaling protein levels are also altered, but in a different way
compared with SSRI-treated and healthy women. Altered placental gene
and protein expression could lead to an altered placental function, which
in turn could affect the development of the fetus.

Fluoxetine treatment of human embryos alters the timing to different
developmental stages. Also, fluoxetine induces the expression of several
proteins in the embryo, as well as proteins secreted by the embryo, compared with control embryos. This indicates that the embryonal exposure
to fluoxetine in utero could lead to changes in embryo development.

In SH-SY5Y cells transfected with TrkA, both TPA and NGF activates
Raf-1, but to a different extent and via different pathways. TPA activates
Raf-1 via PKC and NGF via Ras. Although SH-SY5Y cells express very
low amounts of endogenous TrkA, the signaling pathway down-stream
of the receptor seems to be intact.
67
Sammanfattning på svenska (Summary in
Swedish)
Under år 2014 hämtade 10,4% av alla kvinnor i barnafödande ålder i Sverige
ut antidepressiv medicin minst en gång. Även om inte alla är gravida då de
börjar sin behandling, hamnar några av dem förr eller senare i ett läge då de
blir gravida och behöver ta ställning till om de ska fortsätta med sina antidepressiva läkemedel eller ej. Depressionssymtom är vanliga under graviditet
och omkring 4 - 7% uppfyller kriterier för diagnosen pågående medelsvår
depression.
En depression äventyrar inte bara mammans hälsa och välmående, utan
kan också påverka fostrets utveckling och ge beteendestörningar hos både
yngre och äldre barn. Havandeskapsförgiftning och förtidsbörd är några av
de graviditetskomplikationer som beskrivits hos deprimerade gravida kvinnor. Barnen kan också födas tillväxthämmade och försenas i sin utveckling.
Den antidepressiva medicin som förskrivs mest under graviditet är s.k. selektiva serotonin-återupptagningshämmare (SSRI), eftersom de är effektiva
och har relativt få biverkningar. Allvarliga missbildningar på grund av behandlingen finns beskrivna men är sällsynta. Under senare år har indikationer på att SSRI kan påverka graviditetsutfallet och fostrets utveckling presenterats. Vidare har en ökad risk för havandeskapsförgiftning, förtidsbörd och
en försämrad funktion av moderkakan (placentan) noterats. Fostret löper en
ökad risk för minskad tillväxt och låg födelsevikt. Barn till kvinnor som
använt SSRI under graviditeten, har svårare att ställa om sig till livet utanför
livmodern och kan få problem med att andas, lågt blodsocker, amning,
kramper och de får oftare än andra vistas på en neonatal vårdavdelning.
En välfungerande placenta är inte bara viktig för näringstillförseln till
barnet, den fungerar också som en barriär för icke önskvärda substanser, så
som mediciner och giftiga ämnen. I delstudie ett och två i denna avhandling
studerades uttrycket av gener och proteiner i vävnadsprover från placenta
från SSRI-behandlade och friska gravida kvinnor, men även hos obehandlade deprimerade gravida kvinnor. Att ha med deprimerade kvinnor i studien
var av största vikt, då det annars skulle vara svårt att avgöra om eventuella
effekter berodde på SSRI-behandlingen eller depressionen i sig. Det visade
sig att både SSRI-behandling och depression påverkade uttrycket av flera
olika gener i placentan jämfört med den friska kontrollgruppen, en del på ett
unikt sätt för de olika grupperna av kvinnor, medan en del gener påverkades
68
på samma sätt i båda grupperna. Två gener som nedreglerades (uttrycktes
mindre av) i placenta hos både SSRI-behandlade och deprimerade kvinnor
var ROCK1 och ROCK2. Dessa gener är involverade i flera olika intracellulära signaleringsvägar, bl.a. i neurotrofiners signalering. Neurotrofiner är
nervtillväxtfaktorer som framför allt styr tillväxten av nervceller och deras
utskott, s.k. neuriter. Delstudie två visade att nivåerna av vissa proteiner av
betydelse i olika signaleringsvägar nedströms om nervtillväxtfaktorn NGF
ökade hos SSRI-behandlade och deprimerade kvinnor jämfört med nivåerna
hos de friska kvinnorna. Mönstret skiljde sig dock något mellan de båda
grupperna. Förändringar i placentans gen- och proteinuttryck kan påverka
hur den fungerar, vilket i sin tur kan påverka hur embryot och fostret utvecklas.
SSRI anses kunna passera placentan och halter av SSRI har uppmätts i
både navelsträngsblod och fostervatten. Detta betyder att fostret exponeras
för SSRI genom blodet, men också genom att svälja fostervatten. I delstudie
tre undersöktes hur direktexponering av SSRI påverkar embryots utveckling
och proteinuttryck. Humana embryon behandlades med SSRI-preparatet
fluoxetin (FLX) och deras utveckling följdes i ett mikroskåp. Det visade sig
att FLX-behandlade embryon genomgår en lite snabbare utveckling jämfört
med obehandlade embryon, vilket mättes genom skillnader i tidsintervall
mellan olika embryologiska utvecklingsstadier. FLX-behandlingen påverkade också vilka proteiner som producerades av embryot, detta gällde såväl
intracellulär produktion som utsöndring. Två speciellt intressanta proteiner
var urokinasliknande plasminogenaktivator (uPA) och NGF. uPA är involverat i bl.a. bildandet av nervceller, men också i tillväxten av neuriter. Både
uPA och NGF återfanns i den del av embryot som senare utvecklas vidare
till placentan.
I delstudie fyra undersöktes vilka proteiner som är involverade i nervtillväxtfaktorn NGFs signalering. En cell-linje från neuroblastom (SHSY5Y/TrkA) användes som modellsystem. Neuroblastomceller är omogna
nervceller, som kan fås att mogna ut om de behandlas med t.ex. NGF. Det
visade sig att NGF-signaleringen i dessa celler framför allt går via proteinerna Ras och Raf-1. Raf-1 signalerar huvudsakligen via MAPK-vägen, men
det har även visats att Raf-1 kan aktivera ROCK1/2, vilket delstudie två i
avhandlingen också indikerar.
Sammanfattningsvis visar denna avhandling att SSRI och depression påverkar både gen- och proteinuttryck i placenta om än på lite olika sätt, samt
att embryots utveckling och proteinuttryck påverkas av FLX. Även om det är
svårt att påvisa vilka kliniska effekter dessa förändringar ger, bör de beaktas
när beslut fattas gällande en gravid kvinnas fortsatta behandling eller ej under en pågående graviditet.
69
Acknowledgements
This thesis work was conducted at the Department of Women’s and Children’s Health, Uppsala University. I would like to thank all the women who
participated in this study. Without you, this research and thesis would not
exist. I would also like to express my sincere gratitude to all the people
around me (professionally and privately, no-one forgotten) who have supported, helped and encouraged me during these years. Thank you!
I especially want to thank:
Helena Åkerud – my main supervisor, for giving me the opportunity to
finish what I started more than 20 years ago and for believing that it was
possible for me to do this. I also want to thank you for your never ending
support and encouragement and for emergency rescues when I have needed
it. I admire your ability to act like a bridge between clinical and preclinical
aspects in research. I am really looking forward to continue working with
you.
Inger Sundström Poromaa – my co-supervisor, for always believing that
everything is possible and all challenges can be mastered. I also appreciate
your enthusiasm and the way you create a positive environment in the lab
and at the department in general, and for always taking the time to listen.
Thank you also for all the feed-back you have given me lately, even though
you have been at a looooong distance for the last half year.
Jocelien Olivier – my other co-supervisor, for your positive attitude and for
support and giving me hope when life has been tough. I miss you in the lab
and I hope we will continue our little email conversations that drifts off into
whatever pops up in our heads. I also want to thank you for bringing a little
bit of the Netherlands back into my life again! Big bug hugs! And thank you
Roel van Eijk for figure number 1. It’s very nice!
Agneta Skoog-Svanberg and Jan Gustafsson, present and former heads of
the Department – thank you for giving me the opportunity to complete my
doctoral thesis at this department. Also, thank you for keeping this department together so that we can continue doing our work without being too
disturbed by “bigger” decisions made further up in the organization.
70
All former and present, closer or further away, colleagues at the KK lab
(mentioned in no specific order at all and I hope I didn’t forget anyone) –
Charlotte Hellgren, Malin Helmestam, Anna-Karin Hannerfors, Karin
Lindgren, Katja Junus, Elin Bannbers, Malin Olsson, Åsa Edvinsson,
Dick Schijvens, Maria Nelander, Emma Håstad, Lena Liljeström, Cecilia Lundin, Evangelina Elenis, Marianne Kördel, Malin Gingnell, Lina
Bergman, Michaela Granfors - for all happy moments and fruitful and
interesting discussions during coffee and lunch. All of you help to create a
warm, supportive and generous atmosphere in the lab. I want to thank Eva
Davey, Lillemor Källström and Lena Moby - for all the gardening tips,
food recipes and for taking such good care of me. I also want to thank my
former roommate Sara Sohlberg - for being such a source of information (is
there anything you don’t know?) and for all the laughter we have had. I also
want to thank you for paving the way for this dissertation. And my present
roommate, Emma Bränn - thank you for your warm nature and for coping
with me when I have been in the worst phase when writing this thesis. I hope
you are not completely discouraged to continue within research. Sara
Nordqvist, thank you for always wondering how things are going regardless
of whether we meet in the supermarket or in the public baths. It means a lot!
And last, but definitely not least, Theodora Kunovac Kallak, thank you for
your endless engagement in the lab, for being so clear headed and for sharing with me your experience from writing a thesis.
Other supervisors at the department - Anna-Karin Wikström, Alkistis
Skalkidou, Maria Jonsson, Matts Olovson and Anneli Stavreus-Evers –
for always sharing your knowledge, taking your time to help out whenever is
needed, and for all uplifting comments. Thank you, Alkistis and AnnaKarin, for inviting me to Crete and Harsa. These trips have been truly inspiring and great fun.
Hans Lindgren – our IT-guru. Thank you for always helping out at short
notice whenever I run into trouble with my computer. And for acting like I
am not a complete ignorant when it comes to these things.
All administrative personnel that have helped me during the years (and always with happy smiling faces) – I especially want to thank Martin Selinus
for keeping track of all the financial stuff and for excellent support during
this dissertation period, Malin Ghanem for explaining all the employment
issues to me, and Susanne Löberg for arranging stamps, helping me with
DiVA and sorting out massage appointments at short notice.
The fantastic people at RPC – Petra Sandell, Ola Sundgren, Ludvig
Ahrén and Cecilia Ålsjö - for your kindness and enthusiasm whenever I
show up in your lab and for sharing your deep knowledge with me. I especially want to thank Julius Hreinson (even though you have now moved to
71
another town) for all the help when initially setting up the embryo study and
Fatma Gülen Yaldir for spending so much time on helping me set up the
experiments and score the embryos in the middle of running your daily business.
Karin Kårehed och Anna-Karin Olsson – by best friends from my ”old”
lab. We have had so much fun (has anyone found the bottle underneath the
Rudbeck laboratory yet?) and we have faced so many things together. AnnaKarin, I will always be grateful for your friendship and back-up. We have to
make sure we meet more often than at the annual Valborg fire. And Karin,
thank you for introducing me to Helena Åkerud. You are the reason why I
came to the KK lab in the first place. I also appreciate our (sometimes deep)
conversations and your tremendous support during these last years, both
professionally and in private life.
Irja and Karl-Erik (Kaggen) Johansson – for always being there for me.
We haven’t met that much lately, but I have always felt that I can call you
and you take me in with open arms. And I promise, I will do my best to keep
the fig tree alive  .
My “oldest” friends, Annica Andersson and Kerstin Fürst Lugne - for still
being there for me after all these years. Even though we haven’t met for
ages, every time we speak on the phone or send emails, it’s like we met yesterday. That’s true friends! I promise we will get together soon! It’s about
time, isn’t it?!
Anna Sillén – my friend from my University studies. Thank you for all the
fun we had during our study time and for still being there for me. Thank you
also for sharing bits and pieces of your life with me and for sending me supportive and happy comments when I need it the most.
My friends from the choirs Allmänna Sången, Coro Piccolo and Stefanoskören (all mentioned and no-one forgotten) –for the energy boost I get
whenever I sing with you guys. These crucibles of people with different
skills (besides singing) and different backgrounds give me so much. I also
want to thank you for all the laughter both during and between rehearsals
and for truly caring for me. I especially want to thank Barbro Björklund,
conductor of Stefanoskören, for always being there for me. For cheering,
comforting and supporting me regardless of whether it’s related to the choir
or life in general. And for letting me come and go to the choir rehearsals as I
please during this last half-a-year. Now, life will get back to normal and I
can be more involved in the choir again.
72
All other friends around me - the “groggbänk/kräftskive-gänget”, the
Godmothers and Godfathers of my children, and our neighbors the
Haagensen family – I hope you understand how much I appreciate you all.
My relatives – aunts, uncles and cousins with their families – for always
reminding me of that deep-down in my heart I am a “värmlänning”, although
I have actually never lived there.
My sister Anna Sundelius and her family, Petter, Isak and Christina (Stina) – for all the fun you bring, interesting (and serious?) discussions at dinners, and for all the love and support you give me. And thank you, Isak and
Stina, for giving me my only nickname outside my family. I will always be
your “moster Muräna”.
My parents, my late mother, Barbro Söderholm, and my father, Sören
Söderholm – who made me who I am. You have always believed in me and
given me the courage to strive towards my goals, or to stop it if things have
had a bad impact on me. I have always been able to talk to you and discuss
everything, big or small, and you have always listened. You are the best
parents a daughter could ask for.
My family – my husband Joachim (Joche) and my daughters Johanna and
Linnea, for always reminding me of what life is all about. There is just not
enough space to express how much you mean to me. Thank you for coping
with me these last six months when I have had my focus elsewhere. I promise I will not work this much again. And thank you Joche for sharing your
life with me and for all the support you have given me during all these years.
Johanna and Linnea, thank you for inspiring me and bringing me joy. You
are the best! I love you!
I know this became a little bit long, but what can one do when one has so
many good friends and colleagues.
This thesis was supported by grants from The Swedish Research Council,
The Marianne and Marcus Wallenberg Foundation, The Children’s Cancer
Foundation of Sweden, The Swedish Cancer Society, The Swedish Society
of Medicine, and the Family Planning Fund in Uppsala.
73
References
1.
Gorman LL, O'Hara MW, Figueiredo B, Hayes S, Jacquemain F,
Kammerer MH, et al. Adaptation of the structured clinical interview for
DSM-IV disorders for assessing depression in women during pregnancy
and post-partum across countries and cultures. The British journal of
psychiatry Supplement. 2004 Feb;46:s17-23.
2. Andersson L, Sundstrom-Poromaa I, Bixo M, Wulff M, Bondestam K,
aStrom M. Point prevalence of psychiatric disorders during the second
trimester of pregnancy: a population-based study. Am J Obstet Gynecol.
2003 Jul;189(1):148-54.
3. Melville JL, Gavin A, Guo Y, Fan MY, Katon WJ. Depressive disorders
during pregnancy: prevalence and risk factors in a large urban sample.
Obstetrics and gynecology. 2010 Nov;116(5):1064-70.
4. Patkar AA, Bilal L, Masand PS. Pharmacotherapy of depression in
pregnancy. Annals of clinical psychiatry : official journal of the American
Academy of Clinical Psychiatrists. 2004 Apr-Jun;16(2):87-100.
5. Johnson GL. Birth outcomes in pregnant women taking fluoxetine. The
Journal of family practice. 1997 Jan;44(1):32.
6. Cantwell R, Clutton-Brock T, Cooper G, Dawson A, Drife J, Garrod D, et
al. Saving Mothers' Lives: Reviewing maternal deaths to make motherhood
safer: 2006-2008. The Eighth Report of the Confidential Enquiries into
Maternal Deaths in the United Kingdom. BJOG : an international journal of
obstetrics and gynaecology. 2011 Mar;118 Suppl 1:1-203.
7. Esscher A, Hogberg U, Haglund B, Essen B. Maternal mortality in Sweden
1988-2007: more deaths than officially reported. Acta obstetricia et
gynecologica Scandinavica. 2013 Jan;92(1):40-6.
8. Goodman JH, Chenausky KL, Freeman MP. Anxiety disorders during
pregnancy: a systematic review. The Journal of clinical psychiatry. 2014
Oct;75(10):e1153-84.
9. Vesga-Lopez O, Blanco C, Keyes K, Olfson M, Grant BF, Hasin DS.
Psychiatric disorders in pregnant and postpartum women in the United
States. Archives of general psychiatry. 2008 Jul;65(7):805-15.
10. Nast I, Bolten M, Meinlschmidt G, Hellhammer DH. How to measure
prenatal stress? A systematic review of psychometric instruments to assess
psychosocial stress during pregnancy. Paediatric and perinatal
epidemiology. 2013 Jul;27(4):313-22.
11. Cox JL, Holden JM, Sagovsky R. Detection of postnatal depression.
Development of the 10-item Edinburgh Postnatal Depression Scale. The
British journal of psychiatry : the journal of mental science. 1987
Jun;150:782-6.
12. Wickberg B, Hwang CP. The Edinburgh Postnatal Depression Scale:
validation on a Swedish community sample. Acta psychiatrica
Scandinavica. 1996 Sep;94(3):181-4.
74
13. Rubertsson C, Borjesson K, Berglund A, Josefsson A, Sydsjo G. The
Swedish validation of Edinburgh Postnatal Depression Scale (EPDS)
during pregnancy. Nordic journal of psychiatry. 2011 Dec;65(6):414-8.
14. National Institute for Health and Care Excellence. Antenatal and postnatal
mental health: Clinical management and service guidance (update).
(Clinical Guideline 123.). 2014 (http://guidance.nice.org.uk/CG192).
15. Cuijpers P, Smit F. Excess mortality in depression: a meta-analysis of
community studies. Journal of affective disorders. 2002 Dec;72(3):227-36.
16. Lyness JM, Niculescu A, Tu X, Reynolds CF, 3rd, Caine ED. The
relationship of medical comorbidity and depression in older, primary care
patients. Psychosomatics. 2006 Sep-Oct;47(5):435-9.
17. Saarni SI, Suvisaari J, Sintonen H, Pirkola S, Koskinen S, Aromaa A, et al.
Impact of psychiatric disorders on health-related quality of life: general
population survey. The British journal of psychiatry : the journal of mental
science. 2007 Apr;190:326-32.
18. Nierenberg AA, Rapaport MH, Schettler PJ, Howland RH, Smith JA,
Edwards D, et al. Deficits in psychological well-being and quality-of-life in
minor depression: implications for DSM-V. CNS neuroscience &
therapeutics. 2010 Aug;16(4):208-16.
19. Moussavi S, Chatterji S, Verdes E, Tandon A, Patel V, Ustun B.
Depression, chronic diseases, and decrements in health: results from the
World Health Surveys. Lancet. 2007 Sep 8;370(9590):851-8.
20. Greenberg PE, Birnbaum HG. The economic burden of depression in the
US: societal and patient perspectives. Expert opinion on pharmacotherapy.
2005 Mar;6(3):369-76.
21. Simon GE, Barber C, Birnbaum HG, Frank RG, Greenberg PE, Rose RM,
et al. Depression and work productivity: the comparative costs of treatment
versus nontreatment. Journal of occupational and environmental medicine /
American College of Occupational and Environmental Medicine. 2001
Jan;43(1):2-9.
22. Mathers CD, Loncar D. Projections of global mortality and burden of
disease from 2002 to 2030. PLoS medicine. 2006 Nov;3(11):e442.
23. Cowen PJ. Classification of depressive disorders. Current topics in
behavioral neurosciences. 2013;14:3-13.
24. Parker G, Fletcher K, Hyett M, Hadzi-Pavlovic D, Barrett M, Synnott H.
Measuring melancholia: the utility of a prototypic symptom approach.
Psychological medicine. 2009 Jun;39(6):989-98.
25. Kendler KS. The diagnostic validity of melancholic major depression in a
population-based sample of female twins. Archives of general psychiatry.
1997 Apr;54(4):299-304.
26. Howard LM, Oram S, Galley H, Trevillion K, Feder G. Domestic violence
and perinatal mental disorders: a systematic review and meta-analysis.
PLoS medicine. 2013;10(5):e1001452.
27. Andersson L, Sundstrom-Poromaa I, Wulff M, Astrom M, Bixo M.
Implications of antenatal depression and anxiety for obstetric outcome.
Obstetrics and gynecology. 2004 Sep;104(3):467-76.
28. Miller LJ, LaRusso EM. Preventing postpartum depression. The Psychiatric
clinics of North America. 2011 Mar;34(1):53-65.
29. Krishnan V, Nestler EJ. The molecular neurobiology of depression. Nature.
2008 Oct 16;455(7215):894-902.
75
30. Krishnan V, Nestler EJ. Linking molecules to mood: new insight into the
biology of depression. The American journal of psychiatry. 2010
Nov;167(11):1305-20.
31. Palazidou E. The neurobiology of depression. British medical bulletin.
2012;101:127-45.
32. Sacher J, Wilson AA, Houle S, Rusjan P, Hassan S, Bloomfield PM, et al.
Elevated brain monoamine oxidase A binding in the early postpartum
period. Archives of general psychiatry. 2010 May;67(5):468-74.
33. Bailara KM, Henry C, Lestage J, Launay JM, Parrot F, Swendsen J, et al.
Decreased brain tryptophan availability as a partial determinant of postpartum blues. Psychoneuroendocrinology. 2006 Apr;31(3):407-13.
34. Doornbos B, Fekkes D, Tanke MA, de Jonge P, Korf J. Sequential
serotonin and noradrenalin associated processes involved in postpartum
blues. Progress in neuro-psychopharmacology & biological psychiatry.
2008 Jul 1;32(5):1320-5.
35. Lommatzsch M, Hornych K, Zingler C, Schuff-Werner P, Hoppner J,
Virchow JC. Maternal serum concentrations of BDNF and depression in the
perinatal period. Psychoneuroendocrinology. 2006 Apr;31(3):388-94.
36. Steudte S, Stalder T, Dettenborn L, Klumbies E, Foley P, Beesdo-Baum K,
et al. Decreased hair cortisol concentrations in generalised anxiety disorder.
Psychiatry research. 2011 Apr 30;186(2-3):310-4.
37. Herbert J. Cortisol and depression: three questions for psychiatry.
Psychological medicine. 2013 Mar;43(3):449-69.
38. Vrshek-Schallhorn S, Doane LD, Mineka S, Zinbarg RE, Craske MG,
Adam EK. The cortisol awakening response predicts major depression:
predictive stability over a 4-year follow-up and effect of depression history.
Psychological medicine. 2013 Mar;43(3):483-93.
39. Kammerer M, Adams D, Castelberg Bv BV, Glover V. Pregnant women
become insensitive to cold stress. BMC pregnancy and childbirth. 2002
Nov 19;2(1):8.
40. Campbell EA, Linton EA, Wolfe CD, Scraggs PR, Jones MT, Lowry PJ.
Plasma corticotropin-releasing hormone concentrations during pregnancy
and parturition. The Journal of clinical endocrinology and metabolism.
1987 May;64(5):1054-9.
41. Allolio B, Hoffmann J, Linton EA, Winkelmann W, Kusche M, Schulte
HM. Diurnal salivary cortisol patterns during pregnancy and after delivery:
relationship to plasma corticotrophin-releasing-hormone. Clinical
endocrinology. 1990 Aug;33(2):279-89.
42. Altemus M, Deuster PA, Galliven E, Carter CS, Gold PW. Suppression of
hypothalmic-pituitary-adrenal axis responses to stress in lactating women.
The Journal of clinical endocrinology and metabolism. 1995
Oct;80(10):2954-9.
43. Skalkidou A, Hellgren C, Comasco E, Sylven S, Sundstrom Poromaa I.
Biological aspects of postpartum depression. Women's health (London,
England). 2012 Nov;8(6):659-72.
44. Hellgren C, Akerud H, Skalkidou A, Sundstrom-Poromaa I. Cortisol
awakening response in late pregnancy in women with previous or ongoing
depression. Psychoneuroendocrinology. 2013 Dec;38(12):3150-4.
76
45. Conroy S, Pariante CM, Marks MN, Davies HA, Farrelly S, Schacht R, et
al. Maternal psychopathology and infant development at 18 months: the
impact of maternal personality disorder and depression. Journal of the
American Academy of Child and Adolescent Psychiatry. 2012
Jan;51(1):51-61.
46. Pearson RM, Evans J, Kounali D, Lewis G, Heron J, Ramchandani PG, et
al. Maternal depression during pregnancy and the postnatal period: risks
and possible mechanisms for offspring depression at age 18 years. JAMA
psychiatry (Chicago, Ill). 2013 Dec;70(12):1312-9.
47. Lovejoy MC, Graczyk PA, O'Hare E, Neuman G. Maternal depression and
parenting behavior: a meta-analytic review. Clinical psychology review.
2000 Aug;20(5):561-92.
48. El Marroun H, Jaddoe VW, Hudziak JJ, Roza SJ, Steegers EA, Hofman A,
et al. Maternal use of selective serotonin reuptake inhibitors, fetal growth,
and risk of adverse birth outcomes. Archives of general psychiatry. 2012
Jul;69(7):706-14.
49. Olivier JD, Akerud H, Kaihola H, Pawluski JL, Skalkidou A, Hogberg U,
et al. The effects of maternal depression and maternal selective serotonin
reuptake inhibitor exposure on offspring. Front Cell Neurosci. 2013;7:73.
50. Waters CS, Hay DF, Simmonds JR, van Goozen SH. Antenatal depression
and children's developmental outcomes: potential mechanisms and
treatment options. European child & adolescent psychiatry. 2014
Oct;23(10):957-71.
51. Grote NK, Bridge JA, Gavin AR, Melville JL, Iyengar S, Katon WJ. A
meta-analysis of depression during pregnancy and the risk of preterm birth,
low birth weight, and intrauterine growth restriction. Archives of general
psychiatry. 2010 Oct;67(10):1012-24.
52. Olivier JD, Akerud H, Sundstrom Poromaa I. Antenatal depression and
antidepressants during pregnancy: Unraveling the complex interactions for
the offspring. European journal of pharmacology. 2014 Aug 2.
53. Uguz F, Sonmez EO, Sahingoz M, Gokmen Z, Basaran M, Gezginc K, et
al. Maternal generalized anxiety disorder during pregnancy and fetal brain
development: a comparative study on cord blood brain-derived
neurotrophic factor levels. Journal of psychosomatic research. 2013
Oct;75(4):346-50.
54. Uguz F, Onder Sonmez E, Sahingoz M, Gokmen Z, Basaran M, Gezginc K,
et al. Neuroinflammation in the fetus exposed to maternal obsessivecompulsive disorder during pregnancy: a comparative study on cord blood
tumor necrosis factor-alpha levels. Comprehensive psychiatry. 2014
May;55(4):861-5.
55. Grant KA, McMahon C, Austin MP, Reilly N, Leader L, Ali S. Maternal
prenatal anxiety, postnatal caregiving and infants' cortisol responses to the
still-face procedure. Developmental psychobiology. 2009 Dec;51(8):62537.
56. Barbey JT, Roose SP. SSRI safety in overdose. The Journal of clinical
psychiatry. 1998;59 Suppl 15:42-8.
57. Kieler H, Artama M, Engeland A, Ericsson O, Furu K, Gissler M, et al.
Selective serotonin reuptake inhibitors during pregnancy and risk of
persistent pulmonary hypertension in the newborn: population based cohort
study from the five Nordic countries. Bmj. 2012;344:d8012.
77
58. Cooper WO, Willy ME, Pont SJ, Ray WA. Increasing use of
antidepressants in pregnancy. Am J Obstet Gynecol. 2007 Jun;196(6):544
e1-5.
59. Hayes RM, Wu P, Shelton RC, Cooper WO, Dupont WD, Mitchel E, et al.
Maternal antidepressant use and adverse outcomes: a cohort study of
228,876 pregnancies. Am J Obstet Gynecol. 2012 Jul;207(1):49 e1-9.
60. Alwan S, Reefhuis J, Rasmussen SA, Friedman JM. Patterns of
antidepressant medication use among pregnant women in a United States
population. Journal of clinical pharmacology. 2011 Feb;51(2):264-70.
61. Iversen LL. Role of transmitter uptake mechanisms in synaptic
neurotransmission. British journal of pharmacology. 1971 Apr;41(4):57191.
62. Heikkinen T, Ekblad U, Palo P, Laine K. Pharmacokinetics of fluoxetine
and norfluoxetine in pregnancy and lactation. Clinical pharmacology and
therapeutics. 2003 Apr;73(4):330-7.
63. Noorlander CW, Ververs FF, Nikkels PG, van Echteld CJ, Visser GH,
Smidt MP. Modulation of serotonin transporter function during fetal
development causes dilated heart cardiomyopathy and lifelong behavioral
abnormalities. PloS one. 2008;3(7):e2782.
64. Hendrick V, Stowe ZN, Altshuler LL, Hwang S, Lee E, Haynes D.
Placental passage of antidepressant medications. The American journal of
psychiatry. 2003 May;160(5):993-6.
65. Loughhead AM, Fisher AD, Newport DJ, Ritchie JC, Owens MJ, DeVane
CL, et al. Antidepressants in amniotic fluid: another route of fetal exposure.
The American journal of psychiatry. 2006 Jan;163(1):145-7.
66. Elder K, Dale. B. In-Vitro Fertilization: Cambridge University Press; 2010.
67. Norwitz ER, Schust DJ, Fisher SJ. Implantation and the survival of early
pregnancy. The New England journal of medicine. 2001 Nov
8;345(19):1400-8.
68. Gude NM, Roberts CT, Kalionis B, King RG. Growth and function of the
normal human placenta. Thrombosis research. 2004;114(5-6):397-407.
69. Hagberg H, Maršál K, Westgren M, Engqvist J. Obstetrik. Lund:
Studentlitteratur; 2008.
70. Gardner DK, Schoolcraft WB. Culture and transfer of human blastocysts.
Current opinion in obstetrics & gynecology. 1999 Jun;11(3):307-11.
71. Braude P, Bolton V, Moore S. Human gene expression first occurs between
the four- and eight-cell stages of preimplantation development. Nature.
1988 Mar 31;332(6163):459-61.
72. Machtinger R, Racowsky C. Morphological systems of human embryo
assessment and clinical evidence. Reproductive biomedicine online. 2013
Mar;26(3):210-21.
73. Meseguer M, Herrero J, Tejera A, Hilligsoe KM, Ramsing NB, Remohi J.
The use of morphokinetics as a predictor of embryo implantation. Human
reproduction. 2011 Oct;26(10):2658-71.
74. Wong CC, Loewke KE, Bossert NL, Behr B, De Jonge CJ, Baer TM, et al.
Non-invasive imaging of human embryos before embryonic genome
activation predicts development to the blastocyst stage. Nature
biotechnology. 2010 Oct;28(10):1115-21.
75. Kaser DJ, Racowsky C. Clinical outcomes following selection of human
preimplantation embryos with time-lapse monitoring: a systematic review.
Human reproduction update. 2014 Sep-Oct;20(5):617-31.
78
76. The Istanbul consensus workshop on embryo assessment: proceedings of an
expert meeting. Human reproduction. 2011 Jun;26(6):1270-83.
77. Conaghan J, Chen AA, Willman SP, Ivani K, Chenette PE, Boostanfar R, et
al. Improving embryo selection using a computer-automated time-lapse
image analysis test plus day 3 morphology: results from a prospective
multicenter trial. Fertil Steril. 2013 Aug;100(2):412-9 e5.
78. Gardner DK, Sakkas D. Assessment of embryo viability: the ability to
select a single embryo for transfer--a review. Placenta. 2003 Oct;24 Suppl
B:S5-12.
79. Hardarson T, Van Landuyt L, Jones G. The blastocyst. Human
reproduction. 2012 Aug;27 Suppl 1:i72-91.
80. Gardner DK, Lane M, Stevens J, Schlenker T, Schoolcraft WB. Blastocyst
score affects implantation and pregnancy outcome: towards a single
blastocyst transfer. Fertil Steril. 2000 Jun;73(6):1155-8.
81. Zhang S, Lin H, Kong S, Wang S, Wang H, Wang H, et al. Physiological
and molecular determinants of embryo implantation. Molecular aspects of
medicine. 2013 Oct;34(5):939-80.
82. Carson DD, Bagchi I, Dey SK, Enders AC, Fazleabas AT, Lessey BA, et
al. Embryo implantation. Developmental biology. 2000 Jul 15;223(2):21737.
83. Altmae S, Reimand J, Hovatta O, Zhang P, Kere J, Laisk T, et al. Research
resource: interactome of human embryo implantation: identification of gene
expression pathways, regulation, and integrated regulatory networks.
Molecular endocrinology (Baltimore, Md). 2012 Jan;26(1):203-17.
84. Lessey BA. Assessment of endometrial receptivity. Fertil Steril. 2011
Sep;96(3):522-9.
85. Mitchell JA, Hammer RE, Goldman H. Serotonin-induced disruption of
implantation in the rat: II. Suppression of decidualization. Biol Reprod.
1983 Aug;29(1):151-6.
86. Nava-Ocampo AA, Koren G. Human teratogens and evidence-based
teratogen risk counseling: the Motherisk approach. Clinical obstetrics and
gynecology. 2007 Mar;50(1):123-31.
87. Polifka JE, Friedman JM. Medical genetics: 1. Clinical teratology in the
age of genomics. CMAJ : Canadian Medical Association journal = journal
de l'Association medicale canadienne. 2002 Aug 6;167(3):265-73.
88. Cassina M, Salviati L, Di Gianantonio E, Clementi M. Genetic
susceptibility to teratogens: state of the art. Reproductive toxicology. 2012
Sep;34(2):186-91.
89. Daud AN, Bergman JE, Bakker MK, Wang H, de Walle HE, Plosch T, et
al. Pharmacogenetics of drug-induced birth defects: the role of
polymorphisms of placental transporter proteins. Pharmacogenomics. 2014
May;15(7):1029-41.
90. Amenta F, Vega JA, Ricci A, Collier WL. Localization of 5hydroxytryptamine-like immunoreactive cells and nerve fibers in the rat
female reproductive system. The Anatomical record. 1992 Jul;233(3):47884.
91. Bodis J, Hartmann G, Tinneberg HR, Torok A, Hanf V, Papenfuss F, et al.
Relationship between the monoamine, progesterone and estradiol content in
follicular fluid of preovulatory graafian follicles after superovulation
treatment. Gynecologic and obstetric investigation. 1993;35(4):232-5.
79
92. Amireault P, Dube F. Serotonin and its antidepressant-sensitive transport in
mouse cumulus-oocyte complexes and early embryos. Biol Reprod. 2005
Aug;73(2):358-65.
93. Basu B, Desai R, Balaji J, Chaerkady R, Sriram V, Maiti S, et al. Serotonin
in pre-implantation mouse embryos is localized to the mitochondria and
can modulate mitochondrial potential. Reproduction (Cambridge, England).
2008 May;135(5):657-69.
94. Cikos S, Fabian D, Makarevich AV, Chrenek P, Koppel J. Biogenic
monoamines in preimplantation development. Human reproduction. 2011
Sep;26(9):2296-305.
95. Il'kova G, Rehak P, Vesela J, Cikos S, Fabian D, Czikkova S, et al.
Serotonin localization and its functional significance during mouse
preimplantation embryo development. Zygote (Cambridge, England). 2004
Aug;12(3):205-13.
96. Khozhai LI, Puchkov VF, Otellin VA. [The effect of a serotonin deficiency
on mammalian embryonic development]. Ontogenez. 1995 SepOct;26(5):350-5.
97. Kim CW, Choe C, Kim EJ, Lee JI, Yoon SY, Cho YW, et al. Dual effects
of fluoxetine on mouse early embryonic development. Toxicology and
applied pharmacology. 2012 Nov 15;265(1):61-72.
98. Montiel JF, Kaune H, Maliqueo M. Maternal-fetal unit interactions and
eutherian neocortical development and evolution. Frontiers in
neuroanatomy. 2013;7:22.
99. Jauniaux E, Van Oppenraaij RH, Burton GJ. Obstetric outcome after early
placental complications. Current opinion in obstetrics & gynecology. 2010
Dec;22(6):452-7.
100. John R, Hemberger M. A placenta for life. Reproductive biomedicine
online. 2012 Jul;25(1):5-11.
101. Oufkir T, Arseneault M, Sanderson JT, Vaillancourt C. The 5-HT 2A
serotonin receptor enhances cell viability, affects cell cycle progression and
activates MEK-ERK1/2 and JAK2-STAT3 signalling pathways in human
choriocarcinoma cell lines. Placenta. 2010 May;31(5):439-47.
102. Cruz MA, Gallardo V, Miguel P, Carrasco G, Gonzalez C. Serotonininduced vasoconstriction is mediated by thromboxane release and action in
the human fetal-placental circulation. Placenta. 1997 Mar-Apr;18(2-3):197204.
103. Bottalico B, Larsson I, Brodszki J, Hernandez-Andrade E, Casslen B,
Marsal K, et al. Norepinephrine transporter (NET), serotonin transporter
(SERT), vesicular monoamine transporter (VMAT2) and organic cation
transporters (OCT1, 2 and EMT) in human placenta from pre-eclamptic and
normotensive pregnancies. Placenta. 2004 Jul;25(6):518-29.
104. Viau M, Lafond J, Vaillancourt C. Expression of placental serotonin
transporter and 5-HT 2A receptor in normal and gestational diabetes
mellitus pregnancies. Reproductive biomedicine online. 2009
Aug;19(2):207-15.
105. Gaspar P, Cases O, Maroteaux L. The developmental role of serotonin:
news from mouse molecular genetics. Nature reviews Neuroscience. 2003
Dec;4(12):1002-12.
106. Bonnin A, Goeden N, Chen K, Wilson ML, King J, Shih JC, et al. A
transient placental source of serotonin for the fetal forebrain. Nature. 2011
Apr 21;472(7343):347-50.
80
107. Hemels ME, Einarson A, Koren G, Lanctot KL, Einarson TR.
Antidepressant use during pregnancy and the rates of spontaneous
abortions: a meta-analysis. The Annals of pharmacotherapy. 2005
May;39(5):803-9.
108. Rahimi R, Nikfar S, Abdollahi M. Pregnancy outcomes following exposure
to serotonin reuptake inhibitors: a meta-analysis of clinical trials.
Reproductive toxicology. 2006 Nov;22(4):571-5.
109. Johansen RL, Mortensen LH, Andersen AM, Hansen AV, StrandbergLarsen K. Maternal use of selective serotonin reuptake inhibitors and risk
of miscarriage - assessing potential biases. Paediatric and perinatal
epidemiology. 2015 Jan;29(1):72-81.
110. Stephansson O, Kieler H, Haglund B, Artama M, Engeland A, Furu K, et
al. Selective serotonin reuptake inhibitors during pregnancy and risk of
stillbirth and infant mortality. JAMA : the journal of the American Medical
Association. 2013 Jan 2;309(1):48-54.
111. Qiu C, Williams MA, Calderon-Margalit R, Cripe SM, Sorensen TK.
Preeclampsia risk in relation to maternal mood and anxiety disorders
diagnosed before or during early pregnancy. American journal of
hypertension. 2009 Apr;22(4):397-402.
112. Toh S, Mitchell AA, Louik C, Werler MM, Chambers CD, Hernandez-Diaz
S. Antidepressant use during pregnancy and the risk of preterm delivery
and fetal growth restriction. Journal of clinical psychopharmacology. 2009
Dec;29(6):555-60.
113. Reis M, Kallen B. Delivery outcome after maternal use of antidepressant
drugs in pregnancy: an update using Swedish data. Psychological medicine.
2010 Oct;40(10):1723-33.
114. Chambers CD, Hernandez-Diaz S, Van Marter LJ, Werler MM, Louik C,
Jones KL, et al. Selective serotonin-reuptake inhibitors and risk of
persistent pulmonary hypertension of the newborn. The New England
journal of medicine. 2006 Feb 9;354(6):579-87.
115. Wen SW, Yang Q, Garner P, Fraser W, Olatunbosun O, Nimrod C, et al.
Selective serotonin reuptake inhibitors and adverse pregnancy outcomes.
Am J Obstet Gynecol. 2006 Apr;194(4):961-6.
116. Toh S, Mitchell AA, Louik C, Werler MM, Chambers CD, Hernandez-Diaz
S. Selective serotonin reuptake inhibitor use and risk of gestational
hypertension. The American journal of psychiatry. 2009 Mar;166(3):320-8.
117. Kallen B. Neonate characteristics after maternal use of antidepressants in
late pregnancy. Archives of pediatrics & adolescent medicine. 2004
Apr;158(4):312-6.
118. Oberlander TF, Warburton W, Misri S, Aghajanian J, Hertzman C.
Neonatal outcomes after prenatal exposure to selective serotonin reuptake
inhibitor antidepressants and maternal depression using population-based
linked health data. Archives of general psychiatry. 2006 Aug;63(8):898906.
119. Suri R, Altshuler L, Hendrick V, Rasgon N, Lee E, Mintz J. The impact of
depression and fluoxetine treatment on obstetrical outcome. Archives of
women's mental health. 2004 Jul;7(3):193-200.
120. Malm H, Klaukka T, Neuvonen PJ. Risks associated with selective
serotonin reuptake inhibitors in pregnancy. Obstetrics and gynecology.
2005 Dec;106(6):1289-96.
81
121. Palmsten K, Setoguchi S, Margulis AV, Patrick AR, Hernandez-Diaz S.
Elevated risk of preeclampsia in pregnant women with depression:
depression or antidepressants? American journal of epidemiology. 2012
May 15;175(10):988-97.
122. Palmsten K, Huybrechts KF, Michels KB, Williams PL, Mogun H,
Setoguchi S, et al. Antidepressant use and risk for preeclampsia.
Epidemiology (Cambridge, Mass). 2013 Sep;24(5):682-91.
123. De Vera MA, Berard A. Antidepressant use during pregnancy and the risk
of pregnancy-induced hypertension. British journal of clinical
pharmacology. 2012 Aug;74(2):362-9.
124. El Marroun H, White T, Verhulst FC, Tiemeier H. Maternal use of
antidepressant or anxiolytic medication during pregnancy and childhood
neurodevelopmental outcomes: a systematic review. European child &
adolescent psychiatry. 2014 Oct;23(10):973-92.
125. Davis RL, Rubanowice D, McPhillips H, Raebel MA, Andrade SE, Smith
D, et al. Risks of congenital malformations and perinatal events among
infants exposed to antidepressant medications during pregnancy.
Pharmacoepidemiology and drug safety. 2007 Oct;16(10):1086-94.
126. Wogelius P, Norgaard M, Gislum M, Pedersen L, Munk E, Mortensen PB,
et al. Maternal use of selective serotonin reuptake inhibitors and risk of
congenital malformations. Epidemiology (Cambridge, Mass). 2006
Nov;17(6):701-4.
127. Chambers CD, Johnson KA, Dick LM, Felix RJ, Jones KL. Birth outcomes
in pregnant women taking fluoxetine. The New England journal of
medicine. 1996 Oct 3;335(14):1010-5.
128. Alwan S, Reefhuis J, Rasmussen SA, Olney RS, Friedman JM. Use of
selective serotonin-reuptake inhibitors in pregnancy and the risk of birth
defects. The New England journal of medicine. 2007 Jun 28;356(26):268492.
129. Louik C, Lin AE, Werler MM, Hernandez-Diaz S, Mitchell AA. Firsttrimester use of selective serotonin-reuptake inhibitors and the risk of birth
defects. The New England journal of medicine. 2007 Jun 28;356(26):267583.
130. Diav-Citrin O, Shechtman S, Weinbaum D, Wajnberg R, Avgil M, Di
Gianantonio E, et al. Paroxetine and fluoxetine in pregnancy: a prospective,
multicentre, controlled, observational study. British journal of clinical
pharmacology. 2008 Nov;66(5):695-705.
131. Kieler H, Malm H, Artama M, Engeland A, Furu K, Gissler M, et al. Use of
antidepressants and association with elective termination of pregnancy:
population based case-control study. BJOG : an international journal of
obstetrics and gynaecology. 2014 Nov 14.
132. Kallen B, Olausson PO. Maternal use of selective serotonin re-uptake
inhibitors and persistent pulmonary hypertension of the newborn.
Pharmacoepidemiology and drug safety. 2008 Aug;17(8):801-6.
133. Croen LA, Grether JK, Yoshida CK, Odouli R, Hendrick V. Antidepressant
use during pregnancy and childhood autism spectrum disorders. Archives
of general psychiatry. 2011 Nov;68(11):1104-12.
134. Rai D, Lee BK, Dalman C, Golding J, Lewis G, Magnusson C. Parental
depression, maternal antidepressant use during pregnancy, and risk of
autism spectrum disorders: population based case-control study. Bmj.
2013;346:f2059.
82
135. Oberlander TF, Reebye P, Misri S, Papsdorf M, Kim J, Grunau RE.
Externalizing and attentional behaviors in children of depressed mothers
treated with a selective serotonin reuptake inhibitor antidepressant during
pregnancy. Archives of pediatrics & adolescent medicine. 2007
Jan;161(1):22-9.
136. Oberlander TF, Papsdorf M, Brain UM, Misri S, Ross C, Grunau RE.
Prenatal effects of selective serotonin reuptake inhibitor antidepressants,
serotonin transporter promoter genotype (SLC6A4), and maternal mood on
child behavior at 3 years of age. Archives of pediatrics & adolescent
medicine. 2010 May;164(5):444-51.
137. Misri S, Reebye P, Kendrick K, Carter D, Ryan D, Grunau RE, et al.
Internalizing behaviors in 4-year-old children exposed in utero to
psychotropic medications. The American journal of psychiatry. 2006
Jun;163(6):1026-32.
138. ACOG Practice Bulletin: Clinical management guidelines for obstetriciangynecologists number 92, April 2008 (replaces practice bulletin number 87,
November 2007). Use of psychiatric medications during pregnancy and
lactation. Obstetrics and gynecology. 2008 Apr;111(4):1001-20.
139. Levi-Montalcini R. The nerve growth factor: thirty-five years later. The
EMBO journal. 1987 May;6(5):1145-54.
140. Aloe L. Rita Levi-Montalcini and the discovery of NGF, the first nerve cell
growth factor. Archives italiennes de biologie. 2011 Jun;149(2):175-81.
141. Cantarella G, Lempereur L, Presta M, Ribatti D, Lombardo G, Lazarovici
P, et al. Nerve growth factor-endothelial cell interaction leads to
angiogenesis in vitro and in vivo. FASEB journal : official publication of
the Federation of American Societies for Experimental Biology. 2002
Aug;16(10):1307-9.
142. Tometten M, Blois S, Arck PC. Nerve growth factor in reproductive
biology: link between the immune, endocrine and nervous system?
Chemical immunology and allergy. 2005;89:135-48.
143. Dissen GA, Romero C, Hirshfield AN, Ojeda SR. Nerve growth factor is
required for early follicular development in the mammalian ovary.
Endocrinology. 2001 May;142(5):2078-86.
144. Dissen GA, Romero C, Paredes A, Ojeda SR. Neurotrophic control of
ovarian development. Microscopy research and technique. 2002 Dec
15;59(6):509-15.
145. Chaves RN, Alves AM, Lima LF, Matos HM, Rodrigues AP, Figueiredo
JR. Role of nerve growth factor (NGF) and its receptors in folliculogenesis.
Zygote (Cambridge, England). 2013 May;21(2):187-97.
146. Dissen GA, Garcia-Rudaz C, Paredes A, Mayer C, Mayerhofer A, Ojeda
SR. Excessive ovarian production of nerve growth factor facilitates
development of cystic ovarian morphology in mice and is a feature of
polycystic ovarian syndrome in humans. Endocrinology. 2009
Jun;150(6):2906-14.
147. Toti P, Ciarmela P, Florio P, Volpi N, Occhini R, Petraglia F. Human
placenta and fetal membranes express nerve growth factor mRNA and
protein. Journal of endocrinological investigation. 2006 Apr;29(4):337-41.
148. Tometten M, Klapp BF, Joachim R, Fest S, Zenclussen AC, Peters EM, et
al. Nerve growth factor and its functional receptor TrkA are up-regulated in
murine decidual tissue of stress-triggered and substance P-mediated
abortion. American journal of reproductive immunology (New York, NY :
1989). 2004 Jan;51(1):86-93.
83
149. Dhobale MV, Pisal HR, Mehendale SS, Joshi SR. Differential expression
of human placental neurotrophic factors in preterm and term deliveries.
International journal of developmental neuroscience : the official journal of
the International Society for Developmental Neuroscience. 2013
Dec;31(8):719-23.
150. Kaplan DR, Hempstead BL, Martin-Zanca D, Chao MV, Parada LF. The
trk proto-oncogene product: a signal transducing receptor for nerve growth
factor. Science (New York, NY). 1991 Apr 26;252(5005):554-8.
151. Chao MV, Hempstead BL. p75 and Trk: A two-receptor system. Trends in
Neurosciences. 1995 7//;18(7):321-6.
152. Chao MV. The p75 neurotrophin receptor. Journal of neurobiology. 1994
Nov;25(11):1373-85.
153. Stephens RM, Loeb DM, Copeland TD, Pawson T, Greene LA, Kaplan
DR. Trk receptors use redundant signal transduction pathways involving
SHC and PLC-gamma 1 to mediate NGF responses. Neuron. 1994
Mar;12(3):691-705.
154. Greene LA, Kaplan DR. Early events in neurotrophin signalling via Trk
and p75 receptors. Curr Opin Neurobiol. 1995 Oct;5(5):579-87.
155. Qui MS, Green SH. PC12 cell neuronal differentiation is associated with
prolonged p21ras activity and consequent prolonged ERK activity. Neuron.
1992 Oct;9(4):705-17.
156. Thomas SM, DeMarco M, D'Arcangelo G, Halegoua S, Brugge JS. Ras is
essential for nerve growth factor- and phorbol ester-induced tyrosine
phosphorylation of MAP kinases. Cell. 1992 Mar 20;68(6):1031-40.
157. Yamaguchi Y, Katoh H, Yasui H, Mori K, Negishi M. RhoA inhibits the
nerve growth factor-induced Rac1 activation through Rho-associated
kinase-dependent pathway. The Journal of biological chemistry. 2001 Jun
1;276(22):18977-83.
158. Sahai E, Olson MF, Marshall CJ. Cross-talk between Ras and Rho
signalling pathways in transformation favours proliferation and increased
motility. The EMBO journal. 2001 Feb 15;20(4):755-66.
159. Sahai E, Ishizaki T, Narumiya S, Treisman R. Transformation mediated by
RhoA requires activity of ROCK kinases. Current biology : CB. 1999 Feb
11;9(3):136-45.
160. Ishizaki T, Maekawa M, Fujisawa K, Okawa K, Iwamatsu A, Fujita A, et
al. The small GTP-binding protein Rho binds to and activates a 160 kDa
Ser/Thr protein kinase homologous to myotonic dystrophy kinase. The
EMBO journal. 1996 Apr 15;15(8):1885-93.
161. Ehrenreiter K, Piazzolla D, Velamoor V, Sobczak I, Small JV, Takeda J, et
al. Raf-1 regulates Rho signaling and cell migration. The Journal of cell
biology. 2005 Mar 14;168(6):955-64.
162. Xing J, Ginty DD, Greenberg ME. Coupling of the RAS-MAPK pathway
to gene activation by RSK2, a growth factor-regulated CREB kinase.
Science (New York, NY). 1996 Aug 16;273(5277):959-63.
163. Xing J, Kornhauser JM, Xia Z, Thiele EA, Greenberg ME. Nerve growth
factor activates extracellular signal-regulated kinase and p38 mitogenactivated protein kinase pathways to stimulate CREB serine 133
phosphorylation. Molecular and cellular biology. 1998 Apr;18(4):1946-55.
164. Hansen-Algenstaedt N, Algenstaedt P, Schaefer C, Hamann A, Wolfram L,
Cingoz G, et al. Neural driven angiogenesis by overexpression of nerve
growth factor. Histochemistry and cell biology. 2006 Jun;125(6):637-49.
84
165. Takei Y, Laskey R. Tumor necrosis factor alpha regulates responses to
nerve growth factor, promoting neural cell survival but suppressing
differentiation of neuroblastoma cells. Mol Biol Cell. 2008 Mar;19(3):85564.
166. Barouch R, Kazimirsky G, Appel E, Brodie C. Nerve growth factor
regulates TNF-alpha production in mouse macrophages via MAP kinase
activation. Journal of leukocyte biology. 2001 Jun;69(6):1019-26.
167. El Zein N, D'Hondt S, Sariban E. Crosstalks between the receptors tyrosine
kinase EGFR and TrkA and the GPCR, FPR, in human monocytes are
essential for receptors-mediated cell activation. Cellular signalling. 2010
Oct;22(10):1437-47.
168. Howe LR, Leevers SJ, Gomez N, Nakielny S, Cohen P, Marshall CJ.
Activation of the MAP kinase pathway by the protein kinase raf. Cell. 1992
Oct 16;71(2):335-42.
169. Kolch W, Heidecker G, Kochs G, Hummel R, Vahidi H, Mischak H, et al.
Protein kinase C alpha activates RAF-1 by direct phosphorylation. Nature.
1993 Jul 15;364(6434):249-52.
170. Corbit KC, Foster DA, Rosner MR. Protein kinase Cdelta mediates
neurogenic but not mitogenic activation of mitogen-activated protein kinase
in neuronal cells. Molecular and cellular biology. 1999 Jun;19(6):4209-18.
171. Marais R, Light Y, Mason C, Paterson H, Olson MF, Marshall CJ.
Requirement of Ras-GTP-Raf complexes for activation of Raf-1 by protein
kinase C. Science (New York, NY). 1998 Apr 3;280(5360):109-12.
172. Fujise A, Mizuno K, Ueda Y, Osada S, Hirai S, Takayanagi A, et al.
Specificity of the high affinity interaction of protein kinase C with a
physiological substrate, myristoylated alanine-rich protein kinase C
substrate. The Journal of biological chemistry. 1994 Dec
16;269(50):31642-8.
173. Ouimet CC, Wang JK, Walaas SI, Albert KA, Greengard P. Localization of
the MARCKS (87 kDa) protein, a major specific substrate for protein
kinase C, in rat brain. The Journal of neuroscience : the official journal of
the Society for Neuroscience. 1990 May;10(5):1683-98.
174. Stumpo DJ, Bock CB, Tuttle JS, Blackshear PJ. MARCKS deficiency in
mice leads to abnormal brain development and perinatal death. Proceedings
of the National Academy of Sciences of the United States of America. 1995
Feb 14;92(4):944-8.
175. Pahlman S, Hoehner JC, Nanberg E, Hedborg F, Fagerstrom S, Gestblom
C, et al. Differentiation and survival influences of growth factors in human
neuroblastoma. European journal of cancer (Oxford, England : 1990).
1995;31A(4):453-8.
176. Henriksson MA, Pahlman S. Neuroblastoma research progress--a 2011
perspective. Seminars in cancer biology. 2011 Oct;21(4):215-6.
177. Pahlman S, Odelstad L, Larsson E, Grotte G, Nilsson K. Phenotypic
changes of human neuroblastoma cells in culture induced by 12-Otetradecanoyl-phorbol-13-acetate. International journal of cancer Journal
international du cancer. 1981 Nov 15;28(5):583-9.
178. Azar CG, Scavarda NJ, Reynolds CP, Brodeur GM. Multiple defects of the
nerve growth factor receptor in human neuroblastomas. Cell growth &
differentiation : the molecular biology journal of the American Association
for Cancer Research. 1990 Sep;1(9):421-8.
85
179. Lavenius E, Gestblom C, Johansson I, Nanberg E, Pahlman S. Transfection
of TRK-A into human neuroblastoma cells restores their ability to
differentiate in response to nerve growth factor. Cell growth &
differentiation : the molecular biology journal of the American Association
for Cancer Research. 1995 Jun;6(6):727-36.
180. Blume-Jensen P, Wernstedt C, Heldin CH, Ronnstrand L. Identification of
the major phosphorylation sites for protein kinase C in kit/stem cell factor
receptor in vitro and in intact cells. The Journal of biological chemistry.
1995 Jun 9;270(23):14192-200.
181. Kutlu T, Karagozler AA, Gozukara EM. Relationship among placental
cadmium, lead, zinc, and copper levels in smoking pregnant women.
Biological trace element research. 2006 Winter;114(1-3):7-17.
182. Zhu MJ, Du M, Nathanielsz PW, Ford SP. Maternal obesity up-regulates
inflammatory signaling pathways and enhances cytokine expression in the
mid-gestation sheep placenta. Placenta. 2010 May;31(5):387-91.
183. Stone WL, Bailey B, Khraisha N. The pathophysiology of smoking during
pregnancy: a systems biology approach. Frontiers in bioscience (Elite
edition). 2014;6:318-28.
184. Rampono J, Proud S, Hackett LP, Kristensen JH, Ilett KF. A pilot study of
newer antidepressant concentrations in cord and maternal serum and
possible effects in the neonate. The international journal of
neuropsychopharmacology / official scientific journal of the Collegium
Internationale Neuropsychopharmacologicum. 2004 Sep;7(3):329-34.
185. Hambiliki F, Hanrieder J, Bergquist J, Hreinsson J, Stavreus-Evers A,
Wanggren K. Glycoprotein 130 promotes human blastocyst development in
vitro. Fertil Steril. 2013 May;99(6):1592-9.
186. Naessen T, Kushnir MM, Chaika A, Nosenko J, Mogilevkina I, Rockwood
AL, et al. Steroid profiles in ovarian follicular fluid in women with and
without polycystic ovary syndrome, analyzed by liquid chromatographytandem mass spectrometry. Fertil Steril. 2010 Nov;94(6):2228-33.
187. Nilsson S, Ramstrom M, Palmblad M, Axelsson O, Bergquist J.
Explorative study of the protein composition of amniotic fluid by liquid
chromatography electrospray ionization Fourier transform ion cyclotron
resonance mass spectrometry. Journal of proteome research. 2004 JulAug;3(4):884-9.
188. Amano M, Fukata Y, Kaibuchi K. Regulation and functions of Rhoassociated kinase. Experimental cell research. 2000 Nov 25;261(1):44-51.
189. Nakagawa O, Fujisawa K, Ishizaki T, Saito Y, Nakao K, Narumiya S.
ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming
protein serine/threonine kinase in mice. FEBS letters. 1996 Aug
26;392(2):189-93.
190. Schofield AV, Bernard O. Rho-associated coiled-coil kinase (ROCK)
signaling and disease. Critical reviews in biochemistry and molecular
biology. 2013 Jul-Aug;48(4):301-16.
191. Di Cunto F, Imarisio S, Hirsch E, Broccoli V, Bulfone A, Migheli A, et al.
Defective neurogenesis in citron kinase knockout mice by altered
cytokinesis and massive apoptosis. Neuron. 2000 Oct;28(1):115-27.
192. Wei L, Roberts W, Wang L, Yamada M, Zhang S, Zhao Z, et al. Rho
kinases play an obligatory role in vertebrate embryonic organogenesis.
Development. 2001 Aug;128(15):2953-62.
86
193. Greenbaum D, Colangelo C, Williams K, Gerstein M. Comparing protein
abundance and mRNA expression levels on a genomic scale. Genome
biology. 2003;4(9):117.
194. Vogel C, Marcotte EM. Insights into the regulation of protein abundance
from proteomic and transcriptomic analyses. Nature reviews Genetics.
2012 Apr;13(4):227-32.
195. Shi J, Wei L. Rho kinases in cardiovascular physiology and
pathophysiology: the effect of fasudil. Journal of cardiovascular
pharmacology. 2013 Oct;62(4):341-54.
196. Shimizu T, Fukumoto Y, Tanaka S, Satoh K, Ikeda S, Shimokawa H.
Crucial role of ROCK2 in vascular smooth muscle cells for hypoxiainduced pulmonary hypertension in mice. Arteriosclerosis, thrombosis, and
vascular biology. 2013 Dec;33(12):2780-91.
197. Laeno AM, Tamashiro DA, Alarcon VB. Rho-associated kinase activity is
required for proper morphogenesis of the inner cell mass in the mouse
blastocyst. Biol Reprod. 2013 Nov;89(5):122.
198. Lunghi L, Ferretti ME, Medici S, Biondi C, Vesce F. Control of human
trophoblast function. Reproductive biology and endocrinology : RB&E.
2007;5:6.
199. Zeck W, Widberg C, Maylin E, Desoye G, Lang U, McIntyre D, et al.
Regulation of placental growth hormone secretion in a human trophoblast
model--the effects of hormones and adipokines. Pediatric research. 2008
Apr;63(4):353-7.
200. Goldstein LD, Reynolds CP, Perez-Polo JR. Isolation of human nerve
growth factor from placental tissue. Neurochemical research. 1978
Apr;3(2):175-83.
201. Heinrich G, Meyer TE. Nerve growth factor (NGF) is present in human
placenta and semen, but undetectable in normal and Paget's disease blood:
measurements with an anti-mouse-NGF enzyme immunoassay using a
recombinant human NGF reference. Biochemical and biophysical research
communications. 1988 Aug 30;155(1):482-6.
202. Kanai-Azuma M, Kanai Y, Matsuda H, Kurohmaru M, Tachi C, Yazaki K,
et al. Nerve growth factor promotes giant-cell transformation of mouse
trophoblast cells in vitro. Biochemical and biophysical research
communications. 1997 Feb 13;231(2):309-15.
203. Frank P, Barrientos G, Tirado-Gonzalez I, Cohen M, Moschansky P, Peters
EM, et al. Balanced levels of nerve growth factor are required for normal
pregnancy progression. Reproduction (Cambridge, England). 2014
Aug;148(2):179-89.
204. Masumoto A, Hirooka Y, Shimokawa H, Hironaga K, Setoguchi S,
Takeshita A. Possible involvement of Rho-kinase in the pathogenesis of
hypertension in humans. Hypertension. 2001 Dec 1;38(6):1307-10.
205. Uehata M, Ishizaki T, Satoh H, Ono T, Kawahara T, Morishita T, et al.
Calcium sensitization of smooth muscle mediated by a Rho-associated
protein kinase in hypertension. Nature. 1997 Oct 30;389(6654):990-4.
206. Mukai Y, Shimokawa H, Matoba T, Kandabashi T, Satoh S, Hiroki J, et al.
Involvement of Rho-kinase in hypertensive vascular disease: a novel
therapeutic target in hypertension. FASEB journal : official publication of
the Federation of American Societies for Experimental Biology. 2001
Apr;15(6):1062-4.
87
207. Moriki N, Ito M, Seko T, Kureishi Y, Okamoto R, Nakakuki T, et al. RhoA
activation in vascular smooth muscle cells from stroke-prone spontaneously
hypertensive rats. Hypertension research : official journal of the Japanese
Society of Hypertension. 2004 Apr;27(4):263-70.
208. Ark M, Yilmaz N, Yazici G, Kubat H, Aktas S. Rho-associated protein
kinase II (rock II) expression in normal and preeclamptic human placentas.
Placenta. 2005 Jan;26(1):81-4.
209. Friel AM, Hynes PG, Sexton DJ, Smith TJ, Morrison JJ. Expression levels
of mRNA for Rho A/Rho kinase and its role in isoprostane-induced
vasoconstriction of human placental and maternal vessels. Reproductive
sciences (Thousand Oaks, Calif). 2008 Feb;15(2):179-88.
210. Bai JZ, Mon Y, Krissansen GW. Kinectin participates in microtubuledependent hormone secretion in pancreatic islet beta-cells. Cell biology
international. 2006 Nov;30(11):885-94.
211. Tran H, Pankov R, Tran SD, Hampton B, Burgess WH, Yamada KM.
Integrin clustering induces kinectin accumulation. Journal of cell science.
2002 May 15;115(Pt 10):2031-40.
212. Hotta K, Tanaka K, Mino A, Kohno H, Takai Y. Interaction of the Rho family
small G proteins with kinectin, an anchoring protein of kinesin motor.
Biochemical and biophysical research communications. 1996 Aug 5;225(1):6974.
213. Wan B, Wang XR, Zhou YB, Zhang X, Huo K, Han ZG. C12ORF39, a
novel secreted protein with a typical amidation processing signal.
Bioscience reports. 2010 Feb;30(1):1-10.
214. Wakabayashi J, Zhang Z, Wakabayashi N, Tamura Y, Fukaya M, Kensler
TW, et al. The dynamin-related GTPase Drp1 is required for embryonic
and brain development in mice. The Journal of cell biology. 2009 Sep
21;186(6):805-16.
215. Nelissen EC, van Montfoort AP, Dumoulin JC, Evers JL. Epigenetics and
the placenta. Human reproduction update. 2011 May-Jun;17(3):397-417.
216. Barker DJ, Bull AR, Osmond C, Simmonds SJ. Fetal and placental size and
risk of hypertension in adult life. Bmj. 1990 Aug 4;301(6746):259-62.
217. Barker DJ. Intrauterine programming of adult disease. Molecular medicine
today. 1995 Dec;1(9):418-23.
218. Leon DA. Fetal growth and adult disease. European journal of clinical
nutrition. 1998 Jan;52 Suppl 1:S72-8; discussion S8-82.
219. Forsen T, Osmond C, Eriksson JG, Barker DJ. Growth of girls who later
develop coronary heart disease. Heart (British Cardiac Society). 2004
Jan;90(1):20-4.
220. Forsen TJ, Eriksson JG, Osmond C, Barker DJ. The infant growth of boys
who later develop coronary heart disease. Annals of medicine.
2004;36(5):389-92.
221. Stein CE, Fall CH, Kumaran K, Osmond C, Cox V, Barker DJ. Fetal growth
and coronary heart disease in south India. Lancet. 1996 Nov
9;348(9037):1269-73.
222. Barker DJ. The developmental origins of adult disease. Journal of the
American College of Nutrition. 2004 Dec;23(6 Suppl):588S-95S.
223. Seckl JR, Holmes MC. Mechanisms of disease: glucocorticoids, their
placental metabolism and fetal 'programming' of adult pathophysiology.
Nature clinical practice Endocrinology & metabolism. 2007 Jun;3(6):47988.
88
224. Chapman K, Holmes M, Seckl J. 11beta-hydroxysteroid dehydrogenases:
intracellular gate-keepers of tissue glucocorticoid action. Physiological
reviews. 2013 Jul;93(3):1139-206.
225. Holmes MC, Abrahamsen CT, French KL, Paterson JM, Mullins JJ, Seckl
JR. The mother or the fetus? 11beta-hydroxysteroid dehydrogenase type 2
null mice provide evidence for direct fetal programming of behavior by
endogenous glucocorticoids. The Journal of neuroscience : the official
journal of the Society for Neuroscience. 2006 Apr 5;26(14):3840-4.
226. Welberg LA, Seckl JR, Holmes MC. Inhibition of 11beta-hydroxysteroid
dehydrogenase, the foeto-placental barrier to maternal glucocorticoids,
permanently programs amygdala GR mRNA expression and anxiety-like
behaviour in the offspring. The European journal of neuroscience. 2000
Mar;12(3):1047-54.
227. Hompes T, Izzi B, Gellens E, Morreels M, Fieuws S, Pexsters A, et al.
Investigating the influence of maternal cortisol and emotional state during
pregnancy on the DNA methylation status of the glucocorticoid receptor
gene (NR3C1) promoter region in cord blood. Journal of psychiatric
research. 2013 Jul;47(7):880-91.
228. Conradt E, Lester BM, Appleton AA, Armstrong DA, Marsit CJ. The roles
of DNA methylation of NR3C1 and 11beta-HSD2 and exposure to
maternal mood disorder in utero on newborn neurobehavior. Epigenetics :
official journal of the DNA Methylation Society. 2013 Dec;8(12):1321-9.
229. Bonnin A, Levitt P. Fetal, maternal, and placental sources of serotonin and
new implications for developmental programming of the brain.
Neuroscience. 2011 Dec 1;197:1-7.
230. Buznikov GA, Lambert HW, Lauder JM. Serotonin and serotonin-like
substances as regulators of early embryogenesis and morphogenesis. Cell
and tissue research. 2001 Aug;305(2):177-86.
231. Delle Piane L, Lin W, Liu X, Donjacour A, Minasi P, Revelli A, et al.
Effect of the method of conception and embryo transfer procedure on midgestation placenta and fetal development in an IVF mouse model. Human
reproduction. 2010 Aug;25(8):2039-46.
232. Farin PW, Piedrahita JA, Farin CE. Errors in development of fetuses and
placentas from in vitro-produced bovine embryos. Theriogenology. 2006
Jan 7;65(1):178-91.
233. Morbeck DE, Paczkowski M, Fredrickson JR, Krisher RL, Hoff HS,
Baumann NA, et al. Composition of protein supplements used for human
embryo culture. Journal of assisted reproduction and genetics. 2014
Dec;31(12):1703-11.
234. Morbeck DE, Krisher RL, Herrick JR, Baumann NA, Matern D, Moyer T.
Composition of commercial media used for human embryo culture. Fertil
Steril. 2014 Sep;102(3):759-66 e9.
235. Dumoulin JC, Land JA, Van Montfoort AP, Nelissen EC, Coonen E,
Derhaag JG, et al. Effect of in vitro culture of human embryos on
birthweight of newborns. Human reproduction. 2010 Mar;25(3):605-12.
236. Nelissen EC, Van Montfoort AP, Coonen E, Derhaag JG, Geraedts JP,
Smits LJ, et al. Further evidence that culture media affect perinatal
outcome: findings after transfer of fresh and cryopreserved embryos.
Human reproduction. 2012 Jul;27(7):1966-76.
89
237. Vergouw CG, Kostelijk EH, Doejaaren E, Hompes PG, Lambalk CB, Schats
R. The influence of the type of embryo culture medium on neonatal
birthweight after single embryo transfer in IVF. Human reproduction. 2012
Sep;27(9):2619-26.
238. Noh H, Hong S, Huang S. Role of urokinase receptor in tumor progression
and development. Theranostics. 2013;3(7):487-95.
239. Lino N, Fiore L, Rapacioli M, Teruel L, Flores V, Scicolone G, et al. uPAuPAR molecular complex is involved in cell signaling during neuronal
migration and neuritogenesis. Developmental dynamics : an official
publication of the American Association of Anatomists. 2014
May;243(5):676-89.
240. Poluha W, Poluha DK, Ross AH. TrkA neurogenic receptor regulates
differentiation of neuroblastoma cells. Oncogene. 1995 Jan 5;10(1):185-9.
241. Silverman E, Frodin M, Gammeltoft S, Maller JL. Activation of p90 Rsk1
is sufficient for differentiation of PC12 cells. Molecular and cellular
biology. 2004 Dec;24(24):10573-83.
90
Acta Universitatis Upsaliensis
Digital Comprehensive Summaries of Uppsala Dissertations
from the Faculty of Medicine 1095
Editor: The Dean of the Faculty of Medicine
A doctoral dissertation from the Faculty of Medicine, Uppsala
University, is usually a summary of a number of papers. A few
copies of the complete dissertation are kept at major Swedish
research libraries, while the summary alone is distributed
internationally through the series Digital Comprehensive
Summaries of Uppsala Dissertations from the Faculty of
Medicine. (Prior to January, 2005, the series was published
under the title “Comprehensive Summaries of Uppsala
Dissertations from the Faculty of Medicine”.)
Distribution: publications.uu.se
urn:nbn:se:uu:diva-248527
ACTA
UNIVERSITATIS
UPSALIENSIS
UPPSALA
2015