You are on page 1of 13

CNS Spectrums

http://journals.cambridge.org/CNS

Additional services for CNS Spectrums:

Email alerts: Click here


Subscriptions: Click here
Commercial reprints: Click here
Terms of use : Click here

The role of reproductive hormones in postpartum depression

Crystal Edler Schiller, Samantha Meltzer-Brody and David R. Rubinow

CNS Spectrums / Volume 20 / Issue 01 / February 2015, pp 48 - 59


DOI: 10.1017/S1092852914000480, Published online: 29 September 2014

Link to this article: http://journals.cambridge.org/abstract_S1092852914000480

How to cite this article:


Crystal Edler Schiller, Samantha Meltzer-Brody and David R. Rubinow (2015). The role of reproductive hormones in
postpartum depression. CNS Spectrums, 20, pp 48-59 doi:10.1017/S1092852914000480

Request Permissions : Click here

Downloaded from http://journals.cambridge.org/CNS, IP address: 203.64.11.45 on 25 Mar 2015


CNS Spectrums (2015), 20, 48–59. © Cambridge University Press 2014
doi:10.1017/S1092852914000480

REVIEW ARTICLE

The role of reproductive hormones in postpartum


depression
Crystal Edler Schiller,* Samantha Meltzer-Brody, and David R. Rubinow

Psychiatry Department, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA

Despite decades of research aimed at identifying the causes of postpartum depression (PPD), PPD remains common,
and the causes are poorly understood. Many have attributed the onset of PPD to the rapid perinatal change in
reproductive hormones. Although a number of human and nonhuman animal studies support the role of reproductive
hormones in PPD, several studies have failed to detect an association between hormone concentrations and PPD. The
purpose of this review is to examine the hypothesis that fluctuations in reproductive hormone levels during pregnancy
and the postpartum period trigger PPD in susceptible women. We discuss and integrate the literature on animal models
of PPD and human studies of reproductive hormones and PPD. We also discuss alternative biological models of PPD to
demonstrate the potential for multiple PPD phenotypes and to describe the complex interplay of changing reproductive
hormones and alterations in thyroid function, immune function, hypothalamic–pituitary–adrenal (HPA) axis function,
lactogenic hormones, and genetic expression that may contribute to affective dysfunction. There are 3 primary lines of
inquiry that have addressed the role of reproductive hormones in PPD: nonhuman animal studies, correlational studies
of postpartum hormone levels and mood symptoms, and hormone manipulation studies. Reproductive hormones
influence virtually every biological system implicated in PPD, and a subgroup of women seem to be particularly
sensitive to the effects of perinatal changes in hormone levels. We propose that these women constitute a “hormone-
sensitive” PPD phenotype, which should be studied independent of other PPD phenotypes to identify underlying
pathophysiology and develop novel treatment targets.

Received 9 June 2014; Accepted 14 August 2014; First published online 29 September 2014
Key words: estrogen, progesterone, allopregnanolone, postpartum depression.

Despite decades of research aimed at identifying the families. Many have speculated that PPD is caused, at
causes of postpartum depression (PPD) and developing least in part, by the rapid change in the reproductive
effective methods of screening, prevention, and treat- hormones estradiol and progesterone before and imme-
ment, PPD remains common, affecting between 7% and diately after delivery.2 Although a number of human and
20% of women following delivery.1 PPD is one of the nonhuman animal studies suggest that changes in
most important public health problems that we can reproductive hormone levels contribute to PPD,3–8
address: it not only affects women at a highly vulnerable several studies have failed to detect an association
time, but it also has deleterious effects on children and between hormone concentrations and PPD symptoms.9–11
For example, cross-sectional human studies examining
between-group differences in ovarian hormones levels
* Address for correspondence: Dr. Crystal Edler Schiller, 234 Medical
and depressive symptoms during the postpartum period
School Wing D, Campus Box 7160, University of North Carolina at
Chapel Hill School of Medicine, Chapel Hill, NC 27599-7160, USA.
have failed to demonstrate an association between
(Email: crystal_schiller@med.unc.edu) absolute estrogen and progesterone concentrations and
We thank Sarah Johnson and Erin Richardson for assisting with the PPD.9–11 In contrast, studies in which PPD was treated
literature review. This work was supported by the UNC Building Inter- with estradiol have successfully reduced depressive
disciplinary Careers in Women’s Health (BIRCWH) Career Development symptoms,5,12 and animal studies have demonstrated
Program (K12 HD001441) and the National Institute of Mental Health of
that estradiol and progesterone withdrawal provoke
the National Institutes of Health under Award Number R21MH101409.
The content is solely the responsibility of the authors and does not
depression-like behavior.4,7,8
necessarily represent the official views of the National Institutes of The mixed results regarding the role of estradiol and
Health. progesterone in PPD are likely due to 3 factors. First, the
REPRODUCTIVE HORMONES AND POSTPARTUM DEPRESSION 49

PPD diagnosis contains enormous variability. A post- imaging and genetic studies as they pertain to the
partum depressive episode can meet the diagnostic hormonal contribution to affective dysregulation during
criteria in a number of different ways, which results in the perinatal period.
women with very different symptom presentations
receiving the same diagnosis. Two women could share
Defining PPD
only one symptom of major depression, experience
timing of onset of the episode during very different The Diagnostic and Statistical Manual of Mental
hormonal conditions (eg, first trimester of pregnancy Disorders, Fifth Edition (DSM-5) expanded the defini-
versus first week postpartum), and both receive a PPD tion of PPD to include major depressive episodes with a
diagnosis. Thus, PPD likely represents a number of perinatal onset as those beginning in either pregnancy or
depressive phenotypes, which may in large part account within the first 4 weeks postpartum.19 Although PPD and
for the difficulty in identifying any one biological or non-perinatal major depressive disorder have the same
hormonal factor central to the disorder. DSM diagnostic criteria (ie, depressed mood, anhedonia,
Second, based on epidemiologic studies of risk, social sleep and appetite disturbance, impaired concentration,
and psychological factors play a large role in the psychomotor disturbance, lethargy, feelings of worthless-
pathogenesis of PPD. For example, decreased social ness or guilt, and suicidal ideation),19 the symptoms of
support, poor quality social support, and poor marital psychomotor agitation and lethargy are more prominent
satisfaction increase the risk of PPD.13–15 The number of in PPD than MDD.20 Additional symptoms of PPD include
previous episodes of depression, a history of PPD, and mood lability and preoccupation with infant well-being.
depression during pregnancy are also significant risk PPD also is frequently associated with symptoms of
factors for PPD.15–17 PPD, like any mood disorder, is anxiety, ruminative thoughts, and panic attacks.21 Indeed,
therefore best seen as a clinical integration of risk most women with PPD have comorbid anxiety disorders.21
and protective factors that culminate in the triggering Recent estimates suggest that 7% of women experience an
of a mood episode in the context of a biological episode of major depression in the first 3 months
(or reproductive) state. following delivery, and the prevalence increases to 20%
Third, the existing studies have used widely diverging when episodes of minor depression are also included.1
methods to examine how reproductive hormones influ- The majority of existing studies suggest that PPD is
ence depressive symptoms: some have examined absolute no more common than non-postpartum depression22;
hormone concentrations in those with and without the however, the largest epidemiological study to date
disorder,9–11 some have examined the change in hormone demonstrated an increased risk of depression during the
levels during pregnancy and the immediate postpartum postpartum period.23
period and the attendant changes in depressive symp- PPD is distinguished from the postpartum blues,
toms,10,18 some have administered hormones to well which is defined by a normative, mild increase in negative
individuals at high risk for PPD,3 and some have used affect in the days following delivery.22 Also distinct from
hormones as a treatment for PPD.5,12 Any biological PPD is postpartum psychosis, which has a rapid onset
model of PPD must account for all 3 of these problems. associated with hallucinations or bizarre delusions, mood
The purpose of this review is to examine the evidence swings, disorganized behavior, and cognitive impair-
for a reproductive hormone model of PPD in which ment.24,25 Many cases of postpartum psychosis are
fluctuating reproductive hormone levels trigger affective manifestations of bipolar disorder,26,27 which may present
dysregulation. We will define PPD and discuss the as mania for the first time during the postpartum period.
diagnostic issues that contribute to difficulties in The perturbation in mood, limited reality testing, and
identifying a single biomarker for the disorder. We will gross functional impairment make postpartum psychosis
discuss alternative biological models of PPD to demon- particularly dangerous for mothers and babies.24
strate the potential for multiple PPD phenotypes and to An important limitation of the DSM criteria for PPD
describe the complex interplay of changing reproductive is that they are not mechanistically based, which is
hormones and alterations in thyroid function, immune why the National Institute of Mental Health (NIMH)
function, HPA axis function, lactogenic hormones, Research Domain Criteria (RDoC) project may be an
and genetic expression that may contribute to affective ideal framework for studying PPD. The RDoC project
dysfunction. We will present animal models and human advocates study of basic dimensions of functioning
studies of reproductive hormones and PPD, and discuss (eg, emotion processing) across multiple units of analysis
methodological issues that have contributed to conflict- (eg, genetic risk and epigenetic modification, limbic
ing findings in the literature. We will provide evidence of system, self-reported affective state) in a specific context
a “hormone-sensitive” PPD phenotype, and discuss the (eg, reproductive hormonal state). The RDoC initiative,
potential neurobiological pathophysiology of PPD for therefore, allows researchers to go beyond the DSM
this group of women. Finally, we will review human brain criteria to identify women who demonstrate patterns of
50 C. E. SCHILLER ET AL.

affective dysregulation related to reproductive states that physiologic levels of gonadal steroids modulate the
and to examine the underlying neurobiological patho- neurocircuitry involved in normal affective states. In a
physiology. For example, while some previous studies study of healthy women, regional cerebral blood flow
have strictly defined PPD according to the DSM criteria, (rCBF) was attenuated in the dorsolateral prefrontal
most have used more inclusive criteria, including cortex, inferior parietal lobule, and posterior inferior
episodes of depression that began before or during temporal cortex during GnRH agonist-induced hypogo-
pregnancy and carried over into the postpartum period nadism, whereas the characteristic pattern of cortical
and episodes with an onset several months following activation re-emerged during both estradiol and proges-
delivery. A study by Forty et al28 demonstrated that terone addback.38 Studies of neural activity during
defining PPD onset within 8 weeks of delivery is optimal the menstrual cycle have compared activation across
for studying the biological triggering of affective dysre- menstrual phases within subjects. Goldstein et al39
gulation. Using this definition, Deligiannidis et al29 found increased amygdala activity during the late
identified functional neural correlates of postpartum follicular phase (higher estradiol levels) compared to
depressive symptoms that occur in the context of the early follicular phase (lower estradiol levels), and
changing reproductive hormone and neurosteroid levels. Protopopescu et al40 demonstrated increased activity in
the medial orbitofrontal cortex (a region that exerts
inhibitory control over amygdalar function) during the
Biological Models of PPD in Humans luteal phase (higher estradiol levels) compared with the
follicular phase (relatively lower estradiol levels). The
Reproductive hormone model of PPD
opposite was true for the lateral orbitofrontal cortex,
Many have hypothesized a role for reproductive hor- suggesting that sensory and evaluative neural functions
mones in PPD because of the temporal association are suppressed in the days prior to menstruation.40
between the substantial and rapid changes in hormone Ovarian hormones also modulate neural reward func-
concentrations that occur at delivery and the onset of tion, with increased activation of the superior orbito-
depressive symptoms.11 However, there are several frontal cortex and amygdala during reward anticipation
more reasons for hypothesizing that reproductive hor- and of the midbrain, striatum, and left ventrolateral
mones play a role in PPD. First, reproductive hormones prefrontal cortex during reward delivery in the follicular
play a major role in basic emotion processing, arousal, phase (compared with the luteal phase).41 Thus, there is
cognition, and motivation, and thus, may contribute to evidence that reproductive hormones influence the
PPD indirectly by influencing the psychological and biological systems and neural circuits implicated in
social risk factors. However, reproductive hormones also depression directly, suggesting that the hormone
regulate each of the biological systems implicated in instability inherent in the perinatal period could
major depression, which suggests that hormones may contribute to mood dysregulation in PPD.
impact a woman’s risk for PPD directly. In the forebrain
and hippocampus, ovariectomy decreases and estradiol
Alternative biological models
increases brain-derived neurotrophic factor (BDNF)
levels,30 which are decreased by depression and stress The hormonal changes of pregnancy and the postpartum
and increased by antidepressants.31 Estradiol also period do not occur in isolation: several other biological
increases cAMP response element-binding (CREB) pro- systems are altered during pregnancy and have been
tein activity32 and the neurotrophin receptor protein implicated in PPD. Alterations in any of these systems
trkA,33 and it decreases GSK-3 beta activity34 in the rat may provoke PPD independent of the changing hormo-
brain similar to antidepressant medications. Progesterone nal milieu, which would suggest that there are a number
also regulates neurotransmitter synthesis, release, and of PPD phenotypes, each with their own relevant
transport.35 For example, progesterone upregulates BDNF biomarkers. Thus far, the search for one biomarker for
expression in the hippocampus and cerebral cortex.36 The the general category of PPD has been elusive, and further
relevance of gonadal steroids to affective regulation is research is needed to determine whether there are
further suggested by modulatory effects on stress and the multiple PPD phenotypes with distinct etiologies. It also
HPA axis, neuroplasticity, cellular energetics, immune stands to reason that perturbations of other biological
activation, and cortical activity,37 all processes that have systems act in concert with rapidly changing hormone
been implicated as dysfunctional in depression. levels to contribute to affective dysregulation. Indeed,
Of particular note are the manifold effects of gonadal reproductive hormones have been shown modulate all of
steroids on brain function as revealed by brain imaging the other biological systems implicated in PPD: thyroid
studies. These studies, by employing positron emission function,42 lactogenic function,43 the hypothalamic–
tomography (PET) or functional magnetic resonance pituitary–adrenal (HPA) axis,44,45 and the immune
imaging (fMRI) in asymptomatic women, have demonstrated system.46 As such, we will discuss the potential contribution
REPRODUCTIVE HORMONES AND POSTPARTUM DEPRESSION 51

of each of these systems to affective dysregulation during axis function normalizes at approximately 12 weeks
pregnancy and the postpartum period, and we will discuss postpartum.74 The effects of pregnancy on HPA axis
the evidence of a genetic susceptibility to PPD. function may be at least partially attributable to the
Thyroid hormones have been proposed as a biomarker effects of estrogen on corticosteroid binding globulin,75
of PPD in large part because of the presumed relationship CRH gene expression,76 and circulating corticotropin
between thyroid dysfunction and major depression47: concentrations.44 Similar to the HPA axis dysregulation
depression accompanies thyroid pathologies,48,49 thyroid seen in nonpuerperal depression, basal concentrations of
dysregulation accompanies depression,50,51 and the plasma cortisol are increased in women with PPD, and
administration of thyroid hormones is thought to suppression of cortisol by dexamethasone is blunted.59
augment and accelerate antidepressant treatment.52,53 In one study, for women with PPD, there was no
Estrogen increases thyroxine-binding globulin (TBG) association between ACTH and cortisol levels in
and consequently increases circulating thyroxine (T4) response to a stress test, whereas in nondepressed
levels.54,55 Thyroid dysfunction is associated with preg- control women, there was a more regulated association
nancy56 and may contribute to PPD in some women.57,58 with cortisol levels rising following the increase in
However, previous studies have failed to detect a clear ACTH.77 Some evidence suggests that higher cortisol
association between thyroid hormone dysregulation and levels at the end of pregnancy are associated with
PPD in the majority of patients.59–61 increased blues symptoms.78 However, it remains
The lactogenic hormones oxytocin and prolactin have unclear whether HPA dysregulation contributes to the
been implicated in PPD.62 Failed lactation and PPD onset of PPD or occurs as an epiphenomenon.
commonly co-occur, and lactogenic hormones regulate Immune dysregulation has been hypothesized to
not only the synthesis and secretion of breast milk, contribute to the development of PPD.79 During
but also maternal behavior and mood. Oxytocin, in pregnancy, anti-inflammatory cytokines responsible for
particular, may account for the shared pathogenesis of immunosuppression are elevated and promote preg-
unplanned early weaning and PPD.63 Estrogen and nancy maintenance, whereas proinflammatory cytokines
progesterone modulate oxytocin mRNA expression in are downregulated. Delivery abruptly shifts the immune
brain regions associated with maternal behavior and system into a proinflammatory state, which lasts for
lactation.64,65 Lower oxytocin levels during the third several weeks. Patients with depression tend to have
trimester are associated with increased depressive higher levels of the proinflammatory cytokines tumor
symptoms during pregnancy63 and the immediate post- necrosis factor (TNF)-α and interleukin (IL)-6,80 and
partum period.66 In a recent study by Stuebe et al,63 administration of cytokines is associated with the onset of
oxytocin secretion during breastfeeding was inversely depression.81 The immune axis is regulated by estradiol.
associated with depression and anxiety symptoms at Estradiol modulates cytokine production, cytokine recep-
8 weeks postpartum. Although depression and anxiety tor expression, activation of effector cells, both the number
symptoms were not associated with breastfeeding success and function of dendritic cells and antigen presenting
in this study, reduced oxytocin may predispose women to cells, and monocyte and macrophage immune function.82
PPD and subsequently lead to unsuccessful breastfeed- Differential patterns of gene expression that are function-
ing. Moreover, low oxytocin levels in mothers with PPD ally related to differences in immunity have been found to
are associated with low oxytocin levels in fathers and distinguish women with PPD from those without.83
their children, suggesting a potential neuroendocrine Although one recent study identified several prenatal
mechanism for the increased risk of depression in immune markers of PPD,84 other studies have failed to
children of depressed mothers.67 Last, oxytocin has detect an association between immune dysfunction and
also been examined as a potential treatment for a wide postpartum depressive symptoms.85–87 Thus, the role of
range of psychiatric disorders, including PPD, but with immune function in PPD remains unclear.
inconsistent findings to date.68,69 Evidence of a genetic vulnerability to PPD has
HPA axis dysfunction has also been implicated in the emerged from family, candidate gene, genome-wide,
pathogenesis of PPD. HPA axis hyperactivity is one of the and gene manipulation studies. Family and twin studies
most consistent findings in the neuroendocrinology of suggest that PPD aggregates in families,28,88 is herita-
depression.70 Hypercortisolism is associated with depres- ble,89 and may be genetically distinct from nonpuerperal
sive symptoms and is corrected with antidepressant depression.89 Although multiple genes likely contribute to
treatment.70 Additionally, the HPA axis is dysregulated PPD, the role of specific genetic variations remains unclear.
by stress and trauma,71 both of which are known Candidate gene studies of PPD have identified several of
precipitants of PPD.13,72,73 Levels of corticotropin- the same polymorphisms implicated in nonpuerperal
releasing hormone (CRH), adrenocorticotropic hormone depression, including the Val66Met polymorphism of
(ACTH), and cortisol increase substantially during the BDNF gene,90,91 the Val158Met polymorphism of
pregnancy and drop 4 days following delivery.74 HPA the COMT gene,92,93 the BcII polymorphism of the
52 C. E. SCHILLER ET AL.

glucocorticoid receptor and the rs242939 polymorphism of anhedonia. One recent study demonstrated that estradiol
the CRH receptor 1,94 the short version of the serotonin- supplementation and withdrawal alone was sufficient to
transporter linked polymorphic region (5-HTTLPR) geno- provoke immobility during the forced swim test and
type,95,96 3 polymorphisms in the serotonin 2A receptor anhedonic behavior during lateral hypothalamic self-
(HTR2A) gene,97 and 3 polymorphisms at protein kinase stimulation.18 Increased depression-like behavior during
C-β (PRKCB).98 There is also evidence of PPD biomarkers the “postpartum” demonstrated in previous studies could
that are theoretically distinct from those of MDD and that therefore be attributed to estradiol withdrawal alone.
implicate reproductive hormones. For example, poly- The effects of estradiol withdrawal on depressive
morphisms in the estrogen receptor α gene (ESR1) have behavior in nonhuman animals are well documented.
been found to be associated with PPD.98,99 However, to Following bilateral ovariectomy, rats demonstrate
date, the results of candidate gene studies of MDD and PPD increased immobility during the forced swim test, and
have failed to replicate,100 and have not been statistically these effects are reversed by treatment with estradiol
significant after correcting for multiple comparisons.97,98 alone.106,107 In addition, reduced immobility following a
Also, there is little consistency in the specific polymorph- single administration of estradiol lasts 2–3 days, and the
isms tested and identified across studies, which means that behavioral effects are the same as those following
any one genetic variant or set of genetic variants is of fluoxetine treatment.108 The antidepressant effects of
limited utility as a diagnostic indicator. Genomic studies estradiol during the forced swim test appear to involve
aim to address some of these shortcomings, and there have selective actions at intracellular estrogen receptor-β
been a few small genomic studies of PPD to date. In a (ERβ) in the ventral tegmental area109 and, in fact,
genome-wide linkage study of 1,210 women, researchers may require ERβ.110 In addition, abrupt estradiol with-
identified genetic variations on chromosomes 1q21.3- drawal following sustained high estradiol levels results in
q32.1 and 9p24.3-p22.3 that may increase susceptibility elevated brain cortical dehydroepiandrosterone sulfate
to PPD.101 Of particular relevance here, the strongest (DHEA-S), a neuroactive steroid synthesized endogenously
implicated gene was Hemicentin 1 (HMCN1), which in the brain that attenuates GABA-ergic activity and may
contains multiple estrogen binding sites. Although the be relevant to postpartum depressive symptoms.111
results were no longer significant after accounting for Chronic administration of estradiol leads to dopamine
multiple comparisons,101 the association between the receptor upregulation and increased presynaptic dopa-
rs2891230 polymorphism of the HMCN1 gene and PPD mine activity in the striatum,112–114 which, when
was confirmed by a subsequent candidate gene study.102 followed by abrupt estradiol withdrawal, leads to dysre-
Similarly, a genome-wide association study yielded a gulation in brain dopaminergic pathways and depressive
third-trimester biomarker panel of 116 transcripts that symptoms.115
predicted PPD onset with 88% accuracy in both the Estradiol-withdrawal models of PPD have two weak-
discovery sample of 62 women and the independent nesses: (1) they have low face validity as models of PPD,
replication sample of 24 women.103 Of these transcripts, given that the human postpartum period is characterized
ESR1 was the only enriched transcription factor binding by a drop in both estradiol and progesterone (whereas
site, again potentially implicating estrogen in the patho- progesterone drops before delivery in rodents), and
genesis of PPD.103 Estrogen-induced DNA methylation (2) they result in depression without the attendant
change has also been implicated in PPD, which suggests anxiety often seen in women with PPD.116 The addition
that women with PPD have an enhanced sensitivity to of progesterone to hormone withdrawal models of PPD is
estrogen-based DNA methylation reprogramming.104 In relevant, given that progesterone withdrawal provokes
order to serve as reliable biomarkers of PPD, these genetic anxiety. As noted above, progesterone metabolites act on
variants will require replication in larger, independent GABA receptors in the brain, producing sedative-like
samples, which is currently an active area of investigation effects by enhancing γ-aminobutyric acid (GABA)
in the field. neurotransmission.117 Abrupt decreases in progesterone
are associated with anxiety,118 and treatment with
progesterone reduces anxiety.119 The anxiolytic effects
Reproductive Hormone Models of PPD in Rodents
of progesterone appear to be mediated by the progester-
Nonhuman animal studies largely support the role of one metabolite allopregnanolone (ALLO).120 Indeed,
reproductive hormones in PPD. Ovariectomized rats postpartum rats show increased depressive behavior
treated with 17β-estradiol and progesterone followed by (increased immobility, decreased struggling and swimming)
vehicle only, in order to induce a hormone withdrawal compared with pregnant rats, and this effect appears to be
state similar to the rodent postpartum period, show mediated by low hippocampal ALLO levels during the
increased immobility during the forced swim test,4,7 a postpartum period.120
behavioral indicator of despair, and decreased sucrose To examine the effects of concurrent estradiol and
consumption and preference,105 a behavioral indicator of progesterone withdrawal, Suda et al8 created a novel
REPRODUCTIVE HORMONES AND POSTPARTUM DEPRESSION 53

rodent model of PPD by administering hormone levels without effect.12 At baseline, 16 of the 23 patients had
that are more consistent with human pregnancy than rat serum estradiol concentrations consistent with gonadal
pregnancy. The concurrent withdrawal of estradiol and failure. All women in the study received sublingual
progesterone resulted in decreased immobility during estradiol treatment for 8 weeks. After the first week,
the forced swim test (ie, less depression-like behavior); depressive symptoms significantly decreased, and by the
however, it also resulted in learned helplessness, which end of the 8 weeks, all patients had achieved depressive
was indicated by a failure to avoid repeated foot shocks.8 symptom scores consistent with clinical recovery.
Animals in this study also showed increased anxiety. Although Ahokas et al12 suggest that postpartum
Taken together, the existing animal models suggest that “gonadal failure” is a risk factor for PPD, they did not
the abrupt withdrawal of estradiol alone produces compare estradiol levels in women with and without
behavioral despair and anhedonia, whereas the concur- PPD. Instead, their data support the notion that, in
rent withdrawal of progesterone and estradiol produces susceptible women, low or declining estradiol levels may
learned helplessness and anxiety. However, these studies trigger PPD, while stable or increasing estradiol levels
do not explain how the same putative stimulus (ie, may ameliorate depressive symptoms. Although these
hormone change) is capable of causing depression in treatment studies suggest a role for estradiol in the
some women and not others. pathogenesis of PPD, they are limited by small sample
sizes and lack of control groups, and are confounded by
Fluctuating Reproductive Hormone Levels the effects of stress, lack of sleep, and homeostatic shifts
attendant to childbirth.
Trigger PPD
In order to assess the role of reproductive hormones
There is no consistent or convincing evidence that in PPD directly, Bloch et al3 created a scaled-down
women who develop PPD experience more rapid post- hormonal model of the puerperium wherein euthymic
partum hormone withdrawal, have lower reproductive women with or without a history of PPD were blindly
hormone concentrations during the postpartum period, administered high-dose estradiol and progesterone
or experience greater reductions in hormone levels from during ovarian suppression and then abruptly with-
pregnancy to the postpartum than women without drawn. Women with a history of PPD showed increasing
PPD.9–11,29,121 The onset of depressive symptoms, how- depressive symptoms during hormone addback and
ever, is temporally coincident with the rapid changes in further exacerbation during hormone withdrawal, but
estradiol and progesterone levels that occur at delivery, women lacking a history of PPD experienced no
leading some researchers to view the change in repro- perturbation of mood despite identical hormonal condi-
ductive hormones as a potent stressor in susceptible tions. Increasing depressive symptoms during both
women.11 hormone addback and withdrawal among those with a
Evidence that a subgroup of women is vulnerable to history of PPD is consistent with research demonstrating
perinatal changes in reproductive hormones comes from that one of the biggest risk factors for PPD is depression
treatment studies that have examined the effects of during pregnancy.15 The advantage of this design is that
administering exogenous estradiol to women at high risk the effects of reproductive hormones on mood were
for PPD or those with active PPD symptoms. In a pilot examined without the confounding biological and
study of 11 women with a history of PPD and no other psychosocial stressors associated with childbirth. The
history of affective disorder, participants were prophy- results provide support for a hormone-sensitive PPD
lactically administered oral Premarin, a conjugated phenotype in which reproductive hormone change alone
estrogen, immediately following delivery to prevent is sufficient to provoke mood dysregulation in otherwise
estrogen withdrawal and the onset of depressive symp- euthymic women.
toms.6 Ten of the 11 women remained well during the Some have hypothesized that the source of PPD
postpartum period and for the first year following vulnerability is in abnormal neural responses to the
delivery.6 A later double-blind, placebo-controlled study normal perinatal fluctuations in reproductive hormones.
of 61 women with PPD that began within 3 months PPD is characterized by abnormal activation of the same
following delivery showed that women treated with brain regions that are implicated in nonpuerperal major
estradiol (n = 34) (delivered via a transdermal patch) depression: the amygdala, insula, striatum, orbitofrontal
improved significantly more than women who received cortex, and dorsomedial prefrontal cortex.122–124 PPD is
placebo (n = 27), although nearly half of the women in also associated with reduced connectivity between the
both groups were also taking antidepressant medication.5 amygdala and prefrontal regions, which implicates
A subsequent study examined the effects of estradiol dysregulation of the limbic system in the neural
treatment on a group of 23 women with severe post- pathophysiology of PPD.123 Despite similar levels of
partum depression, many of whom had attempted treat- circulating progesterone and ALLO to controls, women
ment with antidepressant medication or psychotherapy with PPD also show reduced resting state functional
54 C. E. SCHILLER ET AL.

connectivity between the anterior cingulate cortex, The identification of biomarkers in humans is difficult
amygdala, hippocampus, and dorsolateral prefrontal because of a lack of experimental control over the
cortex in the context of the postnatal decline progester- patient’s environment and genetic background, as well
one and ALLO.29 These neuroimaging studies suggest as inaccessibility of brain tissue required for analysis.
that the neural abnormalities associated with PPD are Gene manipulation studies in nonhuman animals can
unique to the perinatal period and may be unmasked by help model how genetic variants and the environment
changes in circulating reproductive hormone concentra- interact to yield a distinct behavioral phenotype.154
tions. Taken together, the results of the human studies Animal models, which have demonstrated that the
are suggestive of a hormone-sensitive PPD phenotype behavioral effects of maternal care are associated with
characterized by neural abnormalities that are present gene expression changes that persist into adulthood and
during the puerperium when reproductive hormone can be transmitted across generations, provide a potent
concentrations change rapidly. epigenetic model of PPD.154 For example, estradiol
One potential mechanism by which changing repro- withdrawal is clearly associated with estradiol-reversible
ductive hormone levels trigger PPD involves neurosteroid anxiety in a strain-dependent fashion (Schoenrock et al,
regulation of affect. Neurosteroids are metabolites of unpublished manuscript). One genetic knockout model
steroid hormones that are synthesized in the brain and potentially explains both the specificity of affective
nervous system and modulate GABA and glutamate. Two dysregulation during the perinatal period and also the
neurosteroids in particular play a role in affective variation in susceptibility to PPD among women.155 In
dysregulation: dehydroepiandrosterone (DHEA) and this model, Maguire and Mody155 demonstrated a GABAA
ALLO. Abnormal DHEA secretion has been implicated receptor subunit knockout that is behaviorally silent
in major depression,125–129 and DHEA is an effective until an animal is exposed to pregnancy and the
antidepressant in both men and women.130,131 The postpartum state, following which the dam displays
majority of research on neurosteroids in reproductive depression-like behavior and cannibalizes its young.
mood disorders, however, has focused on the progester- Thus, reproductive events may unmask the genetic
one metabolite ALLO. There are several reasons to susceptibility to affective dysregulation. Maguire and
speculate that ALLO plays a role in PPD. ALLO modulates Mody155,156,157 observed that alterations in the GABAA
the GABA receptor, which mediates anxiolysis.132 ALLO receptor δ-subunit occur as ovarian hormone levels
supplementation has anxiolytic effects,133–135 whereas fluctuate during the menstrual cycle, pregnancy, and
ALLO withdrawal produces anxiety and insensitivity to the postpartum period. During pregnancy, the expres-
benzodiazepines.118,136 ALLO levels are decreased in sion of the GABAA receptor δ-subunit is downregulated
depression and increased with successful antidepressant as ALLO levels increase, and at parturition, the expres-
treatment.137–142 ALLO also modulates the biological sion of the GABAA receptor δ-subunit recovers in
processes that are dysregulated in major depressive response to rapidly declining neurosteroid levels.157
disorder, including HPA axis regulation,143–146 neuro- The failure to regulate these receptors during pregnancy
protection,147,148 and immune function.149 ALLO also and the postpartum, consequent to the knockout of the
regulates the neural circuits that are implicated in GABAA receptor δ-subunit, appears to provoke beha-
depression, including the limbic system.150,151 vioral abnormalities consistent with PPD. Thus, as noted
Cortical GABA and ALLO are reduced in postpartum above, GABAA receptor δ-subunit deficient mice exhibit
women, regardless of the presence of PPD, compared normal behaviors prior to pregnancy, but they show
with healthy women in the follicular phase.152 Although insensitivity to ALLO during pregnancy, depression-like
there is no evidence of abnormalities in basal circulating and anxiety-like behavior, and abnormal maternal
ALLO levels in PPD, women with PPD show reduced behavior.155 This model suggests that changes in
resting state functional connectivity between the ante- reproductive hormone concentrations during pregnancy
rior cingulate cortex, amygdala, hippocampus, and and the postpartum period are capable of provoking
dorsolateral prefrontal cortex in the context of the affective dysregulation, particularly in those with a
postnatal decline in ALLO.29 In addition, we recently genetically determined susceptibility.
reported an association between changes in ALLO levels
and depressive symptoms during GnRH agonist-induced
Conclusion
ovarian suppression and ovarian steroid addback in
women with a history of PPD, but not in those without The cross-species role of reproductive hormones in
such a history.153 These studies suggest that, even in depressive behavior suggests a neuroendocrine patho-
the presence of normal absolute levels, perinatal physiology for PPD. PPD, as defined in contemporary
fluctuations in reproductive hormones may precipitate research, includes depression that began during or
symptoms in a vulnerable subpopulation of women as a before pregnancy; depression that occurred in the
result of changing ALLO levels. context of a childhood trauma history, traumatic labor
REPRODUCTIVE HORMONES AND POSTPARTUM DEPRESSION 55

or delivery, subthreshold thyroid dysfunction, psycho- Treatment in PMDD: Steroid Hormone Mechanisms in
social stress, or sleep deprivation; and depression that Premenstrual Dysphoric Disorder,” R01MH081837-01.
co-occurred with obsessive-compulsive disorder, post-
traumatic stress disorder, generalized anxiety disorder,
R E F E R E NC E S :
or personality pathology. Logic would preclude consid-
eration of all of these as the same disorder; consequently, 1. Gavin NI, Gaynes BN, Lohr KN, Meltzer-Brody S, Gartlehner G,
when attempting to understand the contribution of Swinson T. Perinatal depression: a systematic review of prevalence
hormonal signaling to postpartum affective dysregula- and incidence. Obstet Gynecol. 2005; 106(5 Pt 1): 1071–1083.
2. Hendrick V, Altshuler LL, Suri R. Hormonal changes in the
tion, it is therefore necessary to carefully define the study
postpartum and implications for postpartum depression.
population and attempt to characterize and disentangle
Psychosomatics. 1998; 39(2): 93–101.
individual PPD phenotypes. The extant literature supports 3. Bloch M, Schmidt PJ, Danaceau M, Murphy J, Nieman L, Rubinow DR.
the existence of a hormone-sensitive PPD phenotype.3 In Effects of gonadal steroids in women with a history of postpartum
order to study the clinical and neuroendocrine correlates depression. Am J Psychiatry. 2000; 157(6): 924–930.
of this phenotype, some researchers have selected 4. Galea LA, Wide JK, Barr AM. Estradiol alleviates depressive-like
symptoms in a novel animal model of post-partum depression. Behav
women with a history of PPD and without a history of
Brain Res. 2001; 122(1): 1–9.
nonpuerperal depressive episodes.3,18 Although these 5. Gregoire AJ, Kumar R, Everitt B, Henderson AF, Studd JW.
studies are primarily relevant for understanding the risk Transdermal oestrogen for treatment of severe postnatal depression.
of PPD recurrence, they represent the first step toward Lancet. 1996; 347(9006): 930–933.
identifying factors that predict first-onset PPD. There is 6. Sichel DA, Cohen LS, Robertson LM, Ruttenberg A, Rosenbaum JF.
Prophylactic estrogen in recurrent postpartum affective disorder.
sufficient evidence to suggest that reproductive hormone
Biol Psychiatry. 1995; 38(12): 814–818.
fluctuations trigger affective dysregulation in sensitive 7. Stoffel EC, Craft RM. Ovarian hormone withdrawal-induced
women. Even within the hormone-sensitive phenotype, “depression” in female rats. Physiol Behav. 2004; 83(3): 505–513.
alterations in multiple biological systems—the immune 8. Suda S, Segi-Nishida E, Newton SS, Duman RS. A postpartum model
system, HPA axis, and lactogenic hormones—likely in rat: behavioral and gene expression changes induced by ovarian
steroid deprivation. Biol Psychiatry. 2008; 64(4): 311–319.
contribute to the pathophysiology of PPD. Studies are
9. Buckwalter JG, Stanczyk FZ, McCleary CA, et al. Pregnancy, the
underway to disentangle the complex interplay of postpartum, and steroid hormones: effects on cognition and mood.
fluctuating reproductive hormones, neurosteroids, HPA Psychoneuroendocrinology. 1999; 24(1): 69–84.
axis reactivity, neural dysfunction, and genetics with a 10. Heidrich A, Schleyer M, Spingler H, et al. Postpartum blues:
specific focus on identifying genomic, brain, and relationship between not-protein bound steroid hormones in
behavior relationships that contribute to affective dys- plasma and postpartum mood changes. J Affect Disord. 1994; 30(2):
93–98.
function in the context of specific reproductive states.
11. O’Hara MW, Schlechte JA, Lewis DA, Varner MW. Controlled
Consistent with the RDoC mission, this line of research prospective study of postpartum mood disorders: psychological,
represents not only an opportunity to identify novel environmental, and hormonal variables. J Abnorm Psychol. 1991;
treatment targets for PPD but also—critically—the potential 100(1): 63–73.
to prevent PPD in susceptible women. 12. Ahokas A, Kaukoranta J, Wahlbeck K, Aito M. Estrogen deficiency
in severe postpartum depression: successful treatment with
sublingual physiologic 17beta-estradiol: a preliminary study. J Clin
Disclosures Psychiatry. 2001; 62(5): 332–336.
13. Beck CT. Predictors of postpartum depression: an update. Nurs Res.
Crystal Schiller has the following disclosures: UNC 2001; 50(5): 275–285.
Building Interdisciplinary Careers in Women’s Health 14. Collins NL, Dunkel-Schetter C, Lobel M, Scrimshaw SC. Social
support in pregnancy: psychosocial correlates of birth outcomes
(BIRCWH) Career Development Program (K12
and postpartum depression. J Pers Soc Psychol. 1993; 65(6):
HD001441), National Institute of Mental Health of the 1243–1258.
National Institutes of Health—Award Number 15. O’Hara MW, Swain AM. Rates and risk of postpartum depression—a
R21MH101409, and Brain and Behavior Research meta-analysis. Int Rev Psychiatry. 1996; 8(1): 37–54.
Foundation. Samantha Meltzer-Brody has the following 16. Bloch M, Rotenberg N, Koren D, Klein E. Risk factors for early
disclosures: Foundation of Hope, research, grant. David postpartum depressive symptoms. Gen Hosp Psychiatry. 2006;
28(1): 3–8.
Rubinow has the following disclosures: active grants
17. O’Hara MW, Neunaber DJ, Zekoski EM. Prospective study of
from NIH/NIMH: “Postdoctoral Training in Reproduc- postpartum depression: prevalence, course, and predictive factors.
tive Mood Disorders,” T32 MH93315; “Identifying J Abnorm Psychol. 1984; 93(2): 158–171.
Biomarkers for Post-Partum Depression in African- 18. Schiller CE, O’Hara MW, Rubinow DR, Johnson AK. Estradiol
American Women.” R01MH095992; “Neuroendocrine modulates anhedonia and behavioral despair in rats and negative
affect in a subgroup of women at high risk for postpartum
Mechanisms of Reproductive Hormone Related Affective
depression. Physiol Behav. 2013; 119: 137–144.
Dysfunction,” R21MH101409; “Depression, Estrogen 19. American Psychiatric Association. Diagnostic and Statistical
Replacement, and Cardiovascular Health in the Peri- Manual of Mental Disorders. 5th ed. Arlington, VA: American
menopause,” 1R01MH087619-01A1; “Continuous OC Psychiatric Publishing; 2013.
56 C. E. SCHILLER ET AL.

20. Bernstein IH, Rush AJ, Yonkers K, et al. Symptom features of 42. Santin AP, Furlanetto TW. Role of estrogen in thyroid function and
postpartum depression: are they distinct? Depress Anxiety. 2008; growth regulation. J Thyroid Res. 2011; 2011: e875125.
25(1): 20–26. 43. Schumacher M, Coirini H, Pfaff DW, McEwen BS. Behavioral effects
21. Wisner KL, Sit DY, McShea MC, et al. Onset timing, thoughts of self- of progesterone associated with rapid modulation of oxytocin
harm, and diagnoses in postpartum women with screen-positive receptors. Science. 1990; 250(4981): 691–694.
depression findings. JAMA Psychiatry. 2013; 70(5): 490–498. 44. Walf AA, Frye CA. Antianxiety and antidepressive behavior
22. O’Hara MW. Postpartum Depression: Causes and Consequences. produced by physiological estradiol regimen may be modulated by
New York: Springer-Verlag; 1995. hypothalamic–pituitary–adrenal axis activity.
23. Vesga-Lopez O, Blanco C, Keyes K, Olfson M, Grant BF, Hasin DS. Neuropsychopharmacology. 2005; 30(7): 1288–1301.
Psychiatric disorders in pregnant and postpartum women in the 45. Roca CA, Schmidt PJ, Altemus M, et al. Differential menstrual cycle
United States. Arch Gen Psychiatry. 2008; 65(7): 805–815. regulation of hypothalamic-pituitary-adrenal axis in women with
24. Sit D, Rothschild AJ, Wisner KL. A review of postpartum psychosis. premenstrual syndrome and controls. J Clin Endocrinol Metab.
J Womens Health (Larchmt). 2006; 15(4): 352–368. 2003; 88(7): 3057–3063.
25. Wisner KL, Peindl K, Hanusa BH. Symptomatology of affective and 46. Butts CL, Sternberg EM. Neuroendocrine factors alter host defense
psychotic llnesses related to childbearing. J Affect Disord. 1994; by modulating immune function. Cell Immunol. 2008; 252(1–2):
30(2): 77–87. 7–15.
26. Brockington IF, Cernik KF, Schofield EM, Downing AR, Francis 47. Bunevicius R, Kusminskas L, Mickuviene N, Bunevicius A, Pedersen CA,
AF, Keelan C. Puerperal psychosis: phenomena and diagnosis. Arch Pop VJM. Depressive disorder and thyroid axis functioning during
Gen Psychiatry. 1981; 38(7): 829–833. pregnancy. World J Biol Psychiatry. 2009; 10(4): 324–329.
27. Kendell RE, Chalmers JC, Platz C. Epidemiology of puerperal 48. Placidi GPA, Boldrini M, Patronelli A, et al. Prevalence of
psychoses. Br J Psychiatry. 1987; 150(5): 662–673. psychiatric disorders in thyroid diseased patients.
28. Forty L, Jones L, Macgregor S, et al. Familiality of postpartum Neuropsychobiology. 1998; 38(4): 222–225.
depression in unipolar disorder: results of a family study. Am J 49. Gulseren S, Gulseren L, Hekimsoy Z, Cetinay P, Ozen C, Tokatlioglu B.
Psychiatry. 2006; 163(9): 1549–1553. Depression, anxiety, health-related quality of life, and disability in
29. Deligiannidis KM, Sikoglu EM, Shaffer SA, et al. GABAergic patients with overt and subclinical thyroid dysfunction. Arch Med Res.
neuroactive steroids and resting-state functional connectivity in 2006; 37(1): 133–139.
postpartum depression: a preliminary study. J Psychiatr Res. 2013; 50. Berent D, Zboralski K, Orzechowska A, Gałecki P. Thyroid
47(6): 816–828. hormones association with depression severity and clinical outcome
30. Sohrabji F, Miranda RC, Toran-Allerand CD. Estrogen differentially in patients with major depressive disorder. Mol Biol Rep. 2014;
regulates estrogen and nerve growth factor receptor mRNAs in adult 41(4): 2419–2425.
sensory neurons. J Neurosci. 1994; 14(2): 459–471. 51. Nemeroff CB, Simon JS, Haggerty JJ Jr, Evans DL. Antithyroid
31. Shimizu E, Hashimoto K, Okamura N, et al. Alterations of serum antibodies in depressed patients. Am J Psychiatry. 1985; 142(7):
levels of brain-derived neurotrophic factor (BDNF) in depressed 840–843.
patients with or without antidepressants. Biol Psychiatry. 2003; 52. Cooper-Kazaz R, Lerer B. Efficacy and safety of triiodothyronine
54(1): 70–75. supplementation in patients with major depressive disorder treated
32. Zhou Y, Watters JJ, Dorsa DM. Estrogen rapidly induces the with specific serotonin reuptake inhibitors. Int J
phosphorylation of the cAMP response element binding protein in Neuropsychopharmacol. 2008; 11(5): 685–699.
rat brain. Endocrinology. 1996; 137(5): 2163–2166. 53. Cooper-Kazaz R, Apter JT, Cohen R, et al. Combined treatment with
33. Sohrabji F, Greene LA, Miranda RC, Toran-Allerand CD. Reciprocal sertraline and liothyronine in major depression: a randomized,
regulation of estrogen and NGF receptors by their ligands in double-blind, placebo-controlled trial. Arch Gen Psychiatry. 2007;
PC12 cells. J Neurobiol. 1994; 25(8): 974–988. 64(6): 679–688.
34. Cardona-Gomez P, Perez M, Avila J, Garcia-Segura L, Wandosell F. 54. Ben-Rafael Z, Struass JF 3rd, Arendash-Durand B, Mastroianni L Jr,
Estradiol inhibits GSK3 and regulates interaction of estrogen Flickinger GL. Changes in thyroid function tests and sex hormone
receptors, GSK3, and beta-catenin in the hippocampus. Mol Cell binding globulin associated with treatment by gonadotropin. Fertil
Neurosci. 2004; 25(3): 363–373. Steril. 1987; 48(2): 318–320.
35. Finocchi C, Ferrari M. Female reproductive steroids and neuronal 55. Arafah BM. Increased need for thyroxine in women with
excitability. Neurol Sci. 2011; 32(Suppl 1): S31–S35. hypothyroidism during estrogen therapy. N Engl J Med. 2001;
36. Pluchino N, Russo M, Santoro AN, Litta P, Cela V, Genazzani AR. 344(23): 1743–1749.
Steroid hormones and BDNF. Neuroscience. 2013; 239: 271–279. 56. Vaidya B, Anthony S, Bilous M, et al. Detection of thyroid
37. Rubinow DR, Girdler SS. Hormones, heart disease, and health: dysfunction in early pregnancy: universal screening or targeted
individualized medicine versus throwing the baby out with the high-risk case finding? J Clin Endocrinol Metab. 2007; 92(1):
bathwater. Depress Anxiety. 2011; 28(4): 282–296. 203–207.
38. Berman KF, Schmidt PJ, Rubinow DR, et al. Modulation of 57. Pedersen CA, Johnson JL, Silva S, et al. Antenatal thyroid correlates
cognition-specific cortical activity by gonadal steroids: a positron- of postpartum depression. Psychoneuroendocrinology. 2007; 32(3):
emission tomography study in women. Proc Natl Acad Sci U S A. 235–245.
1997; 94(16): 8836–8841. 58. Pedersen CA, Stern RA, Pate J, Senger MA, Bowes WA, Mason GA.
39. Goldstein JM, Jerram M, Poldrack R, et al. Hormonal cycle Thyroid and adrenal measures during late pregnancy and the
modulates arousal circuitry in women using functional magnetic puerperium in women who have been major depressed or who
resonance imaging. J Neurosci. 2005; 25(40): 9309–9316. become dysphoric postpartum. J Affect Disord. 1993; 29(2–3):
40. Protopopescu X, Pan H, Altemus M, et al. Orbitofrontal cortex activity 201–211.
related to emotional processing changes across the menstrual cycle. 59. Bloch M, Daly RC, Rubinow DR. Endocrine factors in the etiology of
Proc Natl Acad Sci U S A. 2005; 102(44): 16060–16065. postpartum depression. Compr Psychiatry. 2003; 44(3): 234–246.
41. Dreher JC, Schmidt PJ, Kohn P, Furman D, Rubinow D, Berman KF. 60. Albacar G, Sans T, Martín-Santos R, et al. Thyroid function 48h after
Menstrual cycle phase modulates reward-related neural function delivery as a marker for subsequent postpartum depression.
in women. Proc Natl Acad Sci U S A. 2007; 104(7): 2465–2470. Psychoneuroendocrinology. 2010; 35(5): 738–742.
REPRODUCTIVE HORMONES AND POSTPARTUM DEPRESSION 57

61. Kent GN, Stuckey BG, Allen JR, Lambert T, Gee V. Postpartum 81. Raison CL, Capuron L, Miller AH. Cytokines sing the blues:
thyroid dysfunction: clinical assessment and relationship to inflammation and the pathogenesis of depression. Trends Immunol.
psychiatric affective morbidity. Clin Endocrinol (Oxf). 1999; 51(4): 2006; 27(1): 24–31.
429–438. 82. Cunningham M, Gilkeson G. Estrogen receptors in immunity and
62. Stuebe AM, Grewen K, Pedersen CA, Propper C, Meltzer-Brody S. autoimmunity. Clin Rev Allergy Immunol. 2011; 40(1): 66–73.
Failed lactation and perinatal depression: common problems with 83. Segman RH, Goltser-Dubner T, Weiner I, et al. Blood mononuclear
shared neuroendocrine mechanisms? J Womens Health (Larchmt). cell gene expression signature of postpartum depression. Mol
2012; 21(3): 264–272. Psychiatry. 2010; 15(1): 93–100, 102.
63. Stuebe AM, Grewen K, Meltzer-Brody S. Association between 84. Krause D, Jobst A, Kirchberg F, et al. Prenatal immunologic
maternal mood and oxytocin response to breastfeeding. J Womens predictors of postpartum depressive symptoms: a prospective study
Health (Larchmt). 2013; 22(4): 352–361. for potential diagnostic markers. Eur Arch Psychiatry Clin Neurosci.
64. Amico JA, Crowley RS, Insel TR, Thomas A, O’Keefe JA. Effect of In press. DOI: 10.1007/s00406-014-0494-8.
gonadal steroids upon hypothalamic oxytocin expression. Adv Exp 85. Blackmore ER, Moynihan JA, Rubinow DR, Pressman EK, Gilchrist M,
Med Biol. 1995; 395: 23–35. O’Connor TG. Psychiatric symptoms and proinflammatory cytokines
65. Broad KD, Kendrick KM, Sirinathsinghji DJS, Keverne EB. Changes in pregnancy. Psychosom Med. 2011; 73(8): 656–663.
in oxytocin immunoreactivity and mRNA expression in the sheep 86. Blackmore ER, Groth SW, Chen DG, Gilchrist MA, O’Connor TG,
brain during pregnancy, parturition and lactation and in response to Moynihan JA. Depressive symptoms and proinflammatory cytokines
oestrogen and progesterone. J Neuroendocrinol. 1993; 5(4): 435–444. across the perinatal period in African American women.
66. Skrundz M, Bolten M, Nast I, Hellhammer DH, Meinlschmidt G. J Psychosom Obstet Gynaecol. 2013; 35(1): 8–15.
Plasma oxytocin concentration during pregnancy is associated with 87. Okun ML, Luther J, Prather AA, Perel JM, Wisniewski S, Wisner KL.
development of postpartum depression. Changes in sleep quality, but not hormones predict time to postpartum
Neuropsychopharmacology. 2011; 36(9): 1886–1893. depression recurrence. J Affect Disord. 2011; 130(3): 378–384.
67. Apter-Levy Y, Feldman M, Vakart A, Ebstein RP, Feldman R. Impact 88. Murphy-Eberenz K, Zandi PP, March D, et al. Is perinatal
of maternal depression across the first 6 years of life on the child’s depression familial? J Affect Disord. 2006; 90(1): 49–55.
mental health, social engagement, and empathy: the moderating 89. Treloar SA, Martin NG, Bucholz KK, Madden PA, Heath AC.
role of oxytocin. Am J Psychiatry. 2013; 170(10): 1161–1168. Genetic influences on post-natal depressive symptoms: findings
68. Kim S, Soeken TA, Cromer SJ, Martinez SR, Hardy LR, Strathearn L. from an Australian twin sample. Psychol Med. 1999; 29(3): 645–654.
Oxytocin and postpartum depression: delivering on what’s known 90. Figueira P, Malloy-Diniz L, Campos SB, et al. An association study
and what’s not. Brain Res. 2014; 1580: 219–232. between the Val66Met polymorphism of the BDNF gene and
69. Mah BL, Van IJzendoorn MH, Smith R, Bakermans-Kranenburg MJ. postpartum depression. Arch Womens Ment Health. 2010; 13(3):
Oxytocin in postnatally depressed mothers: its influence on mood 285–289.
and expressed emotion. Prog Neuropsychopharmacol Biol 91. Comasco E, Sylvén SM, Papadopoulos FC, Oreland L, Sundström-
Psychiatry. 2013; 40: 267–272. Poromaa I, Skalkidou A. Postpartum depressive symptoms and the
70. Nestler EJ, Barrot M, DiLeone RJ, Eisch AJ, Gold SJ, Monteggia LM. BDNF Val66Met functional polymorphism: effect of season of
Neurobiology of depression. Neuron. 2002; 34(1): 13–25. delivery. Arch Womens Ment Health. 2011; 14(6): 453–463.
71. Heim C, Newport DJ, Wagner D, Wilcox MM, Miller AH, Nemeroff CB. 92. Comasco E, Sylvén SM, Papadopoulos FC, Sundström-Poromaa I,
The role of early adverse experience and adulthood stress in the Oreland L, Skalkidou A. Postpartum depression symptoms: a case-
prediction of neuroendocrine stress reactivity in women: a multiple control study on monoaminergic functional polymorphisms and
regression analysis. Depress Anxiety. 2002; 15(3): 117–125. environmental stressors. Psychiatr Genet. 2011; 21(1): 19–28.
72. Records K, Rice MJ. A comparative study of postpartum depression 93. Alvim-Soares A, Miranda D, Campos SB, Figueira P, Romano-Silva MA,
in abused and nonabused women. Arch Psychiatr Nurs. 2005; 19(6): Correa H. Postpartum depression symptoms associated with Val158Met
281–290. COMT polymorphism. Arch Womens Ment Health. 2013; 16(4):
73. Ross LE, Dennis CL. The prevalence of postpartum depression among 339–340.
women with substance use, an abuse history, or chronic illness: a 94. Engineer N, Darwin L, Nishigandh D, Ngianga-Bakwin K, Smith SC,
systematic review. J Womens Health (Larchmt). 2009; 18(4): 475–486. Grammatopoulos DK. Association of glucocorticoid and type 1
74. Mastorakos G, Ilias I. Maternal and fetal hypothalamic-pituitary- corticotropin-releasing hormone receptors gene variants and risk
adrenal axes during pregnancy and postpartum. Ann N Y Acad Sci. for depression during pregnancy and post-partum. J Psychiatr Res.
2003; 997: 136–149. 2013; 47(9): 1166–1173.
75. Young EA. Glucocorticoid cascade hypothesis revisited: role of 95. Binder EB, Newport DJ, Zach EB, et al. A serotonin transporter gene
gonadal steroids. Depression. 1995; 3(1–2): 20–27. polymorphism predicts peripartum depressive symptoms in an at-
76. Vamvakopoulos NC, Chrousos GP. Evidence of direct estrogenic risk psychiatric cohort. J Psychiatr Res. 2010; 44(10): 640–646.
regulation of human corticotropin-releasing hormone gene 96. Mitchell C, Notterman D, Brooks-Gunn J, et al. Role of mother’s
expression: potential implications for the sexual dimorphism of the genes and environment in postpartum depression. Proc Natl Acad
stress response and immune/inflammatory reaction. J Clin Invest. Sci U S A. 2011; 108(20): 8189–8193.
1993; 92(4): 1896–1902. 97. El-Ibiary SY, Hamilton SP, Abel R, Erdman CA, Robertson PA,
77. Jolley SN, Elmore S, Barnard KE, Carr DB. Dysregulation of the Finley PR. A pilot study evaluating genetic and environmental
hypothalamic-pituitary-adrenal axis in postpartum depression. Biol factors for postpartum depression. Innov Clin Neurosci. 2013;
Res Nurs. 2007; 8(3): 210–222. 10(9–10): 15–22.
78. Handley SL, Dunn TL, Waldron G, Baker JM. Tryptophan, cortisol 98. Costas J, Gratacòs M, Escaramís G, et al. Association study of 44
and puerperal mood. Br J Psychiatry. 1980; 136(5): 498–508. candidate genes with depressive and anxiety symptoms in post-
79. Corwin EJ, Pajer K. The psychoneuroimmunology of postpartum partum women. J Psychiatr Res. 2010; 44(11): 717–724.
depression. J Womens Health (Larchmt). 2008; 17(9): 1529–1534. 99. Pinsonneault JK, Sullivan D, Sadee W, Soares CN, Hampson E,
80. Dowlati Y, Herrmann N, Swardfager W, et al. A meta-analysis of Steiner M. Association study of the estrogen receptor gene ESR1
cytokines in major depression. Biol Psychiatry. 2010; 67(5): with postpartum depression—a pilot study. Arch Womens Ment
446–457. Health. 2013; 16(6): 499–509.
58 C. E. SCHILLER ET AL.

100. Pinheiro RT, Coelho FM, Silva RA, et al. Association of a serotonin 119. Dennerstein L, Spencer-Gardner C, Gotts G, Brown JB, Smith MA,
transporter gene polymorphism (5-HTTLPR) and stressful life Burrows GD. Progesterone and the premenstrual syndrome: a
events with postpartum depressive symptoms: a population- double blind crossover trial. Br Med J (Clin Res Ed). 1985;
based study. J Psychosom Obstet Gynaecol. 2013; 34(1): 29–33. 290(6482): 1617–1621.
101. Mahon PB, Payne JL, MacKinnon DF, et al. Genome-wide linkage 120. Frye CA, Walf AA. Hippocampal 3α,5α-THP may alter depressive
and follow-up association study of postpartum mood symptoms. Am behavior of pregnant and lactating rats. Pharmacol Biochem
J Psychiatry. 2009; 166(11): 1229–1237. Behav. 2004; 78(3): 531–540.
102. Alvim-Soares AM, Miranda DM, Campos SB, Figueira P, Correa H, 121. Chatzicharalampous C, Rizos D, Pliatsika P, et al. Reproductive
Romano-Silva MA. HMNC1 gene polymorphism associated hormones and postpartum mood disturbances in Greek women.
with postpartum depression. Rev Bras Psiquiatr. 2014; 36(1): Gynecol Endocrinol. 2010; 27(8): 543–550.
96–97. 122. Silverman ME, Loudon H, Safier M, et al. Neural dysfunction in
103. Mehta D, Newport DJ, Frishman G, et al. Early predictive postpartum depression: an fMRI pilot study. CNS Spectr. 2007;
biomarkers for postpartum depression point to a role for estrogen 12(11): 853–862.
receptor signaling. Psychol Med. 2014; 44(11): 2309–2322. 123. Moses-Kolko EL, Perlman SB, Wisner KL, James J, Saul AT,
104. Guintivano J, Arad M, Gould TD, Payne JL, Kaminsky ZA. Phillips ML. Abnormally reduced dorsomedial prefrontal cortical
Antenatal prediction of postpartum depression with blood DNA activity and effective connectivity with amygdala in response to
methylation biomarkers. Mol Psychiatry. 2014; 19(5): 560–567. negative emotional faces in postpartum depression. Am J
105. Green AD, Barr AM, Galea LAM. Role of estradiol withdrawal in Psychiatry. 2010; 167(11): 1373–1380.
“anhedonic” sucrose consumption: a model of postpartum 124. Moses-Kolko EL, Fraser D, Wisner KL, et al. Rapid habituation of
depression. Physiol Behav. 2009; 97(2): 259–265. ventral striatal response to reward receipt in postpartum
106. Bekku N, Yoshimura H. Animal model of menopausal depressive- depression. Biol Psychiatry. 2011; 70(4): 395–399.
like state in female mice: prolongation of immobility time in the 125. Goodyer IM, Herbert J, Altham PM, Pearson J, Secher SM, Shiers
forced swimming test following ovariectomy. Psychopharmacology HM. Adrenal secretion during major depression in 8- to 16-year-
(Berl). 2005; 183(3): 300–307. olds, I. Altered diurnal rhythms in salivary cortisol and
107. Bernardi M, Vergoni AV, Sandrini M, Tagliavini S, Bertolini A. dehydroepiandrosterone (DHEA) at presentation. Psychol Med.
Influence of ovariectomy, estradiol and progesterone on the 1996; 26(2): 245–256.
behavior of mice in an experimental model of depression. Physiol 126. Yaffe K, Ettinger B, Pressman A, et al. Neuropsychiatric function
Behav. 1989; 45(5): 1067–1068. and dehydroepiandrosterone sulfate in elderly women: a
108. Estrada-Camarena E, Fernandez-Guasti A, Lopez-Rubalcava C. prospective study. Biol Psychiatry. 1998; 43(9): 694–700.
Antidepressant-like effect of different estrogenic compounds in the 127. Heuser I, Deuschle M, Luppa P, Schweiger U, Standhardt H,
forced swimming test. Neuropsychopharmacology. 2003; 28(5): Weber B. Increased diurnal plasma concentrations of
830–838. dehydroepiandrosterone in depressed patients. J Clin Endocrinol
109. Walf AA, Rhodes ME, Frye CA. Antidepressant effects of ERbeta- Metab. 1998; 83(9): 3130–3133.
selective estrogen receptor modulators in the forced swim test. 128. Michael A, Jenaway A, Paykel ES, Herbert J. Altered salivary
Pharmacol Biochem Behav. 2004; 78(3): 523–529. dehydroepiandrosterone levels in major depression in adults. Biol
110. Walf AA, Frye CA. A review and update of mechanisms of estrogen Psychiatry. 2000; 48(10): 989–995.
in the hippocampus and amygdala for anxiety and depression 129. Schmidt PJ, Murphy JH, Haq N, Danaceau MA, St Clair L. Basal
behavior. Neuropsychopharmacology. 2006; 31(6): 1097–1111. plasma hormone levels in depressed perimenopausal women.
111. Maayan R, Strous RD, Abou-Kaoud M, Weizman A. The effect of Psychoneuroendocrinology. 2002; 27(8): 907–920.
17beta estradiol withdrawal on the level of brain and peripheral 130. Wolkowitz OM, Reus VI, Keebler A, et al. Double-blind treatment
neurosteroids in ovarectomized rats. Neurosci Lett. 2005; 384(1–2): of major depression with dehydroepiandrosterone. Am J
156–161. Psychiatry. 1999; 156(4): 646–649.
112. Bossé R, Rivest R, Di Paolo T. Ovariectomy and estradiol treatment 131. Schmidt P, Daly RC, Bloch M, et al. Dehydroepiandrosterone
affect the dopamine transporter and its gene expression in the monotherapy in midlife-onset major and minor depression. Arch
rat brain. Brain Res Mol Brain Res. 1997; 46(1–2): 343–346. Gen Psychiatry. 2005; 62(2): 154–162.
113. Di Paolo T, Poyet P, Labrie F. Effect of prolactin and estradiol on 132. Majewska MD, Harrison NL, Schwartz RD, Barker JL, Paul SM.
rat striatal dopamine receptors. Life Sci. 1982; 31(25): 2921–2929. Steroid hormone metabolites are barbiturate-like modulators of
114. Di Paolo T, Poyet P, Labrie F. Prolactin and estradiol increase the GABA receptor. Science. 1986; 232(4753): 1004–1007.
striatal dopamine receptor density in intact, castrated and 133. Bitran D, Hilvers RJ, Kellogg CK. Anxiolytic effects of 3α-hydroxy-
hypophysectomized rats. Prog Neuropsychopharmacol Biol 5α[β]-pregnan-20-one: endogenous metabolites of progesterone
Psychiatry. 1982; 6(4–6): 377–382. that are active at the GABAA receptor. Brain Res. 1991; 561(1):
115. Byrnes EM, Byrnes JJ, Bridges RS. Increased sensitivity of 157–161.
dopamine systems following reproductive experience in rats. 134. Wieland S, Lan NC, Mirasedeghi S, Gee KW. Anxiolytic activity of
Pharmacol Biochem Behav. 2001; 68(3): 481–489. the progesterone metabolite 5α-pregnan-3α-ol-20-one. Brain Res.
116. Wenzel A, Haugen EN, Jackson LC, Brendle JR. Anxiety symptoms 1991; 565(2): 263–268.
and disorders at eight weeks postpartum. J Anxiety Disord. 2005; 135. Bitran D, Purdy RH, Kellog CK. Anxiolytic effect of progesterone is
19(3): 295–311. associated with increases in cortical alloprenanolone and GABAA
117. Beckley EH, Finn DA. Inhibition of progesterone metabolism receptor function. Pharmacol Biochem Behav. 1993; 45(2):
mimics the effect of progesterone withdrawal on forced swim test 423–428.
immobility. Pharmacol Biochem Behav. 2007; 87(4): 412–419. 136. Smith SS, Gong QH, Hsu FC, Markowitz RS, ffrench-Mullen JM,
118. Smith SS, Gong QH, Li X, et al. Withdrawal from 3α-OH-5α- Li X. GABA(A) receptor alpha4 subunit suppression prevents
pregnan-20-one using a pseudopregnancy model alters the kinetics withdrawal properties of an endogenous steroid. Nature. 1998;
of hippocampal GABAA-gated current and increases the GABAA 392(6679): 926–930.
receptor α4 subunit in association with increased anxiety. 137. Uzunova V, Sheline Y, Davis JM, et al. Increase in the cerebrospinal
J Neurosci. 1998; 18(14): 5275–5284. fluid content of neurosteroids in patients with unipolar major
REPRODUCTIVE HORMONES AND POSTPARTUM DEPRESSION 59

depression who are receiving fluoxetine or fluvoxamine. Proc Natl 147. Djebaili M, Guo Q, Pettus EH, Hoffman SW, Stein DG. The
Acad Sci U S A. 1998; 95(6): 3239–3244. neurosteroids progesterone and allopregnanolone reduce cell
138. Romeo E, Ströhle A, Spalletta G, et al. Effects of antidepressant death, gliosis, and functional deficits after traumatic brain injury
treatment on neuroactive steroids in major depression. Am J in rats. J Neurotrauma. 2005; 22(1): 106–118.
Psychiatry. 1998; 155(7): 910–913. 148. Sayeed I, Parvez S, Wali B, Siemen D, Stein DG. Direct inhibition of
139. Ströhle A, Romeo E, Hermann B, et al. Concentrations of the mitochondrial permeability transition pore: a possible
3α-reduced neuroactive steroids and their precursors in plasma of mechanism for better neuroprotective effects of allopregnanolone
patients with major depression and after clinical recovery. Biol over progesterone. Brain Res. 2009; 1263: 165–173.
Psychiatry. 1999; 45(3): 274–277. 149. He J, Evans C-O, Hoffman SW, Oyesiku NM, Stein DG.
140. Schüle C, Romeo E, Uzunov DP, et al. Influence of mirtazapine on Progesterone and allopregnanolone reduce inflammatory
plasma concentrations of neuroactive steroids in major depression cytokines after traumatic brain injury. Exp Neurol. 2004; 189(2):
and on 3α-hydroxysteroid dehydrogenase activity. Mol Psychiatry. 404–412.
2005; 11(3): 261–272. 150. Bixo M, Andersson A, Winblad B, Purdy RH, Bäckström T.
141. Eser D, Schüle C, Baghai TC, Romeo E, Rupprecht R. Neuroactive Progesterone, 5α-pregnane-3,20-dione and 3α-hydroxy-5α-
steroids in depression and anxiety disorders: clinical studies. pregnane-20-one in specific regions of the human female brain in
Neuroendocrinology. 2006; 84(4): 244–254. different endocrine states. Brain Res. 1997; 764(1–2): 173–178.
142. Schüle C, Baghai TC, di Michele F, et al. Effects of combination 151. Akwa Y, Purdy RH, Koob GF, Britton KT. The amygdala mediates
treatment with mood stabilizers and mirtazapine on plasma the anxiolytic-like effect of the neurosteroid allopregnanolone in
concentrations of neuroactive steroids in depressed patients. rat. Behav Brain Res. 1999; 106(1–2): 119–125.
Psychoneuroendocrinology. 2007; 32(6): 669–680. 152. Epperson CN, Gueorguieva R, Czarkowski KA, et al. Preliminary
143. Patchev VK, Shoaib M, Holsboer F, Almeida OFX. The evidence of reduced occipital GABA concentrations in puerperal
neurosteroid tetrahydroprogesterone counteracts corticotropin- women: a 1H-MRS study. Psychopharmacology (Berl). 2006;
releasing hormone-induced anxiety and alters the release and gene 186(3): 425–433.
expression of corticotropin-releasing hormone in the rat 153. Schiller CE, Schmidt PJ, Rubinow DR. Allopregnanolone as a
hypothalamus. Neuroscience. 1994; 62(1): 265–271. mediator of affective switching in reproductive mood disorders.
144. Patchev VK, Hassan AHS, Holsboer F, Almeida OFX. The Psychopharmacology (Berl). 2014; 231(17): 3557–3567.
neurosteroid tetrahydroprogesterone attenuates the endocrine 154. Tarantino LM, Sullivan PF, Meltzer-Brody S. Using animal models
response to stress and exerts glucocorticoid-like effects on to disentangle the role of genetic, epigenetic, and environmental
vasopressin gene transcription in the rat hypothalamus. influences on behavioral outcomes associated with maternal
Neuropsychopharmacology. 1996; 15(6): 533–540. anxiety and depression. Front Psychiatry. 2011; 2: 44.
145. Barbaccia ML, Roscetti G, Trabucchi M, et al. The effects of 155. Maguire J, Mody I. GABAAR plasticity during pregnancy: relevance
inhibitors of GABAergic transmission and stress on brain and to postpartum depression. Neuron. 2008; 59(2): 207–213.
plasma allopregnanolone concentrations. Br J Pharmacol. 1997; 156. Maguire JL, Stell BM, Rafizadeh M, Mody I. Ovarian cycle-linked
120(8): 1582–1588. changes in GABA(A) receptors mediating tonic inhibition alter seizure
146. Kehoe P, Mallinson K, McCormick CM, Frye CA. Central susceptibility and anxiety. Nat Neurosci. 2005; 8(6): 797–804.
allopregnanolone is increased in rat pups in response to repeated, 157. Maguire J, Ferando I, Simonsen C, Mody I. Excitability changes
short episodes of neonatal isolation. Brain Res Dev Brain Res. related to GABAA receptor plasticity during pregnancy. J Neurosci.
2000; 124(1–2): 133–136. 2009; 29(30): 9592–9601.

You might also like