Open access peer-reviewed chapter

Influences of Maternal Vulnerability and Antidepressant Treatment during Pregnancy on the Developing Offspring

Written By

Laura Staal and Jocelien DA Olivier

Submitted: 03 October 2018 Reviewed: 21 December 2018 Published: 14 February 2019

DOI: 10.5772/intechopen.83761

From the Edited Volume

Antidepressants - Preclinical, Clinical and Translational Aspects

Edited by Olivier Berend

Chapter metrics overview

1,027 Chapter Downloads

View Full Metrics

Abstract

Maternal vulnerability to adversity has long-term impact on the developing child. About 20% of the pregnant women suffer from affective disorders. Fetal exposure to maternal adversity may lead to detrimental consequences later in life. Maternal affective disorders are increasingly treated with antidepressants, especially selective serotonin reuptake inhibitors (SSRIs). However, the long-term consequences for the offspring after exposure to this medication are unclear. The interplay between maternal adversity and SSRI treatment has been under investigation and here we discuss how maternal adversity and SSRIs are able to shape offspring development. Specifically, we will discuss animal models addressing behavioral outcomes to understand how the prenatal environment influences the health of the developing child across the life span.

Keywords

  • maternal vulnerability
  • maternal depression
  • selective serotonin reuptake inhibitors
  • antidepressants
  • pregnancy
  • neurodevelopment

1. Introduction

Although pregnancy is often portrayed as a time of great joy, that is not the reality for all women. Depressive symptoms during pregnancy are not uncommon; in fact, 20% of women experience some depressive symptoms during any time of their pregnancy [1]. The number of women who suffer from major depression during pregnancy is estimated to be 4–8% [2, 3]. According to the DSM-5, this disorder is characterized by a depressed mood or loss of interest or pleasure in daily activities for more than 2 weeks. Depression is accompanied by impaired social, occupational, and educational functioning. Untreated antenatal depression, that is to say a depressive episode during pregnancy, may have a tremendous effect on the developing child [4].

Maternal vulnerabilities during pregnancy, such as depression, anxiety, or high stress levels due to other reasons, are associated with increased and continued activation of the hypothalamic-pituitary-adrenal (HPA) axis. The continued activation of the HPA axis in depressed patients causes an elevated stress response and increased cortisol levels [5]. About 40% of the cortisol passes through the placenta [6]. Consequently, increased cortisol levels are found in the urine and saliva of the infants of depressed mothers [7].

Fetal exposure to increased maternal stress levels impacts the developing child. For example, high levels of maternal cortisol are associated with reduced neurological development [8] and altered cortisol responses of the unborn child to a stressor [9]. Furthermore, maternal vulnerabilities such as anxiety, depression, and elevated stress levels are associated with the increased fearful temperament and negative behavioral reactivity to novelty in infants [9, 10], and delayed cognitive and neuromotor development [11, 12] that persists into adolescence [13]. In addition, antenatal depression has also been linked with disturbed sleep patterns in infants [14] and in 18 and 30 months old children [15]. Furthermore, antenatal depression has been linked to reduced fetal growth [16, 17] and altered cardiovascular responses to stress [18].

Several studies have looked into the effects of maternal vulnerability on the behavioral development of the child. For example, maternal anxiety, but not antenatal depression is linked to a difficult child temperament at the age of 4–6 months [19], an increase in behavioral and emotional problems at the age of 4 [20] and more internalizing behavior at the age of 8 [21]. Moreover, antenatal depression is associated with delayed development in 18-month-olds [22], increased externalizing behaviors and a slight decrease in IQ in 8-year-old children [21], and violent behavior during adolescence [23]. On the long-term prenatal exposure to maternal depression is associated with a higher chance of developing depression during adolescence [24, 25] and adulthood [26], or risk of developing other psychopathologies [27]. Maternal anxiety as well as maternal depression during pregnancy is correlated with child attention problems at the age of 3 and 4 [28]. Moreover, an increase in reporting symptoms of antenatal depression and anxiety positively correlated with an increase in internalizing behaviors in 4-year-olds [29]. So anxiety and depression appear to have similar but, at the same time, different effects on offspring development; however, it is difficult to discern between maternal anxiety and depression, due to common comorbidity between these mental health conditions [30].

Overall, maternal vulnerability, such as depression and anxiety, during pregnancy can negatively influence the unborn child on both physiological and behavioral levels. However, it remains difficult to discern the direct effects of antenatal depression (an aversive postnatal environment due to a depressed mother), and genetic predisposition to vulnerability on fetal and infant development. In addition, an increasing percentage of women suffering from depression and/or anxiety are treated with antidepressants, which on itself might have tremendous effects on the developing child.

Advertisement

2. Maternal SSRI treatment and offspring development

Pharmacological treatment of antenatal depression and/or anxiety is sometimes unavoidable. The treatment with antidepressants may relieve the symptoms of the depression of the mother and could help in reducing the impact on the unborn child. Nowadays, a considerable number of women are treated with antidepressants during pregnancy. In Europe, this concerns 2–3% of the pregnant women [16, 31], while in the U.S., the occurrence is as high as up to 13% [32, 33]. The most prescribed antidepressants are selective serotonin reuptake inhibitors (SSRIs), because of their good efficacy, few side effects, and therapeutic safety [34]. These drugs work by blocking the serotonin transporter and hereby preventing the reabsorption of the neurotransmitter serotonin into the presynaptic nerve cell. Subsequently, extracellular serotonin levels in the synaptic cleft are increased and more serotonin is available to bind the postsynaptic receptors. Although, SSRIs are considered safe for antenatal use [35], it has been reported that the use of SSRIs during pregnancy may negatively influence the development of the unborn child. SSRIs can cross the placenta and are found in the amniotic fluid [36, 37], affecting therefore not only the mother but also the developing child. This is of extra concern as serotonin plays a key role in embryonal development. During the development of the fetal brain, serotonin acts as a neurotrophic factor, regulating cell division, differentiation, migration, growth cone elongation, dendritic pruning, myelination, and synaptogenesis [38]. In fact, serotonin receptors and serotonergic metabolic enzymes are expressed before serotonin-producing neurons are present in the brain [39]. Thus, changes in the serotonin levels during neurodevelopment, for instance, by the administration of SSRIs during pregnancy, potentially affect a number of processes in the offspring.

Indeed, literature shows a number of side effects in the offspring due to prenatal SSRI exposure. First of all, SSRI exposure during pregnancy has been associated with attenuated basal cortisol levels in neonates [40, 41], and differential cortisol levels in 3-month-old infants in response to a stressor [42]. Also, the neonatal heart rate response to an acute noxious event is attenuated [43]. Furthermore, several behavioral changes have been reported, such as increased internalizing behaviors, such as depression, anxiety, and social withdrawal during childhood [44, 45], increased externalizing behaviors in 4-year-old children [46], and disrupted sleep patterns in newborn [47]. In addition, SSRIs reduce utero-placental blood flow, a mechanism thought to be involved with hypertension in preeclampsia and gestational diabetes [48, 49].

Recently, there has been much interest in the link between antenatal SSRI treatment and the development of autism spectrum disorders (ASDs) in the child. ASD is a neurodevelopmental condition characterized by difficulties in social communication and unusually restricted, repetitive behavior and interests. The available literature shows an association between the prenatal use of SSRIs and the increased risk of ASDs in the child [50, 51, 52, 53, 54]. It is theorized that this is facilitated by an increase in serotonergic activity during brain development [55]. Several studies found abnormal placental histology to be associated with autism diagnosis [56, 57]. Moreover, autistic patients have elevated blood platelet serotonin levels [58, 59]. Taken together, these results imply the serotonergic influences on maternal-fetal interaction, although the exact mechanisms remain elusive.

A possible route for passing adverse intra-uterine effects to the fetus is via epigenetic regulation. For example, increased maternal depressive mood during pregnancy is associated with reduced methylation of the promotor of the gene coding the serotonin transporter (SERT) in both mothers and newborns [60]. These results suggest increased SERT mRNA levels and subsequently modified serotonin levels, contributing to increased vulnerability later in life [61]. St-Pierre and colleagues therefore conclude that all parameters that can alter serotonin homeostasis during early development could lead to structural and functional changes in fetal development and brain circuits [62], which could subsequently result in a predisposition to psychopathology in adulthood.

Thus, several studies have shown an increased risk for developing the child both during antenatal depression and after prenatal SSRI exposure. However, it is difficult to discern between the effects of the SSRIs and the effects of the depression itself, as healthy mothers do not administer antidepressants. For example, meta-analyses show that the risk found for ASD in the offspring after prenatal SSRI exposure is decreased after correcting for maternal mental illness [63]. Thus, the effects of SSRIs mentioned could be solely due to the administration of the SSRIs, or alternatively, the SSRIs are only partially effective and therefore do not eliminate all the adverse effects of the depression, thereby adding up to the adverse effects of antenatal depression [4].

Advertisement

3. Preclinical studies: perinatal SSRI exposure

In humans, it is difficult, if not impossible, to discern the effects of the SSRI and the depression itself on fetal development. It is not ethical to study the effects of SSRIs in healthy pregnant women. In addition, it is impossible to study gene expression and epigenetic changes in the fetal brain as a result of prenatal SSRI exposure. Due to these limitations of human research, researchers often use animal models, specifically rodents, to get a more profound insight into the mechanisms underlying the observations seen in human studies.

It should be noted that the timing of brain development is different in humans compared to rodents. A rodent brain at postnatal day 7–10 is considered to be the rough equivalent of a newborn human infant [64]. Thus, to mimic SSRI exposure during the entire pregnancy in humans, rodents should be exposed both pre- and postnatally. In addition, it is known that SSRIs are also found in breast milk [65], again underlining the need to research both the effects of pre- and postnatal SSRI exposure. For exposure to SSRIs around these time points, we will use the term perinatal SSRI exposure; when timing of the SSRI exposure is of particular importance, we will distinguish between pre- and postnatal exposure to SSRIs.

3.1 Social behavior

As serotonin is a key regulator of social responses and prenatal SSRI exposure is being linked to ASDs, which are characterized by impaired social behavior [54], this behavioral parameter is often addressed in researching the effects of prenatal SSRI exposure in animals.

Social play at juvenile age is an essential behavior in rodents for the development of the necessary social, cognitive, emotional, and physical skills [66]. SSRIs are well described in literature as reducing social play behavior in young rats when prenatally [67] or postnatally [68, 69, 70, 71] administered. These effects of SSRIs seem sex-mediated, as males are more affected than females [70], or females are not even affected at all [71]. This is interesting because this is analogous to the situation, where men are 3–4 times more likely to get diagnosed with ASDs compared to women [72]. In spite of that, not all studies found similar results. In a recent study [73], social play behavior was unaltered after prenatal SSRI exposure. However, in the latter study, social play was assessed with a familiar play partner (littermate), while other studies use a novel conspecific.

Findings on the effects of prenatal SSRI exposure on social behavior at adult age are more conflicting. Olivier and colleagues [67] found reduced social exploration in adult male rats after prenatal SSRI exposure. While another study found that 4 days of postnatal treatment with SSRIs led to increased sniffing, contact with, and total social interaction with a novel conspecific in males [74]. On top of that, other studies found no effect of prenatal SSRI exposure on social exploration in both males and females [75, 76]. In general, studies on the exposure of SSRIs during early development on adult social interaction are unconvincing. Studies on social motivation on the other hand, measured as the preference of a rodent to spend time with a novel conspecific over interaction with an object, appear to be more in line. Decreased social motivation is found in both males and females, when postnatally exposed to SSRIs [69, 70, 71, 77]. On the other hand, prenatal exposure to SSRIs led to an increase in motivation to interact with a conspecific in mice [75]. Thus, while literature on the effects of perinatal SSRI exposure on social behavior during adult is still limited, both timing of the exposure and sex are important factors in the subsequent social development.

Another form of social interaction under the influence of the neurotransmitter serotonin is aggression [78]. Indeed, during both childhood and adulthood, SSRIs are successfully used to reduce aggressive and violent behavior in certain mental disorders [79, 80]. Perinatal exposure to SSRIs, on the other hand, leads to increased externalizing behavior, such as aggression, in children [46]. In rodents, the effects of perinatal SSRI exposure on aggressive behavior are conflicting. Several studies show an increase in male, but not female aggressive behavior [73, 75, 76, 81], while other studies show reduced aggressive behavior in male rodents perinatally exposed to SSRIs [82, 83].

Serotonin is known to be involved in the regulation of maternal care [84]. Thus, perinatal SSRI exposure can be expected to alter maternal caregiving behavior of both the SSRI-treated mother and her female offspring, when they are mothers later in life. Typical maternal behaviors include nest building, gathering the young into the nest, maternal licking, and nursing the pups. Studies on the effect of direct SSRI exposure found an increase in these behaviors [85, 86], or no effect at all [87]. So far, only one study has been performed on the effects of prenatal exposure on maternal care later in life, and interestingly here, they found a reduction in maternal caregiving behaviors [75]. This suggests that direct changes in serotonin levels, such as an increase in extracellular serotonin levels during SSRI treatment and changes in serotonin levels during development differentially alter the quality of maternal care.

Another role for serotonin is its signaling in sexual development, on both the brain and at a behavioral level [88]. In addition, chronic SSRI treatment may result in sexual dysfunction [89]. Not much is known on the effect of perinatal SSRI exposure on the sexual behavior of these children later in life; however, several studies have been performed in rodents. The effect of perinatal SSRI exposure depends on the timing of treatment. When postnatally administered, male sexual behavior later in life is reduced [70, 90, 91, 92, 93]. In contrast when the SSRI is prenatally, or both prenatally and postnatally, administered, there is no effect on sexual behavior of male rodents [94, 95]. Interestingly, there appears to be an opposite effect in female offspring, where postnatal SSRI exposure leads to an increase in sexual behaviors [75, 96]. Thus, apart from the effect of timing, sex of the offspring also differentially alters the effect of perinatal SSRI exposure on reproductive behavior.

3.2 Affective behavior

It has long been established that serotonin is involved in affective disorders [97]. Affective disorders, also called mood disorders, include psychiatric disorders such as major depressive disorder and anxiety disorders.

A large body of preclinical research has shown the relationship between perinatal SSRI exposure and anxiety. Although there are some studies finding no effects, several studies did find an increase in anxiety-like behavior and/or less explorative behavior in an open field test, when rodents are perinatally exposed to SSRIs [67, 83, 98, 99, 100, 101, 102, 103, 104]. An increase in anxiety-like behavior in the elevated plus maze and the novelty-suppressed feeding test are also found [67, 98, 99, 105]. Nevertheless, there are also studies that did not find effects at all [87, 91, 106, 107, 108, 109, 110, 111], and two studies even found a decrease in behaviors related to anxiety [92, 112]. Even though results differ among studies, these differences are not clearly linked to sex of the offspring or timing of the SSRI exposure. So even though there appears to be a clear link between perinatal SSRI exposure and anxiety later in life, to get a more profound insight into the mechanisms behind this and the role of timing and sex, more researches have to be done.

It is difficult to determine if a rodent is depressed, moreover, to determine if it is even possible for rodents to experience depression. However, rodents can show behavior characteristics of the behaviors, and humans show during episodes of major depression. Such behaviors encompass despair and anhedonia [113]. To measure behavioral despair in rodents, the forced swim test is usually performed [114]. In this test, the animal is placed in an unescapable container filled with water, forcing the rodent to swim. After making efforts to escape the animal may eventually stop his efforts and become immobile. The amount of time spent immobile is used as a measure of helplessness and behavioral despair. As reviewed [115], many, but not all, studies performing the forced swim test after perinatal SSRI exposure found an increase in immobility [83, 99, 100, 105, 106, 109, 110, 116, 117, 118, 119, 120, 121]. Three studies did not find an effect [67, 87, 111], and three studies even found a decrease in immobility [103, 112, 122]. The reviewers propose that these differences may be due to strain effects, some strains could be more susceptible to early life SSRI exposure, while others are resistant. In addition, they point out that the effect of perinatal SSRI exposure is greater, when the animals are exposed during early postnatal period rather than the prenatal period.

Anhedonia is another behavior often assessed as a measure for depression. Anhedonia is the inability or lack of motivation to experience pleasure from rewarding activities and is measured in rodents with the sucrose preference test [123]. In this test, two drinking bottles are placed in the rodent’s home cage. One is filled with water and the other with a sucrose solution. Preference for the sucrose solution is considered as the typical hedonic behavior, and lack of bias toward the sucrose water is characterized as a sign of anhedonia. It appears that only postnatal SSRI exposure increases anhedonia later in life [124], as opposed to prenatal exposure [67, 125]. This once more emphasizes that the moment of exposure is an important factor in assessing the effects of perinatal SSRI exposure.

Thus, not only sex of the offspring, but also the timing of the SSRI exposure appears to play an important role in behavioral development. However, all aforementioned studies are performed in offspring from healthy mothers. In practice, pregnant women are usually treated with SSRIs when they are suffering from anxiety and/or depression. It is likely that both maternal factors and the treatment with SSRIs affect serotonin functioning in the embryo and infant. The interplay of these two factors might shape the development of the fetus in a different way than antenatal depression or prenatal SSRI exposure on its own. Thus, to make a more valid translational step to the human situation, animal models of maternal vulnerability have to be used.

Advertisement

4. Preclinical studies: maternal vulnerability and perinatal SSRI exposure

To induce maternal vulnerability in healthy rodents, researchers often use the early life stress model (ELS). Maternal separation is one of the manipulations often used to create ELS; in this procedure, the offspring is taken away from the mother for few hours during the day, and this happens daily during a period of few early postnatal weeks. This procedure leads to a long-term and intergenerational increase in anxiety and depressive-like behaviors [126, 127, 128, 129, 130, 131]. Female offspring exposed to ELS and showing an increase in anxiety and depressive-live behaviors can be used as a model of maternal vulnerability later in life. Little is known about the interaction of such maternal vulnerability and treatment with SSRIs during pregnancy.

4.1 Social behavior

So far, only two studies have been looking into the combined effects of maternal vulnerability and perinatal SSRI exposure on social play behavior and social interaction of the offspring later in life. In 2017, Gemmel and colleagues [73] showed that the reduction in social play behavior in juveniles due to maternal vulnerability is prevented by perinatal SSRI treatment regardless of the sex of the offspring. This suggests a rescuing effect of SSRIs on social behavior in offspring of stressed mothers. However, social aggressive play was increased in adolescent offspring exposed to perinatal fluoxetine and maternal vulnerability in both sexes. In addition, time grooming a novel conspecific was decreased in males only. In a later study, Gemmel and colleagues [132] did not find such an interaction effect on social behavior in adult offspring. Even though, maternal vulnerability itself decreased social investigation in adult males while perinatal SSRI exposure increased social investigation in adult females and increased social play in adult males. Thus, normalization, by SSRIs, of altered social play and social interaction, due to maternal vulnerability might only be short-lived as it does not persist into adulthood. With regard to aggression, however, long-term protective effects of SSRIs are found [81]. In this study, aggressive behavior was decreased as a result of maternal vulnerability, which was normalized when perinatal SSRI exposure was included in the treatment.

One study has looked into the combined effect of SSRI exposure and maternal vulnerability on offspring sexual development [133]. Perinatal SSRI exposure reduced sexual behavior in male offspring, while interestingly maternal vulnerability alone or the combination of maternal vulnerability and perinatal SSRI exposure did not have any effect on the development of sexual behavior.

So, even though SSRI treatment of vulnerable mothers appears to have protective effects on offspring social development, these findings are not consistent over all types of behavior and differ with the moment of assessment of the offspring.

4.2 Affective behavior

Affective behaviors of the offspring such as anxiety and depression-like behaviors have also been studied after the offspring was exposed to a combination of maternal vulnerability and perinatal SSRI exposure. One study [134] shows that the increase in anxiety due to maternal vulnerability can be reversed by the postnatal administration of SSRIs. When prenatally administered, such a rescuing effect is not found [87]; however, the effect of maternal vulnerability was limited and was only found in males in this study. Two studies assessed depressive-like behavior after perinatal exposure to both maternal vulnerability and SSRIs. Both studies found that SSRIs normalize the increase in immobility in the forced swim test due to maternal vulnerability [87, 135]. Thus so far, the effect maternal vulnerability has on anxiety and depressive-like symptoms in the offspring later in life appear to be reversed by SSRIs.

Advertisement

5. Conclusion

Thus, children from mothers who suffer from anxiety or major depression during their pregnancy are at risk of developing several psychopathologies later in life. Moreover, treatment with SSRIs during pregnancy can also lead to long-term consequences for the children. However, it is difficult to determine if these effects are due to the SSRI treatment, maternal vulnerability, or a combination of both. Preclinical research in rodents shows that perinatal SSRI exposure on itself leads to alterations in social behavior later in life. Specifically, social play in juveniles, sexual behavior, maternal care in females, and aggression in males are influenced. Affective behaviors are also influenced, and both anxiety and depressive-like behaviors are increased due to perinatal SSRI exposure. Both the moments of SSRI exposure, pre- or postnatal, and sex of the offspring appear to be important factors in the development of social and affective behaviors after perinatal SSRI treatment.

Recently, researchers have started to look into the combined effects of maternal vulnerability and perinatal SSRI exposure in preclinical studies to make a more valid translational step to the human situation. Even though only a few studies have been done so far, it seems that, at least some, developmental alterations on offspring behavior due to maternal vulnerability can be normalized by perinatal SSRI exposure. These are interesting and promising results and further investigation into the risks and benefits of SSRI use during pregnancy in appropriate animal models are necessary to help depressed women in their decision to use SSRIs during pregnancy.

Advertisement

Conflict of interest

The authors declare that there is no conflict of interest.

References

  1. 1. Ryan D, Milis L, Misri N. Depression during pregnancy. Canadian Family Physician. 2005;51:1087-1093
  2. 2. Melville JL, Gavin A, Guo Y, Fan M, Katon WJ. Depressive disorders during pregnancy: Prevalence and risk factors in a large urban sample. Obstetrics and Gynecology. 2010;116(5):1064-1070
  3. 3. Kim DR, Snell JL, Ewing GC, O’Reardon J. Neuromodulation and antenatal depression: A review. Neuropsychiatric Disease and Treatment. 2015;11:975-982
  4. 4. Olivier JDA, Åkerud H, Sundström I. Antenatal depression and antidepressants during pregnancy: Unraveling the complex interactions for the offspring. European Journal of Pharmacology. 2015;753:257-262
  5. 5. Field T, Diego M, Hernandez-reif M. Prenatal depression effects on the fetus and newborn: A review. Infant Behavior & Development. 2006;29:445-455
  6. 6. Gitau R, Cameron A, Fisk NM, Glover V. Fetal exposure to maternal cortisol. Lancet. 1998;352:707-708
  7. 7. Kaplan LA, Evans L, Monk C. Effects of mothers’ prenatal psychiatric status and postnatal caregiving on infant biobehavioral regulation: Can prenatal programming be modified? Early Human Development. 2008;84(4):249-256
  8. 8. Ellman LM, Schetter CD, Hobel CJ, Chicz-DeMet A, Glynn LM, Sandman CA. Timing of fetal exposure to stress hormones: Effects on newborn physical and neuromuscular maturation. Developmental Psychobiology. 2008;50(3):232-241
  9. 9. Davis EP, Glynn LM, Waffarn F, Sandman CA. Prenatal maternal stress programs infant stress regulation. Journal of Child Psychology and Psychiatry. 2011;52(2):119-129
  10. 10. Davis EP, Snidman N, Wadhwa PD, Glynn LM, Schetter CD, Sandman CA. Prenatal maternal anxiety and depression predict negative behavioral reactivity in infancy. Infancy. 2004;6(3):319-331
  11. 11. Davis EP, Glynn LM, Schetter CD, Hobel C, Chicz-Demet A, Sandman CA. Prenatal exposure to maternal depression and cortisol influences infant temperament. Journal of the American Academy of Child and Adolescent Psychiatry. 2007;46(6):737-746
  12. 12. Huizink AC, Robles de Medina PG, EJH M, GHA V, Buitelaar JK. Stress during pregnancy is associatedwith developmental outcome in infancy. Journal of Child Psychology and Psychiatry. 2003;44(6):810-818
  13. 13. Mennes M, Stiers P, Lagae L, Van den Bergh B. Long-term cognitive sequelae of antenatal maternal anxiety: Involvement of the orbitofrontal cortex. Neuroscience and Biobehavioral Reviews. 2006;30:1078-1086
  14. 14. Field T, Diego M, Hernandez-Reif M, Figueiredo B, Schanberg S, Kuhn C. Sleep disturbances in depressed pregnant women and their newborns. Infant Behavior & Development. 2007;30:127-133
  15. 15. O’Connor TG, Caprariello P, Blackmore ER, Gregory AM, Glover V, Fleming P. Prenatal mood disturbance predicts sleep problems in infancy and toddlerhood. Early Human Development. 2007;83(7):451-458
  16. 16. El Marroun H, Jaddoe VWV, Hudziak JJ, Roza SJ, Steegers EAP, Hofman A, et al. Maternal use of selective serotonin reuptake inhibitors, fetal growth, and risk of adverse birth outcomes. Archives of General Psychiatry. 2012;69(7):706-714
  17. 17. Henrichs J, Schenk JJ, Roza SJ, van den Berg MP, Schmidt HG, Steegers EAP, et al. Maternal psychological distress and fetal growth trajectories: The generation R study. Psychological Medicine. 2010;40:633-643
  18. 18. Fan F, Zou Y, Tian H, Zhang Y, Zhang J, Ma X, et al. Effects of maternal anxiety and depression during pregnancy in Chinese women on children’s heart rate and blood pressure response to stress. Journal of Human Hypertension. 2016;30:171-176
  19. 19. Austin MP, Hadzi-Pavlovic D, Leader L, Saint K, Parker G. Maternal trait anxiety, depression and life event stress in pregnancy: Relationships with infant temperament. Early Human Development. 2005;81:183-190
  20. 20. O’Connor TG, Heron J, Glover V. Antenatal anxiety predicts child behavioral/emotional problems independently of postnatal depression. Journal of the American Academy of Child and Adolescent Psychiatry. 2002;41(12):1470-1477
  21. 21. Barker ED, Jaffee SR, Uher R, Maughan B. The contribution of prenatal and postnatal maternal anxiety and depression to child maladjustment. Depression and Anxiety. 2011;28(8):696-702
  22. 22. Deave T, Heron J, Evans J, Emond A. The impact of maternal depression in pregnancy on early child development. BJOG : An International Journal of Obstetrics and Gynaecology. 2008;115:1043-1051
  23. 23. Hay DF, Pawlby S, Waters CS, Perra O, Sharp D. Mothers’ antenatal depression and their children’s antisocial outcomes. Child Development. 2010;81(1):149-165
  24. 24. Plant DT, Pariante CM, Sharp D, Pawlby S. Maternal depression during pregnancy and offspring depression in adulthood: Role of child maltreatment. The British Journal of Psychiatry. 2015;207:213-220
  25. 25. Pawlby S, Hay DF, Waters CS, Sharp D, O’Keane V. Antenatal depression predicts depression in adolescent offspring: Prospective longitudinal community-based study. Journal of Affective Disorders. 2009;113(3):236-243
  26. 26. 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. 2013;70(12):1312-1319
  27. 27. Pawlby S, Hay D, Sharp D, Waters CS, Pariante CM. Antenatal depression and offspring psychopathology: The influence of childhood maltreatment. The British Journal of Psychiatry. 2011;199:106-112
  28. 28. Van Batenburg-Eddes T, Brion MJ, Henrichs J, Jaddoe VWV, Hofman A, Verhulst FC, et al. Parental depressive and anxiety symptoms during pregnancy and attention problems in children: A cross-cohort consistency study. Journal of Child Psychology and Psychiatry. 2012;54(5):591-600
  29. 29. 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;163:1026-1032
  30. 30. Glover V. Maternal depression, anxiety and stress during pregnancy and child outcome; what needs to be done. Best Practice & Research. Clinical Obstetrics & Gynaecology. 2014;28(1):25-35
  31. 31. Kieler H, Artama M, Engeland A, Ericsson Ö, 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;344:1-9
  32. 32. Cooper WO, Willy ME, Pont SJ, Ray WA. Increasing use of antidepressants in pregnancy. American Journal of Obstetrics and Gynecology. 2007;196:1-5
  33. 33. Hayes RM, Wu DP, Shelton RC, Cooper O, Dupont WD, Mitchel E, et al. Maternal antidepressant use and adverse outcomes: A cohort study of 228,876 pregnancies. American Journal of Obstetrics and Gynecology. 2012;207(1):49
  34. 34. Barbey JT, Roose SP. SSRI safety in overdose. The Journal of Clinical Psychiatry. 1998;59:42-48
  35. 35. Gentile S. The safety of newer antidepressantsin pregnancy and breastfeeding. Drug Safety. 2005;28(2):137-152
  36. 36. Hostetter A, Stowe ZN, Strader JR, McLaughlin E, Llewellyn A. Dose of selective serotonin uptake inhibitors across pregnancy: Clinical implications. Depression and Anxiety. 2000;11(2):51-57
  37. 37. Loughhead AM, Fisher AD, Newport DJ, Ritchie JC, Owens MJ, CL DV, et al. Antidepressants in amniotic fluid: Another route of fetal exposure. The American Journal of Psychiatry. 2006;163(1):145-147
  38. 38. Gaspar P, Cases O, Maroteaux L. The developmental role of serotonin: News from mouse molecular genetics. Nature Reviews. Neuroscience. 2003;4(12):1002-1012
  39. 39. 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;472(7343):347-350
  40. 40. Brennan PA, Pargas R, Walker EF, Green P, Jeffrey Newport D, Stowe Z. Maternal depression and infant cortisol: Influences of timing, comorbidity and treatment. The Journal of Child Psychology and Psychiatry and Allied Disciplines. 2008;49(10):1099-1107
  41. 41. Pawluski JL, Brain UM, Underhill CM, Hammond GL, Oberlander TF. Prenatal SSRI exposure alters neonatal corticosteroid binding globulin, infant cortisol levels, and emerging HPA function. Psychoneuroendocrinology. 2012;37(7):1019-1028
  42. 42. Oberlander TF, Grunau R, Mayes L, Riggs W, Rurak D, Papsdorf M, et al. Hypothalamic-pituitary-adrenal (HPA) axis function in 3-month old infants with prenatal selective serotonin reuptake inhibitor (SSRI) antidepressant exposure. Early Human Development. 2008;84(10):689-697
  43. 43. Oberlander TF, Eckstein Grunau R, Fitzgerald C, Ellwood A-L, Misri S, Rurak D, et al. Prolonged prenatal psychotropic medication exposure alters neonatal acute pain response. Pediatric Research. 2002;51(4):443-453
  44. 44. Hanley GE, Brain U, Oberlander TF. Prenatal exposure to serotonin reuptake inhibitor antidepressants and childhood behavior. Pediatric Research. 2015;78:174-180
  45. 45. Oberlander TF, Papsdorf M, Brain UM, Misri S, Ross C, Grunau RE. Prenatal effects of selective serotonin reuptake inhibitor antidepressants, serotonin transporter promoter genotype. Archives of Pediatrics & Adolescent Medicine. 2010;164(5):444-451
  46. 46. 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;161:22-29
  47. 47. Zeskind PS, Stephens LE. Maternal selective serotonin reuptake inhibitor use during pregnancy and newborn neurobehavior. Pediatrics. 2004;113(2):368-375
  48. 48. Bolte AC, Van Geijn HP, Dekker GA. Pathophysiology of preeclampsia and the role of serotonin. European Journal of Obstetrics, Gynecology, and Reproductive Biology. 2001;95(1):12-21
  49. 49. Li Y, Hadden C, Singh P, Mercado CP, Murphy P, Dajani NK, et al. GDM-associated insulin deficiency hinders the dissociation of SERT from ERp44 and down-regulates placental 5-HT uptake. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(52):E5697-E5705
  50. 50. Andalib S, Emamhadi MR, Yousefzadeh-Chabok S, Shakouri SK, Høilund-Carlsen PF, Vafaee MS, et al. Maternal SSRI exposure increases the risk of autistic offspring: A meta-analysis and systematic review. European Psychiatry. 2017;45:161-166. DOI: 10.1016/j.eurpsy.2017.06.001
  51. 51. Brown HK, Hussain-Shamsy N, Lunsky Y, Dennis CLE, Vigod SN. The association between antenatal exposure to selective serotonin reuptake inhibitors and autism: A systematic review and meta-analysis. The Journal of Clinical Psychiatry. 2017;78:e48-e58
  52. 52. Kaplan YC, Keskin-Arslan E, Acar S, Sozmen K. Prenatal selective serotonin reuptake inhibitor use and the risk of autism spectrum disorder in children: A systematic review and meta-analysis. Reproductive Toxicology. 2016;66:31-43
  53. 53. Kobayashi T, Matsuyama T, Takeuchi M, Ito S. Autism spectrum disorder and prenatal exposure to selective serotonin reuptake inhibitors: A systematic review and meta-analysis. Reproductive Toxicology. 2016;65:170-178. DOI: 10.1016/j.reprotox.2016.07.016
  54. 54. Gentile S. Prenatal antidepressant exposure and the risk of autism spectrum disorders in children. Are we looking at the fall of gods? Journal of Affective Disorders. 2015;182:132-137
  55. 55. Whitaker-Azmitia PM. Behavioral and cellular consequences of increasing serotonergic activity during brain development: A role in autism? International Journal of Developmental Neuroscience. 2005;23(1):75-83
  56. 56. Anderson GM, Gutknecht L, Cohen DJ, Brailly-Tabard S, Cohen JHM, Ferrari P, et al. Serotonin transporter promoter variants in autism: Functional effects and relationship to platelet hyperserotonemia. Molecular Psychiatry. 2002;7(8):831-836
  57. 57. Walker CK, Anderson KW, Milano KM, Ye S, Tancredi DJ, Pessah IN, et al. Trophoblast inclusions are significantly increased in the placentas of childeren in families at risk for autism. Biological Psychiatry. 2013;74(3):204-211
  58. 58. McBride PA, Anderson GM, Hertzig ME, Snow ME, Thompson SM, Khait VD, et al. Effects of diagnosis, race, and puberty on platelet serotonin levels in autism and mental retardation. Journal of the American Academy of Child and Adolescent Psychiatry. 1998;37(7):767-776
  59. 59. Bijl N, Thys C, Wittevrongel C, De La Marche W, Devriendt K, Peeters H, et al. Platelet studies in autism spectrum disorder patients and first-degree relatives. Molecular Autism. 2015;6(1):1-10
  60. 60. Devlin AM, Brain U, Austin J, Oberlander TF. Prenatal exposure to maternal depressed mood and the MTHFR C677T variant affect SLC6A4 methylation in infants at birth. PLoS One. 2010;5(8):2-9
  61. 61. Philibert R, Madan A, Andersen A, Cadoret R, Packer H, Sandhu H. Serotonin transporter mRNA levels are associated with the methylation of an upstream CpG island. American Journal of Medical Genetics. 2007;144B:101-105
  62. 62. St-Pierre J, Laurent L, King S, Vaillancourt C. Effects of prenatal maternal stress on serotonin and fetal development. Placenta. 2016;48:S66-S71
  63. 63. Andrade C. Antidepressant exposure during pregnancy and risk of autism in the offspring, 1: Meta-review of meta-analyses. The Journal of Clinical Psychiatry. 2017;78(8):e48-e58
  64. 64. Semple B, Blomgren K, Gimlin K, Ferriero D, Noble-Haeusslein L. Brain development in rodents and humans: Identifying benchmarks of maturation and vulnerability to injury across species. Progress in Neurobiology. 2013:1-16
  65. 65. Heikkinen T, Ekblad U, Palo P, Laine K. Pharmacokinetics of fluoxetine and norfluoxetine in pregnancy and lactation. Clinical Pharmacology and Therapeutics. 2003;73(4):330-337
  66. 66. Vanderschuren LJ, Trezza V. What the laboratory rat has taught us about social play behavior: Role in behavioral development and neural mechanisms. Current Topics in Behavioral Neurosciences. 2014;16:189-212
  67. 67. Olivier JDA, Vallès A, Van Heesch F, Afrasiab-Middelman A, Roelofs JJPM, Jonkers M, et al. Fluoxetine administration to pregnant rats increases anxiety-related behavior in the offspring. Psychopharmacology. 2011;217(3):419-432
  68. 68. Homberg JR, Schiepers OJG, Schoffelmeer ANM, Cuppen E, Vanderschuren LJMJ. Acute and constitutive increases in central serotonin levels reduce social play behaviour in peri-adolescent rats. Psychopharmacology. 2007;195(2):175-182
  69. 69. Khatri N, Simpson KL, Lin RCS, Paul IA. Lasting neurobehavioral abnormalities in rats after neonatal activation of serotonin 1A and 1B receptors: Possible mechanisms for serotonin dysfunction in autistic spectrum disorders. Psychopharmacologia. 2014;231(6):1191-1200
  70. 70. Rodriguez-Porcel F, Green D, Khatri N, Swilley Harris S, May WL, Lin RCS, et al. Neonatal exposure of rats to antidepressants affects behavioral reactions to novelty and social interactions in a manner analogous to autistic spectrum disorders. The Anatomical Record. 2011;294(10):1726-1735
  71. 71. Simpson KL, Weaver KJ, de Villers-Sidani E, Lu JY-F, Cai Z, Pang Y, et al. Perinatal antidepressant exposure alters cortical network function in rodents. PNAS. 2011;108(45):18465-18470
  72. 72. Christensen DL, Bilder DA, Zahorodny W, Pettygrove S, Durkin MS, Fitzgerald RT, et al. Prevalence and characteristics of autism spectrum disorder among 4-year-old children in the autism and developmental disabilities monitoring network. Journal of Developmental and Behavioral Pediatrics. 2016;37:1-8
  73. 73. Gemmel M, Hazlett M, Bögi E, De Lacalle S, Hill LA, Kokras N, et al. Perinatal fluoxetine effects on social play, the HPA system, and hippocampal plasticity in pre-adolescent male and female rats: Interactions with pre-gestational maternal stress. Psychoneuroendocrinology. 2017;84:159-171
  74. 74. Ko MC, Lee LJH, Li Y, Lee LJ. Long-term consequences of neonatal fluoxetine exposure in adult rats. Developmental Neurobiology. 2014;74(10):1038-1051
  75. 75. Svirsky N, Levy S, Avitsur R. Prenatal exposure to selective serotonin reuptake inhibitors (SSRI) increases aggression and modulates maternal behavior in offspring mice. Developmental Psychobiology. 2016;58(1):71-82
  76. 76. Kiryanova V, Dyck RH. Increased aggression, improved spatial memory, and reduced anxiety-like behaviour in adult male mice exposed to fluoxetine early in life. Developmental Neuroscience. 2014;36(5):396-408
  77. 77. Zimmerberg B, Germeyan SC. Effects of neonatal fluoxetine exposure on behavior across development in rats selectively bred for an infantile affective trait. Developmental Psychobiology. 2015;57(2):141-152
  78. 78. Duke AA, Bègue L, Bell R, Eisenlohr-moul T. Revisiting the serotonin-aggression relation in humans: A meta-analysis. Psychological Bulletin. 2013;139(5):1148-1172
  79. 79. Blader JC. Pharmacotherapy and postdischarge outcomes of child inpatients admitted for aggressive behavior. Journal of Clinical Psychopharmacology. 2006;26(4):419-425
  80. 80. Bond AJ. Antidepressant treatments and human aggression. European Journal of Pharmacology. 2005;526(1-3):218-225
  81. 81. Kiryanova V, Meunier SJ, Vecchiarelli HA, Hill MN, Dyck RH. Effects of maternal stress and perinatal fluoxetine exposure on behavioral outcomes of adult male offspring. Neuroscience. 2016;320:281-296
  82. 82. Manhaes De Castro R, Barreto Medeiros JM, Mendes Da Silva C, Ferreira LMP, Guedes RCA, Cabrai Filho JE, et al. Reduction of intraspecific aggression in adult rats by neonatal treatment with a selective serotonin reuptake inhibitor. Brazilian Journal of Medical and Biological Research. 2001;34(1):121-124
  83. 83. Lisboa SFS, Oliveira PE, Costa LC, Venâncio EJ, Moreira EG. Behavioral evaluation of male and female mice pups exposed to fluoxetine during pregnancy and lactation. Pharmacology. 2007;80(1):49-56
  84. 84. Zoratto F, Fiore M, Ali SF, Laviola G, Macrì S. Neonatal tryptophan depletion and corticosterone supplementation modify emotional responses in adult male mice. Psychoneuroendocrinology. 2013;38(1):24-39
  85. 85. Johns JM, Joyner PW, McMurray MS, Elliot DL, Hofler VE, Middleton CL, et al. The effects of dopaminergic/serotonergic reuptake inhibition on maternal behavior, maternal aggression, and oxytocin in the rat. Pharmacology, Biochemistry, and Behavior. 2005;81(4):769-785
  86. 86. Pawluski JL, Charlier TD, Fillet M, Houbart V, Crispin HT, Steinbusch HW, et al. Chronic fluoxetine treatment and maternal adversity differentially alter neurobehavioral outcomes in the rat dam. Behavioural Brain Research. 2012;228(1):159-168
  87. 87. Rayen I, van den Hove DL, Prickaerts J, Steinbusch HW, Pawluski JL. Fluoxetine during development reverses the effects of prenatal stress on depressive-like behavior and hippocampal neurogenesis in adolescence. PLoS One. 2011;6(9):e24003
  88. 88. Dohler KD, Jarzab B, Sickmoller PM, Kokocinska D, Kaminski M, Gubala E, et al. Influence of neurotransmitters on sexual differentiation of brain structure and function. Experimental and Clinical Endocrinology. 1991;98:99-109
  89. 89. Balon R. SSRI-associated sexual dysfunction. The American Journal of Psychiatry. 2006;163(9):1504-1509
  90. 90. Gouvea TS, Morimoto HK, de Faria MJ, Moreira EG, Gerardin DC. Maternal exposure to the antidepressant fluoxetine impairs sexual motivation in adult male mice. Pharmacology, Biochemistry, and Behavior. 2008;90:416-419
  91. 91. Harris SS, MacIag D, Simpson KL, Lin RCS, Paul IA. Dose-dependent effects of neonatal SSRI exposure on adult behavior in the rat. Brain Research. 2012;1429:52-60
  92. 92. Maciag D, Simpson KL, Coppinger D, Lu Y, Wang Y, Lin RC, et al. Neonatal antidepressant exposure has lasting effects on behavior and serotonin circuitry. Neuropharmacology. 2006;31:47-57
  93. 93. Rayen I, Steinbusch HW, Charlier TD, Pawluski JL. Developmental fluoxetine exposure and prenatal stress alter sexual differentiation of the brain and reproductive behavior in male offspring. Psychoneuroendocrinology. 2013;38:1618-1629
  94. 94. Chan JSW, Snoeren EMS, Cuppen E, Waldinger MD, Olivier B, Oosting RS. The serotonin transporter plays an important role in male sexual behavior: A study in serotonin transporter knockout rats. The Journal of Sexual Medicine. 2011;8(1):97-108
  95. 95. Cagiano R, Flace P, Bera I, Maries L, Cioca G, Sabatini R, et al. Neurofunctional effects in rats prenatally exposed to fluoxetine. European Review for Medical and Pharmacological Sciences. 2008;12(3):137-148
  96. 96. Rayen I, Steinbusch HW, Charlier TD, Pawluski JL. Developmental fluoxetine exposure facilitates sexual behavior in female offspring. Psychopharmacologia. 2014;231:123-133
  97. 97. Grahame-Smith DG. Serotonin function in affective disorders. Acta Psychiatrica Scandinavica. Supplementum. 1989;350:7-12
  98. 98. Ansorge MS, Zhou M, Lira A, Hen R, Gingrich JA. Early-life blockade of the 5-HT transporter alters emotional behavior in adult mice. Science. 2004;306(5697):879-881
  99. 99. Rebello TJ, Yu Q, Goodfellow NM, Caffrey Cagliostro MK, Teissier A, Morelli E, et al. Postnatal day 2 to 11 constitutes a 5-HT-sensitive period impacting adult mPFC function. The Journal of Neuroscience. 2014;34(37):12379-12393. DOI: 10.1523/JNEUROSCI.1020-13.2014
  100. 100. Sarkar A, Chachra P, Vaidya VA. Postnatal fluoxetine-evoked anxiety is prevented by concomitant 5-HT2A/Creceptor blockade and mimicked by postnatal 5-HT2A/Creceptor stimulation. Biological Psychiatry. 2014;76(11):858-868
  101. 101. Maciag D, Williams L, Coppinger D, Paul IA. Neonatal citalopram exposure produces lasting changes in behavior which are reversed by adult imipramine treatment. European Journal of Pharmacology. 2006;532(3):265-269
  102. 102. Lee LJ, Lee LJH. Neonatal fluoxetine exposure alters motor performances of adolescent rats. Developmental Neurobiology. 2012;72(8):1122-1132
  103. 103. Karpova NN, Lindholm J, Pruunsild P, Timmusk T, Castrén E. Long-lasting behavioural and molecular alterations induced by early postnatal fluoxetine exposure are restored by chronic fluoxetine treatment in adult mice. European Neuropsychopharmacology. 2009;19(2):97-108
  104. 104. Bairy KL, Madhyastha S, Ashok KP, Bairy I, Malini S. Developmental and behavioral consequences of prenatal fluoxetine. Pharmacology. 2007;79:1-11
  105. 105. Zohar I, Shoham S, Weinstock M. Perinatal citalopram does not prevent the effect of prenatal stress on anxiety, depressive-like behaviour and serotonergic transmission in adult rat offspring. The European Journal of Neuroscience. 2015;43(4):590-600
  106. 106. Hansen H, Sanchez C, Meier E. Neonatal administration of the selective serotonin reuptake inhibitor Lu 10-134-C increases forced swimming-induced immobility in adult rats: A putative animal model of depression? The Journal of Pharmacology and Experimental Therapeutics. 1997;283(3):1333-1341
  107. 107. Toffoli LV, Rodrigues GM, Oliveira JF, Silva AS, Moreira EG, Pelosi GG, et al. Maternal exposure to fluoxetine during gestation and lactation affects the DNA methylation programming of rat’s offspring: Modulation by folic acid supplementation. Behavioural Brain Research. 2014;265:142-147
  108. 108. Knaepen L, Rayen I, Charlier TD, Fillet M, Houbart V, van Kleef M, et al. Developmental fluoxetine exposure normalizes the long-term effects of maternal stress on post-operative pain in Sprague-Dawley rat offspring. PLoS One. 2013;8(2):e57608
  109. 109. Glover ME, Pugh PC, Jackson NL, Cohen JL, Fant AD, Akil H, et al. Early-life exposure to the SSRI paroxetine exacerbates depression-like behavior in anxiety/depression-prone rats. Neuroscience. 2015;284:775-797
  110. 110. Boulle F, Pawluski JL, Homberg JR, Machiels B, Kroeze Y, Kumar N, et al. Developmental fluoxetine exposure increases behavioral despair and alters epigenetic regulation of the hippocampal BDNF gene in adult female offspring. Hormones and Behavior. 2016;80:47-57
  111. 111. Coleman FH, Christensen HD, Gonzalez CL, Rayburn WF. Behavioral changes in developing mice after prenatal exposure to paroxetine (Paxil). American Journal of Obstetrics and Gynecology. 1999;181:1166-1171
  112. 112. McAllister BB, Kiryanova V, Dyck RH. Behavioural outcomes of perinatal maternal fluoxetine treatment. Neuroscience. 2012;226:356-366
  113. 113. Krishnan V, Nestler E. Animal models of depression: Molecular perspectives Vaishnav. Current Topics in Behavioral Neurosciences. 2011;7:121-147
  114. 114. Porsolt RD, Le Pichon M, Jalfre M. Depression: A new animal model sensitive to antidepressant treatments. Nature. 1977;266(5604):730-732
  115. 115. Glover ME, Clinton SM. Of rodents and humans: A comparative review of the neurobehavioral effects of early life SSRI exposure in preclinical and clinical research. International Journal of Developmental Neuroscience. 2016;51:50-72
  116. 116. Hilakivi L, Hilakivi I. Increased adult behavioral “despair” in rats neonatally exposed to desipramine or zimeldine: An animal model of depression? Pharmacology, Biochemistry, and Behavior. 1987;28(3):367-369
  117. 117. Velazquez-Moctezuma J, Diaz RO. Neonatal treatment with clomipramine increased immobility in the forced swim test: An attribute of animal models of depression. Pharmacology, Biochemistry, and Behavior. 1992;42(4):737-739
  118. 118. Bhagya V, Srikumar BN, Raju TR, Shankaranarayana Rao BS. The selective noradrenergic reuptake inhibitor reboxetine restores spatial learning deficits, biochemical changes, and hippocampal synaptic plasticity in an animal model of depression. Journal of Neuroscience Research. 2015;93(1):104-120
  119. 119. Yang LM, Hu B, Xia YH, Zhang BL, Zhao H. Lateral habenula lesions improve the behavioral response in depressed rats via increasing the serotonin level in dorsal raphe nucleus. Behavioural Brain Research. 2008;188(1):84-90
  120. 120. Vazquez-Palacios G, Bonilla-Jaime H, Velazquez-Moctezuma J. Antidepressant effects of nicotine and fluoxetine in an animal model of depression induced by neonatal treatment with clomipramine. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2005;29:39-46
  121. 121. Limón-Morales O, Soria-Fregozo C, Arteaga-Silva M, Vázquez-Palacios G, Bonilla-Jaime H. Altered expression of 5-HT1A receptors in adult rats induced by neonatal treatment with clomipramine. Physiology & Behavior. 2014;124:37-44
  122. 122. Mendes-da-Silva C, De Souza SL, Barreto-Medeiros JM, De Freitas-Silva SR, Costa Antunes DE, Urbano Cunha AD, et al. Neonatal treatment with fluoxetine reduces depressive behavior induced by forced swim in adult rats. Arquivos de Neuro-Psiquiatria. 2002;60(4):928-931
  123. 123. Liu M-Y, Yin C-Y, Zhu L-J, Zhu X-H, Xu C, Luo C-X, et al. Sucrose preference test for measurement of stress-induced anhedonia in mice. Nature Protocols. 2018;13(7):1686-1698
  124. 124. Popa D, Lena C, Alexandre C, Adrien J. Lasting syndrome of depression produced by reduction in serotonin uptake during postnatal development: Evidence from sleep, stress, and behavior. The Journal of Neuroscience. 2008;28(14):3546-3554
  125. 125. Francis-Oliveira J, Ponte B, Barbosa APM, Veríssimo LF, Gomes MV, Pelosi GG, et al. Fluoxetine exposure during pregnancy and lactation: Effects on acute stress response and behavior in the novelty-suppressed feeding are age and gender-dependent in rats. Behavioural Brain Research. 2013;252:195-203
  126. 126. van Bodegom M, Homberg JR, Henckens MJAG. Modulation of the hypothalamic-pituitary-adrenal axis by early life stress exposure. Frontiers in Cellular Neuroscience. 2017;11:1-33
  127. 127. Daniels WMU, Pietersen CY, Carstens ME, Stein DJ. Maternal separation in rats leads to anxiety-like behavior and a blunted ACTH response and altered neurotransmitter levels in response to a subsequent stressor. Metabolic Brain Disease. 2004;19:3-14
  128. 128. Houwing DJ, Ramsteijn AS, Riemersma IW, Olivier JDA. Maternal separation induces anhedonia in female heterozygous serotonin transporter knockout rats. Behavioural Brain Research. 2019;356:204-207
  129. 129. Lee JH, Kim HJ, Kim JG, Ryu V, Kim BT, Kang DW, et al. Depressive behaviors and decreased expression of serotonin reuptake transporter in rats that experienced neonatal maternal separation. Neuroscience Research. 2007;58(1):32-39
  130. 130. Wei L, David A, Duman RS, Anisman H, Kaffman A. Early life stress increases anxiety-like behavior in Balbc mice despite a compensatory increase in levels of postnatal maternal care. Hormones and Behavior. 2010;57:396-404
  131. 131. Schmauss C, Lee-McDermott Z, Medina LR. Trans-generational effects of early life stress: The role of maternal behavior. Scientific Reports. 2014;4:1-11
  132. 132. Gemmel M, Kokras N, Dalla C, Pawluski JL. Perinatal fluoxetine prevents the effect of pre-gestational maternal stress on 5-HT in the PFC, but maternal stress has enduring effects on mPFC synaptic structure in offspring. Neuropharmacology. 2018;128:168-180
  133. 133. Rayen I, Steinbusch HWM, Charlier TD, Pawluski JL. Developmental fluoxetine exposure and prenatal stress alter sexual differentiation of the brain and reproductive behavior in male rat offspring. Psychoneuroendocrinology. 2013;38(9):1618-1629
  134. 134. Boulle F, Pawluski JL, Homberg JR, Machiels B, Kroeze Y, Kumar N, et al. Prenatal stress and early-life exposure to fluoxetine have enduring effects on anxiety and hippocampal BDNF gene expression in adult male offspring. Developmental Psychobiology. 2016;58(4):427-438
  135. 135. Salari AA, Fatehi-Gharehlar L, Motayagheni N, Homberg JR. Fluoxetine normalizes the effects of prenatal maternal stress on depression- and anxiety-like behaviors in mouse dams and male offspring. Behavioural Brain Research. 2016;311:354-367

Written By

Laura Staal and Jocelien DA Olivier

Submitted: 03 October 2018 Reviewed: 21 December 2018 Published: 14 February 2019