How to Deliver the Baby if Oxytocin Receptors Are Not Responding

J Midwifery Womens Health. Author manuscript; bachelor in PMC 2015 January 28.

Published in final edited form as:

PMCID: PMC3947469

NIHMSID: NIHMS506762

BEYOND LABOR: THE Role OF NATURAL AND SYNTHETIC OXYTOCIN IN THE TRANSITION TO Maternity

Abstract

Endogenous oxytocin is a key component in the transition to motherhood affecting molecular pathways that buffer stress reactivity, support positive mood, and regulate healthy mothering behaviors (including lactation). Synthetic oxytocin is widely used throughout labor and postpartum care in modern obstetrics. Nevertheless research on the implications beyond labor of maternal exposure to perinatal constructed oxytocin is rare. In this article, we review oxytocin-related biological pathways and behaviors associated with the transition to motherhood, and evidence supporting the need for farther research on potential effects of intrapartum oxytocin beyond labor. We include a primer on oxytocin at the molecular level.

Keywords: oxytocin, nativity, mothering, Pitocin, labor, stress, mood, postpartum depression, lactation, breastfeeding

Introduction

Consequences of routine childbirth interventions on human maternal behavior accept been understudied. Synthetic oxytocin (Pitocin®, Syntocinon®) stimulates uterine smoothen muscle contractility and is widely used in the Us for labor induction, augmentation, and third stage management. While the judicious utilise of constructed oxytocin has many benefits, the biological and behavioral effects of synthetic oxytocin across the immediate clinical uses remain largely unknown. Co-ordinate to the U.South. Centers for Illness Control and Prevention Vital Statistics written report, induction has more than doubled from 1990 (10%) to 2010 (23%).one The best current guess of the augmentation rate in the U.S. is 57%.2 While endogenous oxytocin is well known for its office in labor and lactation, a large body of evidence documents numerous oxytocin-mediated molecular and endocrine pathways that buffer stress reactivity, back up emotional and mental well-being, and promote pro-social and bonding behavior.iii, 4 These behaviors are critical for successful transition to maternity. Given the predominance of synthetic oxytocin in clinical practice, inquiry is needed on how constructed oxytocin may impact the intrinsic regulation of endogenous oxytocin and subsequent oxytocin-related outcomes. In this review, we examine the hypothesis that exposure to synthetic oxytocin during childbirth may play a function in maternal stress reactivity, mood, and mothering behaviors (including lactation).

OXYTOCIN ON THE MOLECULAR LEVEL

Profound changes occur in the oxytocin system during the perinatal menstruum.5, half dozen To preclude preterm birth, oxytocin neurons are kept quiescent during pregnancy through inhibitory mechanisms. The peptide oxytocin continues to accumulate in the posterior pituitary, and at term those inhibitory mechanisms are removed for labor to occur. Oxytocin likewise becomes more available at term through the reduction of enzymatic activity that metabolizes oxytocin in the brain. Based on animal studies, the expression of oxytocin receptors (OTR) increases throughout pregnancy in key areas of the brain that regulate mood, stress and attachment behavior. In humans, the availability of OTR in uterine muscle also increases dramatically at term, preparing for the surges of oxytocin about to be released during birth.

The neurologic origin of oxytocin

Oxytocin is a small neuropeptide consisting of nine amino acids.7 Throughout the human lifespan, specific neurons manufacture oxytocin; these cells are abundant in distinct areas of the mammalian hypothalamus called the paraventricular and supraoptic nuclei. Oxytocin from these cells is carried to and released from the posterior pituitary gland into the apportionment, and from at that place is distributed throughout the body. Within the central nervous system, oxytocin reaches nearly all parts of the brainstem, midbrain, cortex, and spinal column. In addition to hypothalamic product, peripheral organs and tissues also may secrete oxytocin, but the pituitary is believed to be the predominant source of oxytocin in circulation.seven

Oxytocin receptor: how oxytocin affects physiology in the brain and body

To impact target tissues, a receptor must be present and oxytocin presumably must demark to that receptor earlier it can exert cellular action. Oxytocin receptors (OTR) are found throughout the body, with particularly loftier concentrations in the limbic regions of the brain, spinal cavalcade, heart, intestines, immune tissue, uterus, and breast. The OTR belongs to a large family of receptors called G-protein coupled receptors, manufactured by the cell and inserted into the jail cell membrane where they are available for hormone-binding.7 Thousand-protein Coupled Receptors loop in and out of the cell membrane seven times and are coupled to a Thou-protein located on the inside of the cell. Many varieties of K-proteins are known, each initiating a different cascade of events (second messengers) within the cell, which provides specificity to the hormone'south action. The OTR is coupled to a Chiliad-qα. This type of K-protein leads to a rise in intracellular calcium (Ca+) and a muscle prison cell contraction, of particular importance to milk let-down and uterine contractions (see Figure 1).7, 8

An external file that holds a picture, illustration, etc.  Object name is nihms506762f1.jpg

However, when the OTR is on a neuron, the response may be the subsequent release or inhibition of other hormonal neurotransmitters and modulators, such equally serotonin, endogenous opioids and corticotrophin-releasing factor.3 These nervous system interactions are cardinal to understanding how oxytocin released in the brain influences a variety of mental states and behavior. It may too help to explain how the nervous arrangement is stimulated in response to a human or creature'south environs (both external and internal) and afterward leads to the release or inhibition of oxytocin. Additionally, oxytocin binds to other types of receptors, such as vasopressin receptors exerting agonist or adversary effects, thus extending and diversifying the consequences of oxytocin's actions.

Oxytocin-mediated pathways beyond muscle wrinkle

Within all cells, not but neurons, iii other of import functions of the OTR lead to an array of possible cellular actions. ane) Through intracellular 1000-protein activation, phospholipase C causes an even greater amount of Ca+ release into the cell from internal stores. This Ca+ can serve as an independent indicate for diverse functions inside the cell, especially nerves. 2) Another role is the creation of eicosanoids or prostaglandin that can directly increase pain, inflammation, and likely uterine contraction as well. 3) Thirdly, OTR activation may lead to a broad category of cellular events by activating a specific kinase, an enzyme that catalyzes the addition of a phosphate grouping on a specific target molecule or protein. In this example, protein kinase C is activated and requires Ca+. The "downstream" effects of OTR activation will depend on what type of jail cell the receptor is located on. This kinase may initiate a specific action itself or cause some other kinase to activate, and and so another, creating a literal pour of events.7 These events tin can ultimately lead to modifying gene transcription, regulation of the jail cell cycle, apoptosis and/or neurogenesis. The end result for each oxytocin-initiated pathway depends on the type of jail cell involved (uterine, encephalon, heart etc.) and the type of response initiated within that jail cell. OTR activation has the potential for inducing long-lasting biological alterations.

CHALLENGES IN STUDYING OXYTOCIN ON THE MOLECULAR LEVEL—BRAIN VERSUS Trunk

In the intrapartum setting, the half-life of Pitocin® is believed to be only a few minutes,9 and intrapartum plasma levels correlate with Pitocin® dose and rate of assistants,10 nevertheless the dose delivered intravenously may or may not result in an effective uterine contraction pattern. Similarly, oxytocin levels in plasma cannot always exist interpreted equally being meaningful in a detail event on brain activity. In order to understand how perinatal oxytocin exposure has the potential for lasting biologic consequence, it is helpful to empathize some of the constraints that can limit research or the interpretation of research in this field. Many research studies examining endogenous oxytocin in animals and humans rely on blood measurement of the hormone in response to a treatment or intervention, e.g. a stressful event or a social interaction. In that location are several challenges in the interpretation of these measurements.

Firstly, central nervous organisation oxytocin is secreted continuously acting inside the brain and spinal cord, but oxytocin is as well released in pulses into the bloodstream through the posterior pituitary. Pulsatile release of oxytocin occurs when oxytocin neurons of the posterior pituitary depolarize, which are usually in response to specific stimuli (due east.g., uterine stimuli or cues from an baby).eleven, 12 This activation of neurons pulses oxytocin into the blood stream. Data collected in rats suggest correlations betwixt endogenous peripheral (blood stream) and cardinal oxytocin levels, although the degree and significance of these correlations vary.xiii

In addition, there is controversy regarding whether peripherally administered oxytocin (i.eastward. Pitocin® given intravenously or intramuscularly) crosses the blood-brain barrier. Whether whatever oxytocin that does cantankerous changes neuronal action significantly within the nervous arrangement has nonetheless to exist determined.xiv In theory, oxytocin cannot pharmacologically cross due to its relatively large size and hydrophilic nature, however, some animal studies do study depression levels of oxytocin found in the brain following administration in blood15 Interestingly, some electrophysiology-based animal studies advise that maternal oxytocin plays a neuroprotective office within the fetal encephalon during the nascency process, which means it would take to cross both the placental barrier and fetal blood-brain barrier.16 The findings from these studies advise that maternal oxytocin inhibits certain excitatory (GABA) fetal neurons from firing (depolarizing), thereby protecting them during periods of hypoxia (i.e. birth process). Nonetheless, whether or non synthetic or endogenous oxytocin penetrates the maternal encephalon directly has all the same to be proven. While oxytocin does exit the posterior pituitary and is released into circulation, this occurs following neuronal activation (activeness potential). Getting the peripherally circulating hormone into the brain would require either one) an agile transport mechanism to penetrate the tight junctions guarding the microvasculature of the central nervous system (which has withal to exist proven), or 2) it would require a more than porous bulwark to let for improvidence.17 The blood-brain barrier (maternal or fetal) may become more than porous in states of disease or stress.

Lastly, the brain may receive data about peripheral levels of oxytocin through feedback from peripheral nervous system. Peripheral fretfulness may communicate data about oxytocin levels to the encephalon, presumably via a feedback loop (eastward.chiliad. cervical dilation, adrenal gland activity, touch/ nipple stimulation). In that location are also well-studied effects of intranasal administration of synthetic oxytocin on mood and social behavior, nevertheless it remains unknown whether the intranasal route allows oxytocin to enter the brain straight or extraneuronally, or whether it stimulates feed forward effects on endogenous oxytocin via ascending or afferent neuropathways (i.e. the vagus or xth cranial nerve), which are well known to have OTRs.14 xviii Peripheral feedback furnishings of oxytocin, which may be relayed to the encephalon, are difficult to monitor, simply farther complicate the study of synthetic oxytocin. Whether the maternal brain will reliably answer to exogenous oxytocin past decreasing or increasing the synthesis or release of endogenous oxytocin is unknown. In the clinical setting, this blazon of feedback might be seen when Pitocin® is used to initiate an induction of labor just and so can sometimes be close off while the woman continues to labor without the drug. Probable, feedback from peripheral fretfulness messaging about cervical dilation to the brain is in activity, which promotes the woman's endogenous oxytocin release, and this is more than probable than the idea that Pitocin® penetrates the maternal brain directly.

Yet, given all these variables, altered maternal plasma levels persisting across the end of labor have been suggested in 1 written report that evaluated postpartum oxytocin levels in response to breastfeeding 2 days later on birth in women who had different intrapartum and postpartum exposures to synthetic oxytocin (n=xl).nineteen Compared to all other study groups, women exposed to Pitocin® in labor combined with an epidural demonstrated significantly lower oxytocin levels during breastfeeding. Overall, the total quantity of constructed oxytocin administered during parturition was negatively correlated to levels of oxytocin in plasma two days following birth. All of these women had vaginal births and newborns had normal Apgar scores. In these studies the mean elapsing of labor did not differ significantly betwixt groups, nor did blood loss, or newborn weight. The women all initiated breastfeeding within minutes of birth and had the same boilerplate number of feeds in the days prior to the blood sampling. If replicable, this finding suggests that in some cases exposure to Pitocin® may have maternal consequences that last across the nativity experience.

Altering the oxytocin receptor could modify how oxytocin works

Equally reviewed above, the OTR must be present for oxytocin to exert its action. An important consideration for whether synthetic oxytocin may affect maternal physiology is the chapters of the OTR to become saturated. In response to saturation, sometimes a receptor is internalized, (i.e., removed from the cell-membrane where it is presumed to be unavailable and potentially degraded). In the presence of high levels of an agonist, receptor internalization may begin within minutes. The opposite process has been shown to take approximately four hours to resensitize after the agonist is removed.20 However, work on sensitization has focused on myometrial cells, studied in vitro, and information technology is unknown if OTR on neurons will internalize and resensitize on the cell membrane in vivo. If neuronal OTRs undergo a comparable process, this could have implications for maternal behavior.

Another strategy against saturation is that receptors may exist internalized and then down-regulated, through a pause in mRNA gene transcription for the receptor.20 Inquiry on induced labor in humans has focused on sampling the myometrium for expression of the OTR cistron.21 One example is a study that compared women in spontaneous labor with those undergoing an induction of labor and 29 women who planned constituent cesarean commitment. Eighteen of the 33 women in the spontaneous group eventually received augmentation with constructed oxytocin while 26 of 30 women of the induced group did as well. All laboring participants underwent cesarean delivery (indicated for failure to progress or fetal intolerance to labor) and the myometrium was sampled at that time. Oxytocin binding, also as mRNA levels of the OTR, was significantly affected by employ of synthetic oxytocin. Participants with oxytocin-induced labor had a 300-fold downwards-regulation of the OTR cistron in uterine muscle, when compared to receptor availability in spontaneous labor.21 This study suggests that the OTR can down-regulate in the uterus during augmented or induced labor, and points to the demand to study oxytocin binding in other areas of the body such as the maternal brain, breast, heart, intestine or immune system. Whether "agile direction" of 3rd stage of labor also results in down-regulation of receptors has not been reported, but given the prevalence of this exercise, it deserves consideration.

The duration of mRNA downwardly-regulation in the OTR in response to synthetic oxytocin is non yet known. Considering the cellular mechanism for receptor regeneration would include mRNA transcription, translation, protein assembly/folding and ship to the cell membrane, this could take many more hours than simple internalization of the receptor, and full restoration of a functional OTR might require days. Besides, after a given tissue is no longer exposed to a saturating agonist (labor), and if there is no stimuli for releasing endogenous oxytocin (east.g. touch, breastfeeding), the response to the perceived "need" of the system may be unlike between different types of birth and postpartum experiences.

The role of epigenetic regulation of the OTR

On a more long-term level, receptor regulation as well can occur at the level of gene transcription for the receptor through epigenetic modulation. For instance, methylation is i mechanism through which gene expression is down regulated. Zipper of a methyl group (CH3) can occur on specific sites along the Deoxyribonucleic acid sequence. A receptor gene that is more than heavily methylated selectively "silences" the cistron, preventing activation for transcription. Methylation of the OTR gene is one instance of a mechanism that can down-regulate OTR factor expression, with effects that may be heritable. For case, if the OTR gene is silenced, less OTR volition be available on the cell membrane. In turn, the OTR is less available to bind with oxytocin potentially resulting in diminished biological and behavioral outcomes.twenty

There are sensitive periods during mammalian development in which the environs can shape Deoxyribonucleic acid methylation.22 For example, rodent models evidence that early on maternal care can be linked to patterns of methylation in both maternal and offspring phenotypes with a transgenerational effect.23 Emerging evidence supports the hypothesis that epigenetic modification of the OTR has a role in social knowledge, stress reactivity, and social behavioral disorders.24 For example, one study has examined the role of methylation of the OTR in autism-afflicted persons. Hypermethylation of the region of Deoxyribonucleic acid controlling the OTR was seen in blood samples of affected individuals compared to controls (due north=20 matched pairs). This issue also was demonstrated in postmortem encephalon sampling of 8 matched patient-controls, showing a correlation between brain and blood methylation in the OTR.25 Airplane pilot data in rodents advise that normal birth with endogenous oxytocin, every bit well as exposure to intrapartum synthetic oxytocin, may produce epigenetic modulation of the OTR by increasing methylation of sites in the OTR factor of the maternal hypothalamus.26

OXYTOCIN AND TRANSITION TO MOTHERHOOD

The experience of giving birth and becoming a mother, particularly for the offset time, demands a high level of concrete and social interaction. Being able to sensitively intendance for the needs of the infant through synchronous female parent-baby interaction is vital to the continuation of the family unit and species. The postpartum menstruum is as well characterized past drastic hormonal shifts, transition to maternity, coping with new stressors, physical pain, lactation and attachment - all of which involve the endogenous oxytocin system. Furthermore, modern parenting can include fiscal strains, work obligations, social isolation or limited support, and socio-cultural constructs virtually "good" mothering. Within this context, a difficult transition to maternity holds the potential to lead to dysregulated stress reactivity, mood disturbances, susceptibility to less sensitive mothering, asynchronous female parent-infant interaction, and poor infant zipper.

Stress Reactivity

The maternal brain is a distinctive biological land, characterized by a host of biochemical mechanisms supporting the well-being and survival of both mother and infant.half-dozen Significant adaptations occur in the maternal oxytocin system, including protection from the stress and demands of the perinatal period The dramatic rise of oxytocin during physiologic birth may play a role in buffering the stress hormones released past fear and pain during labor according to animal studies and some man work.27 This adaptive response is likely a protective mechanism during the perinatal menstruum, a fourth dimension of intense stress.

The relationship between oxytocin and the HPA (hypothalamic-pituitary-adrenal) axis in the body's response to stressful stimuli has been investigated primarily using rodent models.iii These demonstrate that significant animals have reduced reactivity to stressors via lowered plasma levels of corticotrophin releasing factor, adrenocorticotropin releasing factor, and cortisol. In that location is inhibition of both oxytocin and HPA neurons during pregnancy mainly due to an inhibitory opioid machinery from increased allopregnanolone (a metabolite of progesterone).6 During lactation, oxytocin pulses in the brain increase and the hormone is secreted into circulation. Meanwhile brain levels of oxytocin as well increase and influence the neurons linked to the stress response system. Lactating females react less to stressors and brandish less anxiety-like behavior than non-lactating females. In response to stress oxytocin increases, mayhap as a protective mechanism against continued stress. This can be seen when cortisol and adrenocorticotropin releasing factor decrease subsequently assistants of constructed oxytocin. Conversely, when an oxytocin antagonist is given to rats, their levels of cortisol increase. Withal, in humans the relationships among oxytocin, HPA function, and stress reactivity are less well characterized. Measurements of salivary oxytocin in lactating women advise that oxytocin may increase prior to feeding, when women are preparing to breastfeed.11 There is also a decrease in circulating HPA hormones immediately after breastfeeding is initiated. Likely this is due to oxytocin within the brain exerting an effect on neurons that activate the HPA axis and corticotrophin releasing cistron. Lactating women bear witness increased vagal tone, decreased claret force per unit area and decreased heart rate when compared to non-lactating women, specially in response to a stressor.28 As discussed earlier the vagus nerve detects elevated levels of oxytocin within the body and can feedback to the encephalon via afferent pathways. There is increasing evidence of a role for oxytocin in buffering stress reactivity, suggesting that oxytocin and the HPA systems are intricately linked.27

Maternal Mood

Equally with stress reactivity, a well-regulated oxytocin system is anxiolytic and confers protection against negative mood. Several studies have shown that intranasal assistants of synthetic oxytocin has an anxiolytic effect in psychiatric disorders.14 Whether intrapartum constructed oxytocin confers the same protective function every bit endogenous oxytocin on maternal mental wellness is more difficult to determine, especially in light of the complexity of gimmicky birth practices. Maternal low in the postpartum menstruation is estimated to impact up to 19% of women.29 Women experiencing negative mood are less likely to testify positive mothering behaviors, and are less sensitive to babe needs.30 The decreased quality of mother-baby interaction may lead to suboptimal infant attachment,31 placing infants at risk for poor evolution.32

While there are well-known predictors of postpartum negative mood (eastward.g., poor social support, stressful/adverse life events, and history of depression/feet), subjective and objective birth variables (i.eastward., complications, fashion of delivery, increased utilise of interventions, and maternal perception of the experience) also may be predictors of maternal issue.33, 34 Notwithstanding, trivial is known of the biological underpinnings linking birth variables to postpartum mood, the specific event of exposure to constructed oxytocin has not been teased apart. Apply of synthetic oxytocin is often associated with preexisting complications, but even in low-risk situations synthetic oxytocin tin can precipitate a cascade of interventions and subsequent nascency complications.35 Whether exposure to synthetic oxytocin during childbirth affects postpartum mood is unknown. Even so, based on our cognition of the actions of oxytocin in other situations and in tissues outside of the cardinal nervous organisation, we would conceptualize that any effects of synthetic oxytocin would be dose-dependent and would show individual differences, influenced by context and the history of the female parent.36

Rodent models tin can elucidate molecular pathways of mood that have been evolutionarily conserved in mammals.37 For instance, serotonin and dopamine are mediators of oxytocin'due south anxiolytic actions in both humans and rodents. When endogenous oxytocin is genetically or pharmacologically blocked, anxiety-like and depression-like behavior increases in oxytocin-deficient knockout mice compared to wild-type mice.38 In responding to a stressor, rats bred for "high feet" exhibit a higher release of cardinal oxytocin and greater anxiety-like and low-like symptoms than rats bred for "low anxiety".39 Oxytocin is one of the evolutionarily conserved molecular pathways of mood.

In humans, numerous studies have establish that atypical peripheral oxytocin levels (very high or very low) may exist associated with elevated symptoms of low, anxiety, or postal service-traumatic stress.40 In one perinatal example (due north=74), low levels of oxytocin in late pregnancy were associated with elevated symptoms of depression at ii weeks postpartum, controlling for prepartum symptoms, socio-demographics, and nascence-outcomes.41 Private context, adversities across the life cycle, history of trauma, genotype, and epigenetic processes are all factors that may program the oxytocin system altering (and possibly increasing) sensitivity to synthetic oxytocin during childbirth.

Mothering Behaviors

Endogenous oxytocin's role in mediating the initiation of maternal beliefs has been demonstrated in numerous nonhuman species. Perinatal manipulation of the oxytocin organisation in animals provides potent prove of subsequent dysfunctional maternal behaviors.42–46 For case, in rats, oxytocin clearly mediates the initiation of maternal behavior.44 In ewes, maternal acceptance of their own lamb occurs later on identification, yet a fundamental injection of constructed oxytocin can promote maternal acceptance of conflicting lambs.43 Optimal maternal behavior is blocked in ewes and heifers when central oxytocin is not released in physiologic nascence due to regional anesthesia and subsequent lack of vagino-cervical stimulation.46 In nonhuman primates, optimal maternal behavior tin can be altered past a central injection of synthetic oxytocin or by an oxytocin adversary.42, 45

One detail animal model has been useful in understanding the role of oxytocin in forming social bonds. The socially monogamous prairie vole forms pair bonds, an uncommon phenomenon among rodents of the contrary sexual activity. Information technology is postulated that the function of oxytocin buffers stress-reactivity as a function of social interaction and bonding.4 Enquiry with this model points to the possibility of altering social behavior equally a part of exposure to synthetic oxytocin early in life. For case, in prairie vole pups, exposure to synthetic oxytocin on the first twenty-four hours of life had lasting and dose-dependent effects on the capacity to form pair bonds in later life. In this model, exposure to a low dose of constructed oxytocin facilitated pair bonding later in that pup'southward life, while exposure to a high dose inhibited pair bail formation.47 Exposure to an oxytocin antagonist in the aforementioned time period inhibited subsequent social behaviors including the typical willingness to care for unrelated infants (alloparenting), peradventure mediated by increases in feet.47 In addition, physiologic nativity itself may be critical in the initiation of prairie vole maternal beliefs. Female voles delivered by cesarean surgery have demonstrated infanticidal behavior; while females delivered vaginally (and that underwent a sham surgery following birth) did not.48

A growing body of evidence suggests a link between oxytocin and optimal mothering behaviors in humans every bit well.49–53 Optimal mothering behaviors include affectionate touch, eye-to-middle contact, positive affect, and affectionate language that are characterized past sensitivity to babe cues and synchronous mother-infant interaction.50 Synchronicity in mother-baby interaction has a strong effect on baby affective states. Numerous studies have institute an clan between atypical peripheral oxytocin levels and less optimal mothering behavior.50 Genetic variation (i.eastward., risk alleles), and decreased central bounden, of the OTR gene have as well been associated with less optimal mothering behavior.49, 51, 52 A contempo fMRI report of 15 parent-infant dyads found two singled-out brain -beliefs-oxytocin profiles in mothers displaying synchronous versus intrusive mothering behavior.53 Mothers who displayed synchronicity in mother-baby interaction, had plasma oxytocin levels correlating with neural arrangement in reward-related motivational areas of the brain (left nucleus accumbens and right amygdala). In contrast, mothers who displayed intrusiveness in mother-infant interaction had no meaning correlation between activation of neural advantage areas and oxytocin levels. Additionally, the part of oxytocin in mothering behavior has been linked with the woman's affiliative experiences throughout her life (eastward.g., her own parents, partner and infant).54 Again, it is unclear if any of these relationships are derived or influenced past the birth experience, or apply of synthetic oxytocin. However, these findings do propose that oxytocin plays a key role, beyond labor, in the transition to motherhood.

Lactation

The physiological transition to motherhood also includes establishing lactation. A few contempo studies have examined lactation in the context of synthetic oxytocin and use of epidural anesthesia. For case, in Sweden 351 women who received an epidural were case-control matched with 351 women who did non receive an epidural.55 Breastfeeding success was negatively associated with epidural utilise. Chiefly, women who were augmented with synthetic oxytocin were 3 times less likely to initiate breastfeeding in the start four hours, and two times more likely to requite artificial milk by the time of hospital discharge. Another small study (n=20) examined breastfeeding elapsing in relationship to intrapartum exposure to synthetic oxytocin during induction or augmentation of labor. All mothers had epidural anesthesia. Authors reported an inverse relationship betwixt synthetic oxytocin dose and a shorter duration of exclusive breastfeeding by three months.56

Consequences for the offspring

Evidence for long-term negative consequences for social behavior and the management of stressful experiences accept repeatedly appeared in animal studies of offspring exposed to manipulations past oxytocin in early life. For example, work in piglets revealed that exposure to intranasal oxytocin in early life produced singular, nonreciprocal social behavior and an altered capacity to respond to stressful experiences in afterwards life.57 Several studies in rodents similarly back up the hypothesis that exposure to synthetic oxytocin, especially at high levels, during the perinatal menstruation tin have effects on the offspring.4

Studies of the long-term consequences of perinatal oxytocin exposure for children are less common. All the same, authors recently reported - based on a report in New York City of 3000 total term infants - that Pitocin®-treated infants showed an increase in multiple "agin outcomes" including reductions in Apgar scores (indexed by increased pulse, breathing rate and "reflex irritability") and increased access to the NICU.58 Increased admission to the NICU and other agin effects, with an estimated 30% increment in measures of morbidity, also were seen in a 2012 report from Australia.59 Neurodevelopmental risk for the offspring also was suggested by the finding that the occurrence of attention deficit disorders was twice equally likely in children exposed to Pitocin® during nativity.lx

Several studies have linked exposure to synthetic oxytocin to reductions in lactation, and diminished feeding-related beliefs in the newborn.56, 61 These studies underscore the need for further research, particularly taken in the context of a growing experimental literature in animals linking long-term behavioral outcomes to exposure to synthetic oxytocin in the perinatal period.

CONCLUSION

Oxytocin is a neuroendocrine hormone with complex deportment throughout the body and effects vital to the mother-infant dyad and social well-being. Much remains to be understood about the role oxytocin plays in the transition to motherhood; however, emerging inquiry in both animal and human models highlights the need for a deeper understanding of the part of physiologic birth in female parent-babe biobehavioral outcomes important to the disciplines of midwifery and obstetrics.

Clearly, the role of oxytocin in the body extends far beyond uterine contractility to molecular cell systems that have potential long-term consequences. Downstream molecular effects of naturally-expressed oxytocin and synthetic oxytocin take not been investigated thoroughly in the context of human birth intendance. Midwifery and obstetric research should consider the oxytocin organisation equally a whole, not just the immediate clinical effect, when investigating the role of physiologic birth besides as birth interventions on biobehavioral outcomes in mothers and infants.

Enquiry questions abound regarding the long-term implications of manipulating the oxytocin system during childbirth - an intricate transitional window of time for both female parent and babe. One instance may be early identification of women at-adventure for postpartum mood disorders or lactation difficulties. Identifying at-risk women could potentially exist informed past the interaction of OTR genotype, OTR epigenotype, and differential nascency experiences impacting the regulation of endogenous oxytocin.

While many basic questions remain, we suggest that birth practitioners may benefit from an appreciation of the molecular, developmental and behavioral consequences of one of the most widely used drugs in obstetric practice. Given the lack of clarity and definitive research on the furnishings of oxytocin beyond labor, the dedication of wellness care professionals to minimal-interference in biologically-regulated and evolutionarily-conserved processes is warranted. In that location is peachy potential for interdisciplinary collaboration equally the ubiquitous use of synthetic oxytocin in modern birth continues.

Quick Points

  • Downstream molecular furnishings of constructed oxytocin have rarely been investigated in the context of human nascency care.

  • The office of natural oxytocin includes molecular pathways in the transition to maternity, such as buffering stress reactivity, supporting positive mood and regulating healthy mothering behaviors.

  • Given the action of natural oxytocin on various endocrine pathways, we anticipate that any effects of intrapartum synthetic oxytocin would be dose-dependent and influenced by individual context and maternal history.

  • With the ubiquitous use of synthetic oxytocin in modern nascency care, enquiry questions grow regarding long-term implications of manipulating the oxytocin organization during labor – a complex transitional window of development for both mother and infant.

Acknowledgments

This original review was supported by the National Center for Advancing Translational Sciences, National Institutes of Wellness, through Grant UL1TR000050. The content is solely the responsibleness of the authors and does non necessarily represent the official views of the NIH.

Nosotros thank The Fetzer Establish for their generous back up of research on optimal birth

Biographies

Aleeca F. Bong, PhD, CNM, is an Banana Professor at the Academy of Illinois at Chicago, College of Nursing, Chicago, Illinois, and also a Center for Clinical and Translational Science KL2 Scholar.

Elise Erickson, CNM, MSN, is in clinical practice as a Certified Nurse Midwife and Clinical Supervisor at Columbia Women'due south Clinic, Providence Medical Group in Portland, Oregon.

C. Sue Carter, PhD, is Research Professor in the Section of Psychiatry, University of North Carolina, Chapel Hill, NC 27599

Footnotes

Conflict of involvement disclosure:

The authors report no conflicts of interest.

References

1. Martin JA, Hamilton BE, Ventura SJ, Osterman MJK, Wilson TJ, Mathews T. Births: Last information for 2010. In: Reports NVS, editor. Hyatttsville, Md: National Center for Health Statistics; 2012. [PubMed] [Google Scholar]

2. Declercq ER, Sakala C, Corry MP. Listening to mothers II: report of the 2d national U.Southward. survey of women's childbearing experiences. New York: Childbirth Connection; 2006. Executive summary. [Google Scholar]

iii. Neumann ID, Landgraf R. Balance of brain oxytocin and vasopressin: implications for anxiety, depression, and social behaviors. Trends Neurosci. 2012;35(11):649–659. [PubMed] [Google Scholar]

4. Carter CS, Boone EM, Pournajafi-Nazarloo H, Bales KL. Consequences of early on experiences and exposure to oxytocin and vasopressin are sexually dimorphic. Dev Neurosci. 2009;31(4):332–341. [PMC free article] [PubMed] [Google Scholar]

five. Brunton PJ, Russell JA. Endocrine induced changes in brain function during pregnancy. Brain Res. 2010;1364:198–215. [PubMed] [Google Scholar]

6. Brunton PJ, Russell JA. The expectant brain: adapting for maternity. Nat rev Neurosci. 2008;9(ane):11–25. [PubMed] [Google Scholar]

7. Gimpl Grand, Fahrenholz F. The oxytocin receptor system: structure, function, and regulation. Physiol Rev. 2001;81(2):629–683. [PubMed] [Google Scholar]

8. Devost D, Wrzal P, Zingg HH. Oxytocin receptor signalling. Prog Brain Res. 2008;170:167–176. [PubMed] [Google Scholar]

nine. Arias F. Pharmacology of oxytocin and prostaglandins. Clin Obstet Gynecol. 2000;43(three):455–468. [PubMed] [Google Scholar]

ten. Perry RL, Satin AJ, Barth WH, Valtier S, Cody JT, Hankins GD. The pharmacokenetics of oxytocin as they apply to labor induction. Am J Obstet Gynecol. 1996;174(v):1590–1593. [PubMed] [Google Scholar]

11. White-Traut R, Watanabe G, Pournajafi-Nazarloo H, Schwertz D, Bell A, Carter CS. Detection of salivary oxytocin levels in lactating women. Dev Psychobiol. 2009;51(4):367–373. [PMC gratis article] [PubMed] [Google Scholar]

12. Israel JM, Poulain DA, Oliet SH. Oxytocin-induced postinhibitory rebound firing facilitates bursting activity in oxytocin neurons. J Neurosci. 2008;28(two):385–394. [PMC free article] [PubMed] [Google Scholar]

thirteen. Neumann ID, Torner Fifty, Toschi N, Veenema AH. Oxytocin actions within the supraoptic and paraventricular nuclei: differential effects on peripheral and intranuclear vasopressin release. Am J Physiol Regul Integr Comp Physiol. 2006 [PubMed] [Google Scholar]

fourteen. Churchland PS, Winkielman P. Modulating social behavior with oxytocin: how does it work? What does it mean? Horm Behav. 2012;61(3):392–399. [PMC free article] [PubMed] [Google Scholar]

15. Ermisch A, Barth T, Ruhle HJ, Skopkova J, Hrbas P, Landgraf R. On the blood-brain bulwark to peptides: accumulation of labelled vasopressin, DesGlyNH2-vasopressin and oxytocin past brain regions. Endocrinol Exp. 1985;xix(1):29–37. [PubMed] [Google Scholar]

16. Khazipov R, Tyzio R, Ben-Ari Y. Furnishings of oxytocin on GABA signalling in the foetal encephalon during delivery. Prog Brain Res. 2008;170:243–257. [PubMed] [Google Scholar]

17. McEwen BB. Brain-fluid barriers: relevance for theoretical controversies regarding vasopressin and oxytocin memory research. Adv Pharmacol. 2004;l:531–592. 655–708. [PubMed] [Google Scholar]

18. Uvnas-Moberg Grand. Office of efferent and afferent vagal nervus action during reproduction: integrating function of oxytocin on metabolism and behaviour. Psychoneuroendocrinology. 1994;19(5–seven):687–695. [PubMed] [Google Scholar]

19. Jonas G, Johansson LM, Nissen Eastward, Ejdeback M, Ransjo-Arvidson AB, Uvnas-Moberg One thousand. Effects of intrapartum oxytocin administration and epidural analgesia on the concentration of plasma oxytocin and prolactin, in response to suckling during the second mean solar day postpartum. Breastfeed Med. 2009;4(2):71–82. [PubMed] [Google Scholar]

20. Kimura T, Saji F, Nishimori M, Ogita Grand, Nakamura H, Koyama K, et al. Molecular regulation of the oxytocin receptor in peripheral organs. J Mol Endocrinol. 2003;xxx(two):109–115. [PubMed] [Google Scholar]

21. Phaneuf S, Rodriguez Linares B, TambyRaja RL, MacKenzie IZ, Lopez Bernal A. Loss of myometrial oxytocin receptors during oxytocin-induced and oxytocin-augmented labour. J Reprod Fertil. 2000;120(i):91–97. [PubMed] [Google Scholar]

22. Heim C, Folder EB. Current research trends in early life stress and depression: Review of human studies on sensitive periods, gene-environment interactions, and epigenetics. Exp Neurol. 2012;233(1):102–111. [PubMed] [Google Scholar]

23. Szyf M, McGowan P, Meaney K. The social environment and the epigenome. Environ Mol Mutagen. 2008;49(one):46–60. [PubMed] [Google Scholar]

24. Kumsta R, Hummel Eastward, Chen FS, Heinrichs Yard. Epigenetic regulation of the oxytocin receptor factor: implications for behavioral neuroscience. Forepart Neurosci. 2013;7:83. [PMC gratis article] [PubMed] [Google Scholar]

25. Gregory SG, Connelly JJ, Towers AJ, Johnson J, Biscocho D, Markunas CA, et al. Genomic and epigenetic evidence for oxytocin receptor deficiency in autism. BMC Med. 2009;7:62. [PMC free article] [PubMed] [Google Scholar]

26. Connelly JJ, Kenkel West, Erickson East, Carter CS. Lodge for Neuroscience. Washington D.C.: 2011. Are birth and oxytocin epigenetic events? [Google Scholar]

27. Carter CS, Altemus 1000, Chrousos GP. Neuroendocrine and emotional changes in the postal service-partum period. Prog Brain Res. 2001;133:241–249. [PubMed] [Google Scholar]

28. Altemus 1000, Redwine LS, Leong YM, Frye CA, Porges SW, Carter CS. Responses to laboratory psychosocial stress in postpartum women. Psychosom Med. 2001;63(five):814–821. [PubMed] [Google Scholar]

29. O'Hara MW, McCabe JE. Postpartum depression: current status and future directions. Annu Rev Clin Psychol. 2013;9:379–407. [PubMed] [Google Scholar]

30. Shin H, Park YJ, Ryu H, Seomun GA. Maternal sensitivity: A concept analysis. J Adv Nurs. 2008;64(3):304–314. [PubMed] [Google Scholar]

31. Brook CT. The effects of postpartum depression on maternal-infant interaction: A meta-analysis. Nurs Res. 1995;44(v):298–304. [PubMed] [Google Scholar]

32. Kingston D, Tough Due south, Whitfield H. Prenatal and postpartum maternal psychological distress and babe evolution: a systematic review. Child Psychiatry Hum Dev. 2012;43(5):683–714. [PubMed] [Google Scholar]

33. Blom EA, Jansen PW, Verhulst FC, Hofman A, Raat H, Jaddoe VW, et al. Perinatal complications increase the risk of postpartum depression. The Generation R Study. BJOG. 2010;117(11):1390–1398. [PubMed] [Google Scholar]

34. Waldenstrom U, Hildingsson I, Rubertsson C, Radestad I. A negative nativity experience: Prevalence and adventure factors in a national sample. Nascence. 2004;31(1):17–27. [PubMed] [Google Scholar]

35. Sakala C, Corry M. Evidence-based maternity care: What information technology is and what can be accomplished. New York: Childbirth Connections, Reforming States Group, and Milbank Memorial Fund; 2008. [Google Scholar]

36. Bartz JA, Zaki J, Bolger N, Ochsner KN. Social effects of oxytocin in humans: context and person matter. Trends Cogn Sci. 2011;15(7):301–309. [PubMed] [Google Scholar]

37. Yan HC, Cao X, Das K, Zhu XH, Gao TM. Behavioral animal models of low. Neuroscience message. 2010;26(4):327–337. [PMC costless article] [PubMed] [Google Scholar]

38. Amico JA, Mantella RC, Vollmer RR, Li Ten. Anxiety and stress responses in female oxytocin scarce mice. J Neuroendocrinol. 2004;16(4):319–324. [PubMed] [Google Scholar]

39. Bosch OJ, Meddle SL, Beiderbeck DI, Douglas AJ, Neumann ID. Brain oxytocin correlates with maternal aggression: link to anxiety. J Neurosci. 2005;25(29):6807–6815. [PMC free commodity] [PubMed] [Google Scholar]

40. Cyranowski JM, Hofkens TL, Frank Eastward, Seltman H, Cai HM, Amico JA. Evidence of dysregulated peripheral oxytocin release amidst depressed women. Psychosom Med. 2008;70(9):967–975. [PMC costless article] [PubMed] [Google Scholar]

41. Skrundz Grand, Bolten 1000, Nast I, Hellhammer DH, Meinlschmidt G. Plasma oxytocin concentration during pregnancy is associated with development of postpartum depression. Neuropsychopharmacology. 2011;36(9):1886–1893. [PMC costless article] [PubMed] [Google Scholar]

42. Boccia ML, Goursaud AP, Bachevalier J, Anderson KD, Pedersen CA. Peripherally administered non-peptide oxytocin antagonist, L368,899, accumulates in limbic encephalon areas: a new pharmacological tool for the study of social motivation in non-homo primates. Horm Behav. 2007;52(3):344–351. [PMC complimentary article] [PubMed] [Google Scholar]

43. Keverne EB, Kendrick KM. Oxytocin facilitation of maternal behavior in sheep. Ann N Y Acad Sci. 1992;652:83–101. [PubMed] [Google Scholar]

44. Pedersen CA, Caldwell JD, Walker C, Ayers G, Mason GA. Oxytocin activates the postpartum onset of rat maternal beliefs in the ventral tegmental and medial preoptic areas. Behav Neurosci. 1994;108(half dozen):1163–1171. [PubMed] [Google Scholar]

45. Holman SD, Goy RW. Experiential and hormonal correlates of intendance-giving in rhesus macaques. In: Pryce CR, Martin RD, Skuse D, editors. Motherhood in Man and Nonhuman Primates: Biosocial Determinants. Switzerland: Karger; 1995. [Google Scholar]

46. Williams GL, Gazal OS, Leshin LS, Stanko RL, Anderson LL. Physiological regulation of maternal behavior in heifers: Roles of genital stimulation, intracerebral oxytocin release, and ovarian steroids. Biol Reprod. 2001;65(1):295–300. [PubMed] [Google Scholar]

47. Bales Chiliad, van Westerhuyzen J, Lewis-Reese A, Grotte N, Lanter J, Carter C. Oxytocin has dose-dependent developmental effects on pair-bonding and alloparental care in female prairie voles. Horm Behav. 2007;52(2):274–279. [PMC free article] [PubMed] [Google Scholar]

48. Hayes U, De Vries G. Role of pregnancy and parturition in induction of maternal behavior in prairie voles (Microtus ochrogaster) Horm Behav. 2007;51(2):265–272. [PMC gratis article] [PubMed] [Google Scholar]

49. Bakermans-Kranenburg MJ, van Ijzendoorn MH. Oxytocin receptor (OXTR) and serotonin transporter (v-HTT) genes associated with observed parenting. Soc Cogn Affect Neurosci. 2008;3(2):128–134. [PMC costless article] [PubMed] [Google Scholar]

l. Gordon I, Zagoory-Sharon O, Leckman JF, Feldman R. Oxytocin and the development of parenting in humans. Biol Psychiatry. 2010;68(iv):377–382. [PMC costless article] [PubMed] [Google Scholar]

51. Francis DD, Young LJ, Meaney MJ, Insel TR. Naturally occurring differences in maternal care are associated with the expression of oxytocin and vasopressin (V1a) receptors: Gender differences. J Neuroendocrinol. 2002;14(5):349–353. [PubMed] [Google Scholar]

52. Feldman R, Zagoory-Sharon O, Weisman O, Schneiderman I, Gordon I, Maoz R, et al. Sensitive parenting is associated with plasma oxytocin and polymorphisms in the OXTR and CD38 genes. Biol Psychiatry. 2012;72(iii):175–181. [PubMed] [Google Scholar]

53. Atzil South, Hendler T, Feldman R. Specifying the neurobiological basis of homo zipper: encephalon, hormones, and behavior in synchronous and intrusive mothers. Neuropsychopharmacology. 2011;36(13):2603–2615. [PMC free article] [PubMed] [Google Scholar]

54. Feldman R, Gordon I, Zagoory-Sharon O. Maternal and paternal plasma, salivary, and urinary oxytocin and parent-infant synchrony: considering stress and affiliation components of man bonding. Dev Sci. 2011;fourteen(four):752–761. [PubMed] [Google Scholar]

55. Wiklund I, Norman Thousand, Uvnas-Moberg K, Ransjo-Arvidson AB, Andolf E. Epidural analgesia: breast-feeding success and related factors. Midwifery. 2009;25(2):e31–e38. [PubMed] [Google Scholar]

56. Olza Fernandez I, Marin Gabriel Chiliad, Malalana Martinez A, Fernandez-Canadas Morillo A, Lopez Sanchez F, Costarelli V. Newborn feeding behaviour depressed by intrapartum oxytocin: a airplane pilot study. Acta Paediatr. 2012;101(7):749–754. [PubMed] [Google Scholar]

57. Rault JL, Carter CS, Garner JP, Marchant-Forde JN, Richert BT, Lay DC., Jr Repeated intranasal oxytocin administration in early life dysregulates the HPA axis and alters social beliefs. Physiol Behav. 2013;112–113:40–48. [PubMed] [Google Scholar]

58. Tsimis MS, Buckley AP, Nero D, Laurio A. Oxytocin usage for labor induction or augmentation and adverse neonatal outcomes. American Higher of Obstetricians and Gynecologists 61st Annual Clinical Meeting; New Orleans, LA. 2013. [Google Scholar]

59. Buchanan SL, Patterson JA, Roberts CL, Morris JM, Ford JB. Trends and morbidity associated with oxytocin use in labour in nulliparas at term. Aust North Z J Obstet Gynaecol. 2012;52(ii):173–178. [PubMed] [Google Scholar]

60. Kurth Fifty, Haussmann R. Perinatal Pitocin equally an early ADHD biomarker: neurodevelopmental gamble? J Atten Disord. 2011;xv(5):423–431. [PubMed] [Google Scholar]

61. Bell AF, White-Traut R, Rankin K. Fetal exposure to synthetic oxytocin and the relationship with prefeeding cues inside one hour postbirth. Early Hum Dev. 2013;89(3):137–143. [PubMed] [Google Scholar]

whitehichich.blogspot.com

Source: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3947469/

0 Response to "How to Deliver the Baby if Oxytocin Receptors Are Not Responding"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel