Abstract

Infertility affects a remarkable one in four couples in developing countries. Psychological stress is a ubiquitous facet of life, and although stress affects us all at some point, prolonged or unmanageable stress may become harmful for some individuals, negatively impacting on their health, including fertility. For instance, women who struggle to conceive are twice as likely to suffer from emotional distress than fertile women. Assisted reproductive technology treatments place an additional physical, emotional, and financial burden of stress, particularly on women, who are often exposed to invasive techniques associated with treatment. Stress-reduction interventions can reduce negative affect and in some cases to improve in vitro fertilization outcomes. Although it has been well-established that stress negatively affects fertility in animal models, human research remains inconsistent due to individual differences and methodological flaws. Attempts to isolate single causal links between stress and infertility have not yet been successful due to their multifaceted etiologies. In this review, we will discuss the current literature in the field of stress-induced reproductive dysfunction based on animal and human models, and introduce a recently unexplored link between stress and infertility, the gut-derived hormone, ghrelin. We also present evidence from recent seminal studies demonstrating that ghrelin has a principal role in the stress response and reward processing, as well as in regulating reproductive function, and that these roles are tightly interlinked. Collectively, these data support the hypothesis that stress may negatively impact upon fertility at least in part by stimulating a dysregulation in ghrelin signaling.

Essential Points
  • Infertility affects a remarkable one in four couples worldwide

  • Psychological stress is a major contributor to male and female infertility, at least for some individuals

  • For those individuals who are affected by life stress to a greater extent than others, understanding the underlying mechanisms and the reasons for their increased susceptibility is essential

  • We propose that ghrelin is a compelling link between stress and infertility that may partially explain the individual differences in the way stress affects fertility

Infertility, defined as the inability to achieve a viable pregnancy after 12 months of unprotected intercourse (6 months if the woman is over age 35), affects a remarkable one in four couples worldwide (1, 2). Female and male factors both contribute to infertility, with 30% to 50% attributable to male factors, such as low sperm count, poor sperm quality, hypogonadism, and other abnormalities. Physiological causes of female infertility include ovulation disorders and tubal damage (~50%), as well as 10% to 30% of all cases remaining unexplained (35). Recent data, particularly from animal models (including nonhuman primates) suggest psychological stress may be a major contributor to both male and female infertility (612). Thus, stress can compromise every aspect of fertility, including libido, sperm quality, ovulatory capacity, and implantation (1315) (see Fig. 1). Indeed, stress can be so detrimental to fertility that in at least one species, it is used as a deliberate strategy to suppress fecundity in competitors (16).

Stress influences reproductive function at all levels. Stress can interfere with reproductive function at all levels of the reproductive axis. It can suppress libido, reward, and mating behavior at the level of the brain, particularly the ventral tegmental area. It interferes with the hypothalamic GnRH pulse generator and LH and FSH release from the anterior pituitary. It suppresses oocyte maturation, ovulation at the level of the gonads, as well as increasing the likelihood of ovarian cysts and affecting both ovarian and testicular steroidogenesis. Stress is also detrimental to pregnancy outcomes postconception, reducing the likelihood of successful blastocyst implantation. Adapted from Servier Medical Art under Creative Commons CC-BY license.
Figure 1.

Stress influences reproductive function at all levels. Stress can interfere with reproductive function at all levels of the reproductive axis. It can suppress libido, reward, and mating behavior at the level of the brain, particularly the ventral tegmental area. It interferes with the hypothalamic GnRH pulse generator and LH and FSH release from the anterior pituitary. It suppresses oocyte maturation, ovulation at the level of the gonads, as well as increasing the likelihood of ovarian cysts and affecting both ovarian and testicular steroidogenesis. Stress is also detrimental to pregnancy outcomes postconception, reducing the likelihood of successful blastocyst implantation. Adapted from Servier Medical Art under Creative Commons CC-BY license.

Although the link between stress and reproductive dysfunction has been well-established and extensively investigated in nonhuman animals, where additional variables can be tightly controlled, the question of whether stress directly impedes fertility in humans remains difficult to conclusively examine (1720). Psychological stress experienced by female or male partners of infertile couples has been associated with lower conception rates in at least some cases (2124). However, there have been a number of methodological challenges and conflicting findings facing research in the area. These include the lack of prospective longitudinal studies on the general population (18), the use of nonstandardized measures of stress, and the fact that the majority of human studies have been conducted on couples attending fertility clinics (18, 25), with, likely, underlying physical and/or hormonal causes of infertility. There is also the important consideration that stress is a normal facet of life, thereby making nonstressed control groups very difficult to source. Stress is usually dealt with by an appropriate and regulated neuroendocrine response, but although psychological stress affects us all at some point, individuals differ in the impact this has on their physiology, including fertility. These individual differences may explain why some studies have demonstrated substantial reproductive implications of psychological distress (13, 15, 23, 24, 2631), whereas others have found small effect sizes or no association (3238). It is likely that individuals experiencing extreme or long-term stress or those more vulnerable to its effects may be particularly vulnerable to fertility consequences.

Infertility itself can be highly stressful. Many of those who struggle with infertility seek help from assisted reproductive technology treatments. These treatments, including in vitro fertilization, can involve invasive techniques that place an additional burden of stress on the couple in addition to the stress associated with an inability to conceive (39). The effects of stress may therefore be particularly detrimental for couples undergoing assisted reproductive technology, leading in some couples to more treatment cycles to conceive (28, 3941), resulting in discontinuation of treatment before achieving pregnancy (4244), and potentially contributing to the low overall success rates (45). Similarly, pre-existing psychological conditions, such as anxiety, depression, and high levels of distress, can have a negative effect on assisted reproductive technology outcomes (24, 28, 4650) [but see (5154)]. For instance, Smeenk et al. (48) show anxiety and depression were significantly negatively correlated with pregnancy outcome in a multicenter prospective study in 2001. The same group was not able to replicate this finding in a separate study in 2009 (36). However, patients who are prone to anxiety, depression, or high stress levels may particularly benefit from psychosocial support during the treatments.

Some human evidence suggests intervention to reduce stress may improve the chances of successful pregnancy. A meta-analysis of 21 studies reviewing the efficacy of psychological interventions for infertile patients, from which 13 studies reported pregnancy outcomes, shows a significant positive impact of interventions, including counseling, cognitive-behavioral therapy, mind or body-oriented relaxation, education, psychodynamic or psychoanalytic therapy on pregnancy rates, despite these interventions having no measurable effect on mental health. These improved pregnancy success rates were only evident for couples not receiving assisted reproductive technology treatments (55), highlighting that many factors additional to stress contribute to infertility. Another meta-analysis of 22 studies has also indicated a positive effect of psychotherapy on conception. In this analysis, 45% of subjects in the intervention group reported pregnancy by study completion, compared with 14% of controls, with similar rates of pregnancy achieved in patients receiving assisted reproductive technology treatments and those who were not in specific medical care (56).

As a consequence of these studies, attention is given to stress reduction management in individuals and couples undergoing fertility treatments. The Australian Assisted Reproductive Treatment Act (2008) stipulates a mandatory pretreatment counseling session that includes a discussion of areas of potential stress and strategies for managing these (57). Similar guidelines are implemented in other countries (58). We should note that other studies have shown that short-term attempts to reduce stress levels may have limited or no effect on pregnancy outcomes (35, 59, 60), highlighting the complexity and multifaceted origins of infertility in humans. It will be useful to see large-scale long-term stress intervention studies in infertile humans with accompanying measures of stress perception and circulating stress hormones.

Given the complexity of the interaction between the stress and reproductive axes, links between stress, stress-reduction approaches, and successful conception in humans are inconsistent. Likewise, attempts to isolate single causal links between stress and infertility have not yet been successful. However, stress management has important potential to improve reproductive success rates if we can identify the correct strategies for those people most likely to benefit. It is therefore essential that we begin to identify why fertility may be markedly affected by stress in some individuals, and less so in others. In this review, we discuss the current literature in the field of stress-induced reproductive dysfunction and the multifaceted nature of this interaction, based on nonhuman animal models and human studies. Here we will particularly focus on an important gut hormone, ghrelin, as a compelling link between stress and infertility.

Stress and the Hypothalamic-Pituitary-Adrenal Axis

“Stress” itself has been a controversial concept since its first description in physiology by Cannon and Selye early last century (6165). However, a stressor (the stimulus) can broadly be considered an intense, nonroutine challenge to homeostasis resulting in a nonspecific response that includes general activation of the hypothalamic-pituitary-adrenal (HPA) axis and sympathomedullary systems. Each stressor can also activate a more specific signature response. Here we will refer to “stress” as the stimulus and “the stress response” as the body’s reaction to the stimulus. Although there are myriad types of stress, they can be loosely categorized into “physical” and “psychological” stress based on the immediacy of the impact on the body and the endocrine and neuronal responses they elicit. Physical stressors (interoceptive, homeostatic, systemic) are those involving an immediate disturbance of tissue integrity and a specific activation of the central amygdala and rostral A1 and A2 brainstem noradrenergic cells in addition to general HPA axis and sympathomedullary activation. Psychological stress (neurogenic, psychogenic, emotional) involves a threat to tissue disturbance rather than a direct injury and a specific medial amygdala and caudal A1 and A2 brainstem noradrenergic pattern of neuronal activation (6669).

Acutely, within seconds to minutes, both physical and psychological stress generally activates the sympathomedullary system leading to the release of adrenaline and noradrenaline, which increase heart rate, blood pressure, respiration, and blood glucose levels to facilitate attention and action directed at combatting the stress. Within minutes the HPA axis is activated, with corticotropin-releasing hormone (CRH)- and arginine vasopressin-expressing cells in the medial parvocellular region of the paraventricular nucleus of the hypothalamus stimulating the release of CRH from axonal terminal boutons in the median eminence, which, in turn, stimulates corticotrophs in the anterior pituitary gland to release adrenocorticotropic hormone (ACTH) into systemic circulation. By approximately 20 to 30 minutes after the onset of the stress (70, 71), ACTH is acting at the melanocortin-2 receptors on the adrenal cortex to stimulate the synthesis and release of glucocorticoids into the circulation. Glucocorticoids remain elevated for ~60 to 120 minutes after the onset of an acute stressor (70, 72) and have roles in immunosuppression, glucose uptake and mobilization, fat storage, and memory consolidation, among others. Glucocorticoids also negatively feed back onto glucocorticoid and mineralocorticoid receptors, chiefly in the hippocampus and hypothalamus, to inhibit further activation of the paraventricular nucleus of the hypothalamus (PVN) so suppressing ongoing activation of the HPA axis once the stressor has been resolved (7376) (see Fig. 2).

The HPA axis. Upon encountering a stressor, medial parvocellular CRH and arginine vasopressin (AVP) neurons of the hypothalamus receive input from several brain regions, including the amygdala, brainstem, and prefrontal cortex, and stimulate CRH release into the median eminence. CRH acts on the anterior pituitary through CRH receptor type 1 (CRH-R1), leading to the release of ACTH into circulation, a peptide hormone derived from pro-opiomelanocortin (POMC). ACTH stimulates glucocorticoid release to regulate glucose and fat utilization and storage, memory, immune function, and other stress-coping strategies, as well as negatively feeding back on glucocorticoid and mineralocorticoid receptors in the hypothalamus and hippocampus to suppress further HPA axis activation. Adapted from Servier Medical Art under Creative Commons CC-BY license.
Figure 2.

The HPA axis. Upon encountering a stressor, medial parvocellular CRH and arginine vasopressin (AVP) neurons of the hypothalamus receive input from several brain regions, including the amygdala, brainstem, and prefrontal cortex, and stimulate CRH release into the median eminence. CRH acts on the anterior pituitary through CRH receptor type 1 (CRH-R1), leading to the release of ACTH into circulation, a peptide hormone derived from pro-opiomelanocortin (POMC). ACTH stimulates glucocorticoid release to regulate glucose and fat utilization and storage, memory, immune function, and other stress-coping strategies, as well as negatively feeding back on glucocorticoid and mineralocorticoid receptors in the hypothalamus and hippocampus to suppress further HPA axis activation. Adapted from Servier Medical Art under Creative Commons CC-BY license.

The response to acute stress is typically a highly adaptive phenomenon, enabling the individual to appropriately combat the stressor and recover (7376). However, if a stressor becomes chronic, due to prolonged infection or exposure to substantial life challenges, such as infertility, negative complications can ensue. Chronically elevated glucocorticoids can lead to a variety of complications, including excess weight gain, memory impairments, and mood disorders, and as we will discuss, this also has consequences for fertility.

As outlined in the introduction, although much controversy exists in human research examining the effects of stress on fertility, causal relationships have been demonstrated in animal models. Thus, chronic unpredictable stress in mice has been shown to impair oocyte developmental potential through severe apoptosis and oxidative stress (79). In our own work, we have seen that exposure to psychological stress has an inhibitory effect on copulatory behavior and suppresses the hormonal surge during mating in female rats (6). We have also shown that animals that are programmed to be hyperresponsive to stress demonstrate impaired sexual development and detrimental changes to their ovarian or testicular gametogenesis (6, 77), indicating stress and the HPA axis interact significantly with the reproductive axes.

Stress and the Hypothalamic-Pituitary-Gonadal Axis

Reproduction is an essential function for the perpetuation of the species and, as such, is controlled by a sophisticated regulatory network of neuroendocrine signals that are originated and integrated by the hypothalamic-pituitary-gonadal (HPG) axis. The HPG hormonal cascade begins in the medial preoptic area (mPOA) of the hypothalamus, with the release of gonadotropin-releasing hormone (GnRH) from GnRH neurons. This collection of neurons, known as the GnRH pulse generator, sends axons to the median eminence from where GnRH is released in a synchronized pulsatile manner and is the central control mechanism for the reproductive cycle (7880). This pulsatile GnRH release is driven by several specific mechanisms, including calcium and cyclic adenosine monophosphate signaling (81), electrical activity of GnRH neurons (82), autocrine regulation of the GnRH receptor (83), coupling of the GnRH receptor to G-related proteins (84), expression of G protein-coupled receptor 54 and its endogenous ligand, kisspeptin (85), as well as positive and negative regulation by gonadal steroids (86). The GnRH peptide is secreted from the nerve endings into the hypophyseal portal system, to stimulate the synthesis and the release of the gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), from the anterior pituitary (87). FSH and LH are released in pulses into the blood stream to stimulate the gonadal production of gametes and the release of sex steroids, including estrogen, progesterone, and testosterone (88). The levels of each of the HPG axis hormones are regulated by complex positive and negative feedback loops, and are also influenced by other neuroendocrine signals (see Fig. 3). As such, the HPA and the HPG axes are well known to coregulate one another both centrally and peripherally, with the extent of stress (acute vs chronic), species, sex, and individual differences in resilience affecting the ability of stress to influence reproduction (8991) (see Fig. 4). One of the major structures affected by stress is the hypothalamic GnRH pulse generator, with stress diminishing its ability to stimulate pulsatile release of gonadotropins. This loss of pulsatility may lead to hypothalamic amenorrhea (92), and can be accompanied by reproductive dysregulations associated with other targets along the HPG axis.

The HPG axis. Hypothalamic mPOA kisspeptin (Kiss1) neurons stimulate and gonadotropin inhibitory hormone (GnIH) neurons inhibit GnRH pulsatile release into the median eminence. GnRH release stimulates the release of gonadotropins LH and FSH from the anterior pituitary. This in turn leads to the release of ovarian steroids mainly estrogen and progesterone, that can carry out both negative- and positive-feedback actions depending on the stage of the ovarian cycle. In males, gonadal release of testosterone produces inhibitory actions on GnRH/gonadotropin secretion (negative feedback). Pituitary gonadotropins also stimulate steroidogenesis. Adapted from Servier Medical Art under Creative Commons CC-BY license.
Figure 3.

The HPG axis. Hypothalamic mPOA kisspeptin (Kiss1) neurons stimulate and gonadotropin inhibitory hormone (GnIH) neurons inhibit GnRH pulsatile release into the median eminence. GnRH release stimulates the release of gonadotropins LH and FSH from the anterior pituitary. This in turn leads to the release of ovarian steroids mainly estrogen and progesterone, that can carry out both negative- and positive-feedback actions depending on the stage of the ovarian cycle. In males, gonadal release of testosterone produces inhibitory actions on GnRH/gonadotropin secretion (negative feedback). Pituitary gonadotropins also stimulate steroidogenesis. Adapted from Servier Medical Art under Creative Commons CC-BY license.

Intersection of HPA and HPG axes. CRH can directly inhibit the GnRH pulse generator leading to suppression of pituitary gonadotropins (LH and FSH), in turn resulting in diminished oocyte maturation and steroidogenesis. Glucocorticoids can act directly at the level of the pituitary to suppress gonadotropin release, as well as to exert suppressive effects within the gonads. CRH release from the paraventricular nucleus of the hypothalamus activates locus coeruleus (LC) neurons increasing noradrenaline production. This stress-induced increase in sympathetic activity can then increase sympathetic innervation of the ovary, contributing to the development of ovarian cysts. Adapted from Servier Medical Art under Creative Commons CC-BY license.
Figure 4.

Intersection of HPA and HPG axes. CRH can directly inhibit the GnRH pulse generator leading to suppression of pituitary gonadotropins (LH and FSH), in turn resulting in diminished oocyte maturation and steroidogenesis. Glucocorticoids can act directly at the level of the pituitary to suppress gonadotropin release, as well as to exert suppressive effects within the gonads. CRH release from the paraventricular nucleus of the hypothalamus activates locus coeruleus (LC) neurons increasing noradrenaline production. This stress-induced increase in sympathetic activity can then increase sympathetic innervation of the ovary, contributing to the development of ovarian cysts. Adapted from Servier Medical Art under Creative Commons CC-BY license.

The effects of stress on hypothalamic GnRH neuronal signaling

The role of CRH in stress-induced suppression of GnRH signaling

The inhibitory effects of stress are initiated by CRH-mediated suppression of the GnRH pulse generator, diminishing the subsequent pituitary release of gonadotropins in rats (93), sheep (91, 94), nonhuman primates (95), and humans (96). These inhibitory effects of CRH can be, at least partially, reversed by CRH antagonists as has been shown in rats (9799) and nonhuman primates in response to a mild psychological and metabolic (100), as well as an inflammatory stress (101), although inconsistencies are seen in the primate research, with other studies demonstrating that CRH receptor antagonism paradoxically increases cortisol release, inducing further LH release suppression (12).

The CRH family of neuropeptides integrates the neuroendocrine stress responses in the brain through two distinct receptor subtypes, CRH receptor type 1 (CRH-R1) and type 2 (CRH-R2), with higher binding affinity of CRH to CRH-R1 (102). CRH-R1 is highly expressed in the brain and pituitary and to a lesser extent in peripheral tissues (102, 103). Although the actions of CRH to drive HPA axis activation during stress are primarily mediated by CRH-R1 (104), stress-induced suppression of the GnRH pulse generator is mediated by both CRH-R1 and CRH-R2 (98, 99, 105).

The vulnerability of the GnRH pulse generator to stress has been suggested to be influenced by CRH-GnRH connectivity in the mPOA, a GnRH-rich area. The existence of synaptic connectivity between CRH and GnRH neurons (106) and expression of CRH-R1 receptors in mouse GnRH neurons in this area (107) indicate the possibility of direct actions of CRH on the mPOA GnRH system. In contrast, although the PVN plays a central role in autonomic and neuroendocrine regulation of stress responsiveness (108), PVN CRH neurons do not appear to directly coordinate stress-induced reproductive dysfunction. Lesions of the PVN are unable to block stress-induced suppression of gonadotropin release (109), and no direct connectivity between PVN CRH and GnRH neurons has been detected (110). Furthermore, infusion of calcitonin gene-related peptide, which is known for its role in stress-induced suppression of the HPG axis (111), suppresses LH pulse frequency when infused directly into the mPOA, but not into the PVN (112). As we will discuss in the Ghrelin Signaling in the Stress Response, Reward, and Mood Disorders section, ghrelin can regulate the expression of CRH in stress-sensitive areas including hypothalamus (113) and ghrelin, administrated both centrally and peripherally, mediates its anxiogenic effects via the PVN CRH neurons (114, 115). However, because the inhibitory effects of ghrelin on central reproductive function have been shown in other regions, such as the mPOA (116, 117), it is possible that ghrelin also acts via the activation of CRH neurons in these areas to suppress LH pulsatility.

Additional regulatory mechanisms may influence stress-induced GnRH pulse frequency. Other major stress-regulatory areas, such as the amygdala, particularly the medial and central nuclei, have been implicated in stress-induced suppression of the GnRH pulse generator (118120), with ghrelin, at least indirectly, also influencing these regions during stress (113, 121).

The role of γ-aminobutyric acid in stress-induced suppression of GnRH signaling

In addition to CRH, the inhibitory effect of stress on the GnRH pulse generator activity is likely to be mediated via γ-aminobutyric acid (GABA)-ergic signaling. Most studies have reported inhibitory effects of GABA on GnRH/LH pulsatility (122125), with the inhibitory actions of GABA on GnRH neurons emerging at the time of vaginal opening in the mouse, demonstrating a switch from their postnatal depolarizing profile (126). These inhibitory effects of GABA-ergic signaling have been proposed to be mediated by GABAA receptor activation (122, 124, 125). In female nonhuman primates, blockade of GABAA receptor increases GnRH release and accelerates the onset of puberty (127). In regards to the effects of stress, GABA-ergic signaling is modulated by central administration of CRH (128), by different stressors (120, 129), and by ghrelin (114). GABAA and GABAB receptors in the mPOA are differentially involved in mediating the effects of stress on LH pulsatility, and antagonism of both receptors has been shown to block the CRH-induced inhibition of LH release in rats (130, 131). At the level of the median eminence GABA can act on the GnRH nerve terminals, leading to disruption of estrous cyclicity in rats (123).

Kisspeptin and stress-induced suppression of GnRH signaling

Another key regulator of GnRH signaling that can be influenced by stress and ghrelin is kisspeptin. Kisspeptin is a neuropeptide that regulates fertility by conveying information on systemic levels of sex steroids to GnRH neurons, and thus regulates both tonic and pulsatile GnRH release, playing a critical role in the onset of puberty (85, 132135). The most abundant populations of kisspeptin-expressing neurons are found in the arcuate nucleus of the hypothalamus and preoptic area, particularly the anteroventral periventricular nucleus (134, 136). Kisspeptin neurons are also located in the PVN, and project to limbic structures (137, 138), which are involved in stress-induced suppression of the GnRH pulse generator (120, 128). Different stress paradigms that have been shown to suppress GnRH release also result in downregulation of hypothalamic expression of the kisspeptin gene (Kiss1) and its receptor (Kiss1r) in the mPOA and the arcuate nucleus (139, 140), suggesting that kisspeptin may play a critical role in the stress-induced suppression of GnRH pulsatility and LH release. Similar to stress, ghrelin also has an inhibitory effect on kisspeptin-mediated GnRH pulsatility and LH release (116, 141), as we will discuss in the Ghrelin’s Local and Systemic Role in Hypothalamic-Pituitary Reproductive Control section.

The role of gonadotropin-inhibitory hormone in stress-induced suppression of GnRH signaling

A negative regulator of the HPG axis that has also been implicated in stress-induced reduction of GnRH activity is the gonadotropin-inhibitory hormone, also known as RFamide-related peptide in rodents and humans (142). In rats, exposure to stress leads to hypothalamic upregulation of RFamide-related peptide expression, and this increase is associated with a significant reduction in circulating LH (143). Another recent study has demonstrated that chronic stress-induced reproductive dysfunction in rats is completely resolved by targeted knockdown of hypothalamic RFamide-related peptide-3 (144).

The effects of ghrelin on the activity of RFamide-related peptide-containing neurons have not yet been fully characterized (145). RFamide-related peptide neurons do not express ghrelin receptors, suggesting any effects of ghrelin are indirect or via a yet unidentified receptor (146). RFamide-related peptide neurons, however, coexpress CRH-R1 and glucocorticoid receptors (143), and CRH increases RFamide-related peptide receptor Gpr147 gene expression and suppresses GnRH mRNA levels in GnRH cells in vitro (147), whereas adrenalectomy abolishes the effects of stress on RFamide-related peptide/GnRH signaling (143). Gpr147 is also expressed by kisspeptin neurons (148), and RFamide-related peptide inhibits kisspeptin-induced activation of GnRH neurons (149). These findings suggest that gonadotropin-inhibitory hormone acts directly and indirectly on the GnRH network to mediate the inhibitory effects of stress on reproduction. It is also evident that due to the extreme complexity of the neural networks involved in the control of reproduction, the multiple signaling mechanisms reviewed previously work in conjunction to mediate the effects of stress on fertility.

Stress and pituitary and gonadal function

Glucocorticoids and the regulation of pituitary gonadotropins

Although stress-induced decreases in gonadotropin release are primarily controlled centrally and are influenced by changes to the activity of the hypothalamic GnRH pulse generator, direct subtle changes may also occur locally. Early studies, in several species including humans, demonstrated that glucocorticoids act directly on the pituitary to inhibit its responsiveness to GnRH and reduce the release of gonadotropins in vivo and in vitro (150154). More recent evidence suggests inhibition of the pituitary’s sensitivity to GnRH may also be regulated directly at the pituitary by ghrelin (113, 155, 156), as we will discuss further in the Ghrelin’s Local and Systemic Role in Hypothalamic-Pituitary Reproductive Control section. The effects of glucocorticoids at the pituitary are possibly divergent for LH and FSH, because in vitro release of FSH is increased from rat pituitary cells when treated with glucocorticoids, but LH production is suppressed (157). The potential for direct effects of glucocorticoids on the pituitary are further supported by the evidence that glucocorticoid receptors are expressed by rat (158) and mouse (159) gonadotrophs, and glucocorticoids can act directly upon the anterior pituitary gonadotrophs to suppress GnRH-induced LH subunit β gene expression (159). Synthesis of the β-subunit is the rate-limiting step in LH production (160). Consistent with a direct action of glucocorticoids at the level of the pituitary, studies in ovariectomized ewes have demonstrated that cortisol treatment induces suppression of LH, and this suppression occurs in the absence of a reduction in GnRH pulsatility (161), whereas GR antagonism reverses cortisol- and psychosocial stress-induced suppression in pituitary responsiveness to GnRH (162, 163). Overall, these findings suggest that glucocorticoid-induced suppression of pulsatile LH release may be driven by a reduction of pituitary responsiveness to GnRH. However, the full extent of glucocorticoid (and ghrelin’s) role in compromising the GnRH-driven FSH and LH surges may be poorly estimated due to the technical difficulties in sampling blood sufficiently frequently to obtain accurate surge profiles, particularly in small animal models. Sheep, nonhuman primate, and human studies will prove to be particularly informative in this regard. Another important pituitary hormone known to be influenced by stress and ghrelin is prolactin, and we discuss its role in mediating the effects of stress and ghrelin on reproductive function in Ghrelin as a Regulator of the HPG Axis section.

The effects of glucocorticoids on gonadal function

Exposure to stress is usually accompanied by decreased gonadal steroid production (89, 164). This decreased steroid production may be a result of a stress-induced attenuation of the pituitary reproductive hormone release affecting the gonadal output (165167), as well as by a direct effect of glucocorticoids and sympathetic innervation on the gonadal activity (168, 169), as discussed here and in the the Effects of Catecholamines on Gonadal Function section.

Several studies in humans have demonstrated that increased levels of glucocorticoids due to an exposure to stress can reduce the release of gonadal steroids, with and without a concomitant reduction in plasma LH levels (164, 170172). Bereavement stress (173), as well as experiences of war (174, 175) have been associated with reduced quality of sperm. However, these effects are likely to be related to the perceived degree of stress. As such, there are mixed findings regarding the impact of chronic mild stress, such as job strain, on semen quality. Although some studies have found no association between work-related stress and semen quality or sex steroids (173, 176, 177), others found that higher job strain does lead to significant impairment of sperm quality (178, 179) and decreased levels of testosterone (180). These discrepancies suggest that although occupational stress may be detrimental to male fertility, different measures of job strain such as effort-reward imbalance need to be considered to determine whether work stress has necessarily a negative impact on wellbeing (181). Particularly because job loss and unemployment are also associated with poor sperm quality (177) and lower testosterone (182), these data suggest these effects are likely to be related to increased anxiety and depression, independently linked to poor fertility (22, 171).

The impact of stress on testosterone levels in adult males may be influenced by exposure to early life stress, and thus program increased vulnerability to the effects of adult stress (183). In the testis, glucocorticoids act directly on Leydig cells that express glucocorticoid receptors (184). The access of glucocorticoids to their receptors is controlled by the type 1 isoform of 11β-hydroxysteroid dehydrogenase. In male gonads, this enzyme is exclusively and abundantly expressed in Leydig cells where it catalyses the oxidative inactivation of glucocorticoids. However, in response to severe stress, 11β-hydroxysteroid dehydrogenase1 is saturated and excessive glucocorticoids may cause rapid repression of testosterone production [reviewed in (185)]. Ghrelin has also been shown to modulate testicular function both directly at the testicular level and through its systemic administration, suppressing Sertoli and Leydig cell proliferation (186, 187).

A direct effect of glucocorticoids on the ovary has also been identified. Glucocorticoid receptors are expressed in different cell types within the ovary (188), and their expression is maintained during follicular maturation, ovulation, and pregnancy (189). Similar to glucocorticoid action in the testis, 11β-hydroxysteroid dehydrogenase regulates glucocorticoid action within the ovary. In cultured human granulosa-lutein cells, diminished 11β-hydroxysteroid dehydrogenase activity has been shown to mediate cortisol-induced inhibition of ovarian steroidogenesis (190). Ovarian steroidogenesis is also suppressed locally by ghrelin (191), where it inhibits the expression of other steroid pathway enzymes, such as 3β-hydroxysteroid dehydrogenase, 17β-hydroxysteroid dehydrogenase, and cytochrome P450 aromatase (192).

Glucocorticoids have also been implicated in oocyte maturation along the ovulatory cycle and species specific effects have been noted. Glucocorticoids suppress meiotic maturation in gilt oocytes (193). In contrast, in mouse oocytes, only supraphysiological levels of glucocorticoids inhibit follicle differentiation and oocyte maturation (194). In ewes, inconsistent effects of cortisol and dexamethasone on oocyte maturation have been demonstrated, with no effect of these glucocorticoids on the capacity of the oocytes to undergo fertilization (195). In humans, however, higher levels of cortisol have been detected in the follicular fluid of oocytes that were not fertilized than in the follicular fluid of successfully fertilized oocytes (196), and higher ovarian 11β-hydroxysteroid dehydrogenase 1 activity has been correlated with better fertilization rate (197).

The effects of catecholamines on gonadal function

In addition to the local ovarian effects of glucocorticoids, catecholamines, and in particular noradrenaline, have an important role in follicular maturation and steroidogenesis (198200). β-adrenergic receptors are expressed by the theca-interna ovarian cells, and activation of these receptors leads to increased androgen production (201). A stress-induced increase in sympathetic activity, such as that induced by cold and restraint stress, can increase sympathetic innervation of the ovary, contributing to the development of ovarian cysts in rodents (202, 203), and these effects can be abolished by lesions to the noradrenergic nucleus locus coeruleus (204). Polycystic ovarian morphology and excessive androgen levels are common features of polycystic ovarian syndrome (PCOS). As we will discuss in the Role of Ghrelin Signaling in PCOS section, dysregulation in ghrelin signaling has also been implicated in the pathophysiology of PCOS.

Ghrelin and Its Receptors: Structure, Distribution, and Function

Ghrelin is a gut-derived hormone that was originally associated with feeding behavior and energy homeostasis (205, 206). It has lately been found to have important roles in such diverse biological functions as motivation, memory, vascular function, and neuroprotection after brain injury (207211). Ghrelin is also a key regulator of the endocrine response to stress (212) and of reproduction (155, 191, 213222).

Ghrelin is a 28-amino acid peptide (223). It is principally produced in the stomach as proghrelin where it undergoes posttranslational octanoylation by the enzyme ghrelin-O-acyltransferase (GOAT) to form acylated ghrelin (AG) (224, 225). Ghrelin thus exists in the circulation in at least two major bioactive forms: AG and des-acylated ghrelin (DAG), the unacylated form. There is some evidence that ghrelin is expressed in tissues other than the gut, including pancreas and kidney, pituitary, ovaries, and testes (226229). However, data from studies in mice with the ghrelin reporter tagged to green fluorescent protein (230, 231) suggest that ghrelin in the brain is likely derived from circulating ghrelin having passed through the blood-brain barrier (232) rather than being specifically synthesized in ghrelin-containing neurons (212).

AG was first identified in 1999 as the endogenous ligand for the growth hormone secretagogue receptor (GHSR1a), through which it stimulates growth hormone release (223). GHSR1a is expressed in numerous tissues throughout the body, including the ovaries and testes (228, 229, 233) (see Table 1). It is also expressed in brain regions important in regulating the HPA and HPG axes, and in regions directly involved in these axes, including hypothalamus and pituitary (113, 231, 234). GHSR1a is a G protein-coupled receptor and activation by the endogenous ligand stimulates a phospholipase C/protein kinase C/inositol trisphosphate pathway that triggers inositol trisphosphate-dependent calcium release from intracellular stores. This intracellular calcium combines with calcium entering the cell via voltage-gated l-type calcium channels to stimulate the downstream response (242244). In addition to its ligand-dependent effects, GHSR1a has high constitutive activity, and can signal at approximately 50% of its maximal capacity in the absence of ghrelin, at least in vitro (245). GHSR1a is also able to dimerize with other receptors, such as the dopamine receptor subtype 2 to modulate dopamine signaling (246) and the melanocortin-3 receptor to modulate melanocortin signaling (247, 248).

Table 1.

Distribution of GHSR in Rat and Mouse Hypothalamus and Pituitary

OrganAbbreviationRatMouse
Hypothalamus
 Anterior hypothalamic areaAHA+
 Anteroventral periventricular nucleusAVPe+++
 Arcuate nucleusARC++++++++
 Dorsomedial nucleusDMH+++
 Medial preoptic nucleusMPOA+
 Paraventricular nucleusPVH+++
 Periventricular hypothalamic nucleusPe+
 Retrochiasmatic areaRCA++
 Suprachiasmatic nucleusSCh+++++
 Ventromedial nucleusVMH+++
Pituitary gland
 Anterior pituitary
  Corticotrophs+++
  Gonadotrophs+
  Lactotrophs++
  Somatotrophs++++
 Posterior pituitary
OrganAbbreviationRatMouse
Hypothalamus
 Anterior hypothalamic areaAHA+
 Anteroventral periventricular nucleusAVPe+++
 Arcuate nucleusARC++++++++
 Dorsomedial nucleusDMH+++
 Medial preoptic nucleusMPOA+
 Paraventricular nucleusPVH+++
 Periventricular hypothalamic nucleusPe+
 Retrochiasmatic areaRCA++
 Suprachiasmatic nucleusSCh+++++
 Ventromedial nucleusVMH+++
Pituitary gland
 Anterior pituitary
  Corticotrophs+++
  Gonadotrophs+
  Lactotrophs++
  Somatotrophs++++
 Posterior pituitary

Hypothalamic data are adapted from (234). Pituitary data are adapted from (235). Hypothalamic and pituitary GHSR expression has been identified in rodents (234236), sheep (237), and human (238), as well as nonmammalian species including birds (239) and fish (240, 241).

Symbols in this table are not comparable between organs.

Abbreviations: −, no detectable expression; +, minimal detectable expression; ++, some detectable expression; +++, high expression; ++++, very high expression.

Table 1.

Distribution of GHSR in Rat and Mouse Hypothalamus and Pituitary

OrganAbbreviationRatMouse
Hypothalamus
 Anterior hypothalamic areaAHA+
 Anteroventral periventricular nucleusAVPe+++
 Arcuate nucleusARC++++++++
 Dorsomedial nucleusDMH+++
 Medial preoptic nucleusMPOA+
 Paraventricular nucleusPVH+++
 Periventricular hypothalamic nucleusPe+
 Retrochiasmatic areaRCA++
 Suprachiasmatic nucleusSCh+++++
 Ventromedial nucleusVMH+++
Pituitary gland
 Anterior pituitary
  Corticotrophs+++
  Gonadotrophs+
  Lactotrophs++
  Somatotrophs++++
 Posterior pituitary
OrganAbbreviationRatMouse
Hypothalamus
 Anterior hypothalamic areaAHA+
 Anteroventral periventricular nucleusAVPe+++
 Arcuate nucleusARC++++++++
 Dorsomedial nucleusDMH+++
 Medial preoptic nucleusMPOA+
 Paraventricular nucleusPVH+++
 Periventricular hypothalamic nucleusPe+
 Retrochiasmatic areaRCA++
 Suprachiasmatic nucleusSCh+++++
 Ventromedial nucleusVMH+++
Pituitary gland
 Anterior pituitary
  Corticotrophs+++
  Gonadotrophs+
  Lactotrophs++
  Somatotrophs++++
 Posterior pituitary

Hypothalamic data are adapted from (234). Pituitary data are adapted from (235). Hypothalamic and pituitary GHSR expression has been identified in rodents (234236), sheep (237), and human (238), as well as nonmammalian species including birds (239) and fish (240, 241).

Symbols in this table are not comparable between organs.

Abbreviations: −, no detectable expression; +, minimal detectable expression; ++, some detectable expression; +++, high expression; ++++, very high expression.

Although AG’s interaction with GHSR1a has been well-characterized, the receptor for the more abundant form of circulating ghrelin, DAG (224), is currently unknown. DAG does not act at the GHSR. DAG is known to inhibit the effects of AG (249), but it also has important independent physiological effects, including in stress (250, 251) and potentially in fertility.

Ghrelin Signaling in the Stress Response, Reward, and Mood Disorders

Ghrelin in stress and mood, evidence from clinical studies

In a clinical setting, ghrelin is closely affected by stress, mood, and stress-related disorders, such as anxiety and depression. Women with high levels of interpersonal stress have higher serum ghrelin levels than their less-stressed counterparts (252), and ghrelin is acutely elevated after stress imposed in an experimental setting (253255), or even an anticipation of stress (256). Interestingly, AG is elevated by stress in direct correspondence to the magnitude of stress, so that people with greater glucocorticoid responses to stress have also higher ghrelin levels (253). Several findings suggest ghrelin is involved in the stress response in this context. For instance, exogenous ghrelin stimulates the release of both ACTH and cortisol in humans (257259). A continuous infusion of AG over 5 hours in female rhesus macaques also stimulates cortisol release (260, 261), whereas a bolus injection in the same species has no such effect (262). Studies by Wilson and colleagues (263) further demonstrate that although subordinate female monkeys chronically exposed to social stress do not demonstrate differences in circulating AG, they do have increased sensitivity to the orexigenic effects of ghrelin, even in the fed state. This increased sensitivity stimulates increased consumption of a low caloric diet, when a choice between high and low caloric diets is available. Interestingly, dominant females have reduced consumption of this diet in response to ghrelin. Furthermore, CRH receptor antagonism over 4 days in subordinate and dominant monkeys is efficient at reducing hypercortisolism; however, it stimulates increased caloric intake by subordinates and has anorectic effects in dominant animals (262); again illustrating individual differences in response to stress.

Ghrelin-related dysregulation of the HPA axis can lead to mood disorders, particularly if ghrelin is chronically affected. Individuals with polymorphisms in the gene for preproghrelin can be at increased risk of panic (264) and major depressive disorders (265), and treatment-resistant depressive individuals have elevated ghrelin compared with controls, and those who do respond to treatment (266). With respect to depression, conflicting findings have been seen, which reflect the complexity of ghrelin’s role. Several studies have shown ghrelin is elevated in depression (267269), whereas others have shown no changes (270272). These discrepancies may be accounted for by differences in body mass index or satiety status, because elevated body mass index suppresses, and fasting elevates, ghrelin (273, 274). Thus, Barim et al. (275) found depressive patients have a tendency to have less circulating ghrelin than controls, but body mass index was also lower in this group, potentially independently accounting for the findings. Degree or manifestation of the illness may also play a role because at least one study has found differences in circulating ghrelin depending upon whether the patients were treatment-resistant or -responsive (266).

There is now some support for the idea that elevated ghrelin in depression reflects a role for the peptide in combatting or reducing the effects of this disorder. Thus, treatment interventions that mitigate depression can suppress ghrelin. For instance, electroconvulsive therapy (267, 268), citalopram (275), mirtazapine (276), and other antidepressants (268) reduce circulating ghrelin; albeit that maprotiline elevates it (277). Furthermore, Kluge et al. (278) have shown in a small clinical sample that acute ghrelin administration has a tendency to improve depressive symptoms in men, but not women, with major depressive disorder. It also significantly improves sleep quality in both men and women with depression-related sleep disturbance (278). Transient suppression of catecholamines, which reliably and reversibly induces depressive symptoms, significantly reduces circulating ghrelin in healthy people (279), and a single exposure to exogenous ghrelin led to elevated mood in 30% of healthy people (three of nine) (259).

Ghrelin in stress and mood, mechanistic detail from animal models

Animal models have served to clarify some of the conflicting findings of the human studies. In animal models, as in humans, stress increases ghrelin. With acute stress, including tail pinch and water avoidance, circulating ghrelin and stomach ghrelin mRNA are increased (280282). With ongoing stress, including chronic daily restraint, chronic unpredictable stress, and chronic social defeat, the same pattern is seen (283286), but the increased ghrelin can persist. In mice, increased ghrelin as a result of 10 days daily social defeat was still evident at least 1 month after the last defeat session (286).

“Ghrelin is important for appropriate reproductive function, including development of the reproductive axis.”

Stress-induced ghrelin targets the HPA axis at several levels of the axis, and in healthy individuals under normal conditions probably facilitates an immediate hormonal response that enables effective coping with the cause of the stress (212). GHSR is negligibly expressed in mouse PVN CRH neurons, and ghrelin thus indirectly activates these neurons to stimulate ACTH release from the pituitary (113, 234, 287). In ghrelin knockout (ghr−/−) mice, the PVN c-Fos response to acute restraint is exacerbated compared with wild-type controls, but glucocorticoid release is reduced in the same animals. In the absence of ghrelin, the adrenals are capable of responding normally, because exogenous ACTH causes similar glucocorticoid release in the ghr−/− as in the wild-type. The PVN is also capable of responding normally without ghrelin, because mimicking glucocorticoid release with dexamethasone causes similar activation of this region in both groups (113). GHSR is widely expressed in the anterior pituitary (235). It is also coexpressed with ACTH in corticotropic cells (235), and exogenous ghrelin stimulates ACTH release in vivo (259) and in vitro (288). Thus, ghrelin likely facilitates HPA axis activation by directly stimulating ACTH release from corticotrophs in the anterior pituitary.

Although the PVN CRH cells are not directly activated by ghrelin, the peptide does increase CRH mRNA (280, 289) and does indirectly activate these neurons (287, 290). Among several sites potentially important in relaying stress signals to the PVN following ghrelin stimulation are the amygdala, centrally projecting Edinger Westphal nucleus, locus coeruleus, and the ventral tegmental area (212). Dysregulation of the amygdala is a hallmark of anxiety, depression, and dysfunctional HPA axis responses to stress making it a potentially important site for ghrelin’s activity under stress conditions (291). It is also a key upstream regulator of the GnRH pulse generator (120, 128). Ghrelin signaling via the amygdala is likely indirect or inhibitory. Although the medial amygdala contains abundant GHSR, projects directly to the PVN, and is highly activated following psychological stress, very few of the GHSR-expressing cells are those that express stress-induced c-Fos. Edinger Westphal nucleus urocortin GHSR-containing cells are activated by stress in the presence of ghrelin, and project to the medial amygdala suggesting a possible route by which ghrelin indirectly activates the PVN (113). Ghrelin, and fasting, both strongly activate amygdala activity, as well as increasing CRHR1 mRNA in the region (292294). Intra-amygdala ghrelin injections can also influence the symptoms of depression, although in one study this only occurred in conjunction with calorie restriction (295).

The locus coeruleus also projects directly to the PVN, and these projections are activated by stress to stimulate PVN CRH neurons (296). Intracerebroventricular ghrelin binds to these locus coeruleus catecholaminergic neurons potentially influencing the HPA axis via this pathway (297). The increased sympathetic activity may then increase sympathetic innervation of the ovary, leading to the development of ovarian cysts (202, 203), and contributing to the PCOS phenotype (298, 299). Intracerebroventricular ghrelin similarly increases neuronal activation in the nucleus of the solitary tract, and catecholamine cells from this region can directly activate the PVN and are strongly implicated in regulating an appropriate stress response (67, 297, 300, 301). The ventral tegmental area is another candidate region expressing GHSR, responding to stress, and stimulating the PVN. Activation of the ventral tegmental area may therefore also mediate the effects of ghrelin on HPA axis function. In addition, it has particular significance in ghrelin’s role in the rewarding aspects of reproductive function (302, 303).

Stress-induced ghrelin imbalance regulates brain reward circuitry

As well as, and integrated with, an important role for ghrelin in stress, ghrelin is a key player in responses to motivating stimuli and controlling the feelings of reward. This role has been particularly elucidated with respect to the rewarding effects of food, but also pertains to drugs of abuse and other pleasurable stimuli, likely including sex (304). Thus, conditioned place preference for a food reward in the context of chronic psychosocial stress is not evident in mice lacking the GHSR1a, suggesting ghrelin signaling is essential for such reward behavior (305). Similarly, conditioned place preference for food reward under basal conditions can be blocked with ghrelin antagonists (306). Central ghrelin also increases the preference for highly palatable rewarding foods, including fats and saccharin (307, 308). These ghrelin-induced changes in food preference do not occur in the absence of GHSR (306). The idea that ghrelin itself drives a feeling of reward comes from studies showing satiated mice develop a conditioned place preference to ghrelin alone (302).

To achieve this reward regulation, ghrelin interacts with cortical and mesolimbic areas, including nucleus accumbens, amygdala, and ventral tegmental area (309). GHSR is expressed on midbrain dopamine neurons in the substantia nigra (310) and ventral tegmental area (208), regions important for motivational aspects of multiple behaviors, including sexual behavior (311). Activity of the dopamine system in these regions is elicited by expectation of, or exposure to, pleasurable stimuli, such as sexual experience (312). Ghrelin administration directly into the ventral tegmental area leads to elevated nucleus accumbens dopamine levels (313) and increased feeding behavior (208, 209). Ghrelin directly into the ventral tegmental area also influences motivation to persevere at a task to obtain a food reward. Thus, rats given ventral tegmental area ghrelin had an increase in operant lever pressing and nose pokes to obtain sucrose compared with controls (309, 314). Blocking ghrelin signaling with a ghrelin antagonist suppresses operant performance in calorie restricted rats (314). These effects seem to be particular to the ventral tegmental area because ghrelin administered directly into the nucleus accumbens has no similar effect (309).

In addition to a role for ghrelin in food-related reward, ghrelin signaling can also promote alcohol consumption, and sensitization to other drugs of abuse including cocaine, amphetamines, and nicotine (315322). Indeed, Kaur and Ryabinin (323) have identified that ghrelin’s role in driving alcohol consumption critically involves the Edinger Westphal nucleus, with alcohol stimulating the Edinger Westphal nucleus and ghrelin suppressing this effect, implicating this region as a potential interface between stress and reward processes that can be modulated by ghrelin.

Whether ghrelin plays a specific role in rewarding behavior in mating remains to be explicitly tested. However, many of the mechanisms for such behavior overlap with those for reward in food and drugs of abuse. The mesolimbic dopamine pathway is clearly strongly involved in all types of rewarding behavior, including sexual attraction, sexual pleasure, and interpersonal attachment (324). Nucleus accumbens and ventral tegmental area are both activated in association with mating, and sexual experience can lead to remodeling in these regions, with increases in the number of dendrites and spines in the nucleus accumbens in sexually experienced rats compared with sexually naïve (325327).

To date, few studies have directly examined ghrelin’s role in the rewarding aspects of mating behavior. There is one study that has examined the effects of acute ghrelin on mating, and this found intraperitoneal ghrelin can suppress male ultrasonic calling to receptive females and increase the latency to attack a rival male, evidence of reduced aggression, but preference for female odor was retained (328). These findings are likely to reflect the interplay between ghrelin’s role in reproductive function and in feeding, with acute ghrelin preferentially stimulating feeding-related behavior over mating. In direct assessment of ghrelin’s rewarding role in mating, ghrelin acutely administered peripherally or centrally into the ventral tegmental area and other areas of the dopamine reward system increases sexual motivation and behavior in male mice. These studies have also shown that acute peripheral or central pharmacological suppression of GHSR or genetic deletion of the receptor reduces sexual motivation and behavior in these animals (303, 329). However, persistent elevation of ghrelin induced by chronic stress may have an opposing effect on sexual motivation and behavior, similar to sexual impairment under negative energy balance (330), typically associated with increased ghrelin. We should note here that GHSR-, GOAT-, and ghrelin-knockout mice breed normally under laboratory conditions (Spencer, Sominsky, Andrews, unpublished observations, 2017). However, these conditions are necessarily relatively stress-free and unchallenged. It is likely the absence of an effective ghrelin system would affect motivation to mate as well as other reproductive factors under conditions of chronic stress. In this regard, it will be essential to directly examine mating behaviors, and other fertility factors, in the context of stress in inducible knockouts that have not had the opportunity to developmentally compensate for ghrelin absence.

Ghrelin as a Regulator of the HPG Axis

Ghrelin’s local and systemic role in hypothalamic-pituitary reproductive control

In addition to its role in regulating the stress response and reward, ghrelin is important for appropriate reproductive function, including development of the reproductive axis. It regulates reproductive physiology through its systemic release and local expression, acting at all levels of the HPG axis (331). The local and systemic effects of ghrelin on the adult reproductive system are complex. In regards to its hypothalamic-pituitary reproductive action, AG has been shown to have predominantly inhibitory effects in vivo. Acute intracerebroventricular administration of AG in ovariectomized adult rats suppresses LH pulse frequency, but not the pulse amplitude, suggesting the inhibitory effects of ghrelin on LH release are mediated by its effects at the level of the hypothalamus (155, 332). In support of this, GnRH release by hypothalamic explants from ovariectomized adult rats is inhibited by AG (155). Similar decreases in LH pulse frequency, without concomitant differences in LH pulse amplitude, have been detected in ovariectomized rhesus monkeys subjected to chronic peripheral infusion of AG. These suppressive effects of AG on LH pulsatility are concomitant with an increase in circulating cortisol levels (260), and both of these effects are prevented by CRH receptor antagonist treatment (261), demonstrating the role of ghrelin in the interaction between the HPA and HPG axes. The inhibitory effects of AG on LH pulse frequency also occur in ovariectomized estrogen-replaced fed rats, after acute peripheral administration of AG, and these effects are further exacerbated by overnight fasting (116). Acute AG inhibits LH release throughout the estrous cycle in intact adult female rats (155), intact males, and gonadectomized male and female rats prepuberty, with no changes in FSH release (333). These changes are not apparent in prepubertal intact females (156, 333), indicating that ghrelin’s suppressive effects on GnRH/LH release are reproductive maturity-dependent in females, but potentially not in males. Indeed, another study in male rats has demonstrated that both acute and chronic administration of AG or DAG, as well as an acute coadministration of both peptides, inhibits LH release in prepubertal and adult males, with a similar reduction of FSH in adults (141). The combined inhibitory effects of AG and DAG on LH release that were demonstrated by this group are contrary to the potential antagonistic effects of DAG on the metabolic effects of AG (334). In young adult men, AG has also been shown to suppress LH release, both its pulse frequency and amplitude (217). In women, repeated administration of AG suppresses both LH and FSH (219). Elevated ghrelin levels in exercising women, and women with anorexia nervosa that suffer from chronic energy deficiency, predict menstrual disturbances and hypothalamic amenorrhea (335337). Ghrelin-induced suppression of LH release is similar to that induced by stress (9295). Because energy deficiency is a metabolic stress that leads to an increase in glucocorticoids (338), it is likely that ghrelin and glucocorticoids together communicate inhibitory information to the HPG axis [reviewed in (145)].

Ghrelin has also an inhibitory effect on kisspeptin-stimulated LH release (141). Acute administration of AG, fasting, and their combination suppresses Kiss1 mRNA expression in the mPOA (116), possibly contributing to the inhibitory effects of ghrelin on LH release. However, despite the apparent central inhibitory effects of ghrelin on LH secretion and the mPOA expression of kisspeptin gene, Smith et al. (146) found minimal to no coexpression of GHSR on GnRH, kisspeptin, and tyrosine hydroxylase neurons in the anteroventral periventricular nucleus, or RFamide-related peptide neurons in the dorsomedial hypothalamus, using the GHSR-enhanced green fluorescent protein reporter mouse model. This study demonstrated that over 90% of the GHSR-expressing cells in the anteroventral periventricular nucleus and the periventricular nucleus express estrogen receptor-α (ERα), suggesting that the central inhibitory effects of ghrelin on LH pulsatility may be mediated by estrogen (146). Another study has identified GHSR and ERα coexpression in neurons in distinct hypothalamic nuclei in female mice, including anteroventral periventricular nucleus, the ventrolateral subdivision of the ventromedial nucleus of the hypothalamus, and the arcuate nucleus. Notably, only in the arcuate nucleus was this coexpression, as well as GHSR mRNA expression, mediated by high estrogen levels. The induction of GHSR mRNA expression in the arcuate nucleus by estrogen treatment in ovariectomized mice is specific to neurons expressing ERα and Kiss1 mRNA. Furthermore, direct application of ghrelin induces estrogen-dependent depolarization of arcuate nucleus Kiss1 neurons (117). Estrogen controls energy balance and expenditure, with a reduction in estrogen synthesis and its circulating levels leading to an increase in body weight and adiposity, as is typically seen in women who enter menopause [reviewed in (339)]. Estrogen is also known to regulate stress sensitivity, with reduction in endogenous estrogen enhancing anxiety, and estrogen replacement producing anxiolytic effects at least in some animal models of ovariectomy and in postmenopausal women (340342). A series of studies in subordinate female rhesus monkeys demonstrate that chronic exposure to psychosocial stress modulates physiological responses to estradiol, inducing hypersensitivity to the negative feedback effects of estrogen on LH secretion (12), reduced sensitivity to the anorectic effects of estrogen (343), and impaired anxiolytic effects of estradiol in the context of sexual behavior, even at higher doses (344). These animals also exhibit heightened sensitivity to the orexigenic effects of postprandial AG (262). These effects of chronic stress on the responses to estrogen have been suggested to be at least partially explained by an increase in estrogen-induced GABA-ergic tone in the prefrontal cortex of subordinate females. This social status difference between subordinate and dominant females is reversed by CRH receptor antagonism (345). Studies examining the interactions between estrogen and stress, however, have shown contradictory findings in both animals (346348) and humans [reviewed in (349)]. The different findings reflect differences in behavioral tasks, differences in endogenous and exogenous levels of estradiol and length of exposure to stress. The individual differences in responses to estradiol are particularly important to consider in light of negligible support for beneficial effects of estradiol on mood in postmenopausal women in the Women’s Health Initiative studies (350, 351).

Estrogen-mediated regulation of the stress responses is likely to be mediated by both ERα and ERβ (352), whereas the effects of estrogen on energy balance are primarily mediated by ERα (353). Kiss1-expressing neurons in the arcuate nucleus, a critical region for food intake regulation, coexpress ERα and play a pivotal role in the integration of energy balance signaling and reproduction (354). Although some kisspeptin neurons in this region coexpress ERβ, this receptor does not play an important role in estrogen feedback regulation of GnRH activity (354). Nevertheless, the exact role that estrogen plays in mediating the effects of ghrelin on Kiss1 neurons and hence GnRH pulse generator remains to be established. It is also important to note that there are species differences in the expression of GHSR in brain areas involved in the regulation of reproductive activity, such as mPOA, because GHSR is expressed in this region in mice, but probably not in rats (117, 234), suggesting indirect mechanisms may be involved in the inhibitory effects of AG on Kiss1 expression in this region in rats (116). These differences warrant further species-specific investigation into the central effects of ghrelin on reproductive signaling.

In addition to its central effects on GnRH/LH release, there is a potential for ghrelin to directly influence pituitary LH and FSH secretion, as it does for ACTH (113). In the mouse pituitary, GHSR is exclusively expressed in the anterior pituitary, with higher expression in males than in females, as has been demonstrated using the GHSR-enhanced green fluorescent protein mouse model (235). The highest expression of GHSR is evident in somatotrophs, corresponding with ghrelin’s potent growth hormone releasing activity. Modest expression of GHSR-enhanced green fluorescent protein is found in gonadotrophs and lactotrophs, indicating that ghrelin can act directly at the pituitary level to influence LH, FSH, and prolactin secretion (235), as it does for ACTH (113). Interestingly, the direct effects of AG on pituitary gonadotropins are opposite to its centrally mediated inhibitory effects, with an in vitro application of AG on male and female rat anterior pituitaries dose-dependently potentiating basal LH and FSH production (155, 156, 333). These direct stimulatory effects are estrogen-dependent, with an attenuation of these effects at estrus or after ovariectomy in adult rats, as well as in neonatally estrogenized females prepuberty (155, 156). Cyclic fluctuations in pituitary GHSR have also been reported, with decreased GHSR mRNA expression at estrus and metestrus (155). The involvement of estrogen in the effects of ghrelin on gonadotropin release is not surprising, because the majority of GHSR-expressing cells in the anterior pituitary of both males and females coexpress ERα (235). When AG and GnRH are added simultaneously to the incubation medium, the effects of AG appear to be age-dependent. In adult female rats, AG inhibits GnRH-stimulated LH release in vitro at all stages of the estrous cycle, whereas it potentiates this GnRH-stimulated LH production by anterior pituitaries of intact and ovariectomized prepubertal females (155, 156). In contrast to these in vitro findings, continuous infusion of AG in men has been shown to inhibit spontaneous LH pulsatility, as well as the LH response to naloxone, but not to a GnRH stimulus (355). Naloxone is an opioid antagonist that acts centrally to induce gonadotropin secretion (356). Therefore, these results suggest that despite the ability of AG to directly influence the pituitary function in vitro, its in vivo effects on the release of gonadotropins are predominantly centrally mediated.

At least part of ghrelin’s role in fertility is likely mediated through prolactin. Prolactin is produced by the lactotrophs in the anterior pituitary (357). Besides its crucial role in the initiation and maintenance of lactation, prolactin has a wide variety of physiological roles, including in reproduction, and stress responsiveness (358, 359). Prolactin attenuates the effects of stress, and is thus likely to at least partially mediate the diminished stress responsivity seen during pregnancy and lactation, when prolactin levels are upregulated (360, 361). Prolactin has been recently implicated in resilience to the effects of chronic stress in rats, with those animals that were more resilient to stress demonstrating higher plasma levels of prolactin than their more vulnerable counterparts (362). Hyperprolactinemia, however, is known to suppress GnRH/LH pulsatility (363365), and is a major cause for infertility in males and females (366). This is particularly relevant to our discussion of the effects of ghrelin on fertility, because exogenous AG has been shown to stimulate prolactin release in men and women (257, 258, 367), as well as in human pituitary cells in vitro (368), with no effects of DAG on prolactin levels when administered alone or in combination with AG (369). On the other hand, intracerebroventricular administration of AG in sheep failed to induce changes in circulating prolactin (370). The effects of AG on prolactin release may also be dependent on reproductive maturity and cyclicity, because in prepubertal males and females, as well as in hyperprolactinemic aged female rats, both systemic and central administration of AG inhibited prolactin release, with no changes in prolactin production being evident when pituitary samples were challenged with increasing doses of AG in vitro (371). Other studies on primary cultures of female rat pituitary cells have demonstrated stimulation of prolactin release by GHSR1a agonists [reviewed in (372)]. Similarly, cortistatin, a neuropeptide that binds GHSR1a, was found to increase prolactin release in mice, in vivo and in vitro, and in primary pituitary cell cultures of nonhuman primates, an effect that is blocked by GHSR1a antagonists (373). In mice, GHSR deletion results in an important reduction in the mRNA expression of prolactin and fewer prolactin-positive cells in the pituitary. These changes are associated with reduced expression of the pituitary-specific transcription factor that is essential for differentiation of pituitary cells into somatotrophs, lactotrophs, and thyrotrophs (374). These data suggest that the effects of AG on the release of prolactin are likely to be at the level of the pituitary gland, with potential species and reproductive age-related differences. Because high prolactin inhibits LH release (365), a stimulatory effect of AG on prolactin levels may contribute to its inhibitory effects on LH pulsatility.

The ghrelin system in the gonads

The ghrelin system in the testis

Species-specific mRNA and protein expression of ghrelin and GHSR have been demonstrated in mammalian and nonmammalian ovary and testis [reviewed in (331, 375, 376); see Table 2]. The GHSR gene is persistently expressed in rat testis from infancy to adulthood (377), including after selective elimination of Leydig cells (378). On the other hand, specific GHSR1 amRNA and protein expression becomes detectable only postpuberty in mature nonreplicating Sertoli and Leydig cells (377). In addition, GHSR and GHSR1 amRNA are detected in the seminiferous epithelium at all stages of the spermatogenic cycle, with variable expression across the cycle. Specifically, minimal detection of GHSR and GHSR1 amRNA appears in the rat seminiferous epithelium at stages VII to VIII (377), which are also characterized by the lowest expression of the FSH receptor and hence lower sensitivity to FSH (382, 383). In support of this potential relationship between the expression of GHSR/GHSR1a and FSH responsivity, stimulation of the testes with FSH in vivo significantly upregulates total GHSR and specific GHSR1a gene expression in adult rats, with no effect of human chorionic gonadotropin treatment that acts as LH superagonist in vivo or in vitro on the expression of these ghrelin receptor isoforms (377). In addition, in vivo intratesticular challenge with AG suppresses proliferation of differentiating immature Leydig cells, and is associated with decreased expression of stem cell factor, a primary regulator of Leydig cell development. This inhibitory activity of AG is dependent on FSH signaling, because it is absent in hypophysectomized rats, but restored upon FSH replacement (186). Chronic systemic administration of AG has also been shown to induce morphometric alterations and a reduction in the number and functional capacity of different spermatocytic cells in adult rats (187). In vitro challenge of rat testicular tissue with AG has been shown to increase the expression of GHSR and GHSR1a (377), and to inhibit human chorionic gonadotropin and cyclic adenosine monophosphate-stimulated testosterone secretion (378). Zhu et al. (384) has demonstrated GHSR1a expression in the mouse testis in steroidogenic Leydig cells, Sertoli cells, as well as in germ cells, particularly in transcriptionally inactive elongating/elongated spermatids, indicating that ghrelin may be involved in spermatogenesis/spermiogenesis. Indeed, abnormal spermatogenesis in the testis of leptin deficient ob/ob mice, characterized by arrest at the elongating spermatid stage, is improved by GHSR antagonist treatment (384). In human testis, GHSR1a has also been located in germ cells, but mainly in meiotic pachytene spermatocytes (228), as well as in Leydig and Sertoli cells (228, 385).

Table 2.

Distribution of GHSR in Gonads

Ovaries
 Ovarian follicles (all developmental stages)+
 Granulosa cells+
 Theca cells+
 Luteal cells+
 Oocytes−/+
Testes
 Germ cells+
 Sertoli cells+
 Interstitial tissue (Leydig cells)+
Ovaries
 Ovarian follicles (all developmental stages)+
 Granulosa cells+
 Theca cells+
 Luteal cells+
 Oocytes−/+
Testes
 Germ cells+
 Sertoli cells+
 Interstitial tissue (Leydig cells)+

Gonadal data are from sheep; adapted from (237). Gonadal GHSR expression has been identified in rodents (226, 377, 378), sheep (237), pigs (379), and humans (228, 229, 380, 381).

Abbreviations: −/+, detectable expression in some cells; +, some detectable expression.

Table 2.

Distribution of GHSR in Gonads

Ovaries
 Ovarian follicles (all developmental stages)+
 Granulosa cells+
 Theca cells+
 Luteal cells+
 Oocytes−/+
Testes
 Germ cells+
 Sertoli cells+
 Interstitial tissue (Leydig cells)+
Ovaries
 Ovarian follicles (all developmental stages)+
 Granulosa cells+
 Theca cells+
 Luteal cells+
 Oocytes−/+
Testes
 Germ cells+
 Sertoli cells+
 Interstitial tissue (Leydig cells)+

Gonadal data are from sheep; adapted from (237). Gonadal GHSR expression has been identified in rodents (226, 377, 378), sheep (237), pigs (379), and humans (228, 229, 380, 381).

Abbreviations: −/+, detectable expression in some cells; +, some detectable expression.

Ghrelin itself is also expressed in the testis. In rat testis, the ghrelin gene has been detected at all stages of development, and ghrelin’s immunolocalization has been identified in mature fetal and adult Leydig cells (378, 386). Ghrelin protein expression in rat testis is specific to Leydig cells, because it is undetectable after selective Leydig cell elimination (386). Barriero et al. (386) has also shown that testicular ghrelin mRNA and protein expression in rats is dependent on pituitary LH, with a reduction in ghrelin’s expression after hypophysectomy, and its partial restoration after human chorionic gonadotropin replacement. Furthermore, administration of human chorionic gonadotropin to intact male rats transiently increases testicular ghrelin mRNA levels, with no effect of FSH administration (386), contrary to the FSH-dependent regulation of GHSR1a expression and activity (186, 377). These findings suggest that Sertoli cells, primary responders to FSH, may not play a role in the regulation of ghrelin expression in rat testis (386). Ghrelin immunostaining is also detected in steroidogenic Leydig cells in mice, and is significantly increased in the testis of ob/ob mice that are obese and infertile. Inhibition of ghrelin signaling in these animals, in turn, restores steroidogenic activity, reduces germ cell apoptosis, and improves sperm production (384). Interestingly, however, ghrelin has also been shown to attenuate testicular dysfunction induced by ionizing radiation, heat, cadmium, and chemotherapy in mice and rats (387390), potentially due to ghrelin’s antioxidant properties (213). In human testis, ghrelin is strongly present in steroidogenic Leydig and to a lower extent in Sertoli cells, but is not present in germ cells (228, 385, 391). Ghrelin expression in Leydig cells is negatively correlated with serum testosterone levels, suggesting testicular ghrelin is involved in steroidogenesis (385). Moderate protein expression of ghrelin is also evident in the human rete testis, efferent ductules, epididymis, vas deferens, seminal vesicles, as well as in spermatozoa (391). Contrary to rodent and human testis, in adult sheep ghrelin is also detected in the germ cells, in addition to interstitial (Leydig) and Sertoli cells, with increased immunoreactivity in the germ cells prior to the first meiotic division of the spermatogenic cycle (237).

The ghrelin system in the ovary

GHSR1 amRNA and protein expression has been described in pig, sheep, and human ovary (229, 233, 237, 380). In human ovary, GHSR1a has a wide distribution, and is immunolocalized to oocytes, cuboidal granulosa cells, theca cells, hilus interstitial cells, as well as steroidogenic luteal cells in young, mature, old, and regressing corpus luteum (380). GHSR1a is also expressed in ovarian surface epithelium and in the ciliated cells within the human fallopian tube epithelium at all phases of the cycle (229). In vitro stimulation of cultured human granulosa-lutein cells with AG has been shown to inhibit steroidogenesis, and this effect is prevented by GHSR1a, but not GHSR1b antagonism, despite the more abundant expression of the latter receptor in the granulosa-lutein cells (191). In sheep ovary, GHSR1a is similarly immunolocalized to ovarian follicles at all developmental stages, with stronger signal in the granulosa than the theca cells, low levels of detection in the oocytes, and positive immunostaining in the luteal cells of the corpus luteum (237). GHSR1a presence in the prepubertal porcine follicles was found to mediate ghrelin’s stimulatory effects on estradiol secretion, aromatase activity, and cell proliferation. GHSR1a antagonism, however, did not affect ghrelin-induced suppression of cellular apoptosis suggesting this specific in vitro effect of ghrelin may be independent of GHSR1a binding (233). In rats, chronic in vivo treatment with AG induced a substantial decrease in the number of corpora lutea and in the ovarian volume, but increased the number of ovarian follicles with a reduced mean diameter (214), potentially reflective of the effects of AG on cell proliferation and apoptosis. Another study in rats has demonstrated that chronic treatment with AG, DAG, or their combination during the peripubertal period delays follicular maturation and results in decreased ovarian weight, suggesting the inhibitory effects of ghrelin on ovarian development are only partly dependent on GHSR1a pathways (392). These effects of chronic AG treatment are similar to the deleterious effects of chronic stress on ovarian apoptosis and oocyte developmental potential (7).

Ovarian ghrelin expression has been demonstrated in the cytoplasm of the hilus interstitial cells and granulosa lutein cells in the young and mature corpus luteum, but is not detected in regressing corpus luteum, nor is it detected in oocytes or somatic cells of the ovarian follicles in human ovary (380). The ghrelin gene is also expressed in rat ovary, and its expression levels depend on the stage of the estrous cycle, with lowest detection in proestrus and highest expression in diestrus, during the luteal phase of the cycle. This cyclic expression of the ghrelin gene has been shown to be dependent upon gonadotropin signaling, because administration of GnRH antagonist induces a decrease in ovarian ghrelin mRNA levels throughout the luteal phase of the estrous cycle (226). Ghrelin immunostaining is not detected in growing and preovulatory follicles, but is strongly detected in the cytoplasm of steroidogenic luteal cells in corpus luteum of the current cycle, as well as in regressing corpus luteum (226), a finding that is different to the stage-dependent localization of ghrelin in the corpus luteum of the human ovary (380). Overall the strong and cycle-dependent expression of ghrelin in the corpus luteum suggests ovarian ghrelin may also play a direct role in the regulation of steroidogenesis, in addition to ghrelin’s systemic suppressive effects on the HPG axis function.

Ghrelin’s role in the regulation of puberty

In line with the local and systemic, mostly inhibitory, effects of ghrelin along the HPG axis, and its role in metabolism and energy balance, ghrelin has been shown to play a role in pubertal development. Although the critical role of the metabolic hormone leptin on puberty onset has been well established, the role of ghrelin in the regulation of puberty has not yet been fully elucidated. Few studies have demonstrated in rats that in contrast to the increased sensitivity of female puberty to the permissive effects of leptin (393), male puberty appears to be more sensitive to the effects of ghrelin. Although chronic administration of low doses of ghrelin induced a delay in the onset of puberty in male, but not female rats (394), only a high dose of ghrelin was able to induce a similar delay in vaginal opening females (392). These inhibitory effects on the onset of puberty have been induced by chronic treatment with both AG and DAG, suggesting GHSR1a-dependent and -independent mechanisms may be involved (141, 392). Human data indicate that circulating total ghrelin levels are significantly increased during early postnatal life and then decline with age until the end of puberty and early adulthood (395, 396). This relationship between the decrease in ghrelin and advanced pubertal age is more pronounced in boys than in girls, and is also associated with a negative correlation between ghrelin and insulinlike growth factor 1 (396). Insulinlike growth factor 1 plays an important role in the activation of GnRH pulsatility at puberty (397). These findings therefore suggest that the decline in circulating ghrelin levels during the peripubertal period may facilitate growth and act as a permissive signal on puberty onset. Additional limited information is available from studies in boys with constitutional delay of growth and puberty (CDGP). CDGP is characterized by short stature, delay in bone maturation, and delayed puberty (398). CDGP is also often associated with substantial psychological stress (399401). Circulating ghrelin in boys with CDGP is negatively correlated with anthropometric parameters, such as body mass index, height, and weight (402, 403), as well as testicular volume, gonadotropins, and testosterone (403). It is important to note, however, that these studies report total ghrelin levels and do not distinguish between AG and DAG, which may play independent roles in human pubertal development. Therefore, further investigation is required to elucidate the function of the ghrelin peptides and their mechanisms of action in male and female puberty.

Ghrelin Signaling in Pregnancy

Ghrelin’s role in fertilization and implantation

Ghrelin and its receptor expression in reproductive tissues has also been demonstrated in human, rat, and sheep placentae, endometria, and fallopian tubes (229, 404), as well as in fetal tissues (405, 406), indicating its potential involvement in the course of pregnancy (see Fig. 5). In addition, ghrelin gene expression has been detected in pregnant rat ovary, with higher expression in early, than in late gestation (226).

Ghrelin and pregnancy. Early in pregnancy, ghrelin regulates decidualization of the endometrium and blastocyst–endometrial interactions. Circulating total and acylated (AG) ghrelin levels increase at midgestation in human pregnancy, followed by declining levels during the peripartum period. Maternal ghrelin enters fetal circulation, with both hypo- and hyperghrelinemia leading to adverse effects on neonatal outcomes, including birth weight, neurodevelopment, and fertility. Adapted from Servier Medical Art under Creative Commons CC-BY license.
Figure 5.

Ghrelin and pregnancy. Early in pregnancy, ghrelin regulates decidualization of the endometrium and blastocyst–endometrial interactions. Circulating total and acylated (AG) ghrelin levels increase at midgestation in human pregnancy, followed by declining levels during the peripartum period. Maternal ghrelin enters fetal circulation, with both hypo- and hyperghrelinemia leading to adverse effects on neonatal outcomes, including birth weight, neurodevelopment, and fertility. Adapted from Servier Medical Art under Creative Commons CC-BY license.

Pregnancy begins with successful fertilization and implantation, and these are highly dependent on endometrial receptivity (407). Several factors, such as ovarian steroids, cytokines, and neuropeptides, as well as glucocorticoids, regulate this complex interaction (408411). Glucocorticoids play an important role throughout pregnancy, and maternal glucocorticoids are critical for fetal development [reviewed in (412)], but exposure to excess glucocorticoids due to chronic maternal stress is associated with the development of metabolic disorders and an increased risk of emotional and cognitive disturbances in later life [reviewed in (413)]. Ghrelin is also likely to be involved in the maintenance of pregnancy, including the implantation process (216, 414, 415). In women, ghrelin gene and protein expression are significantly increased in decidualized, compared with nonpregnant endometrium, whereas GHSR expression is evident in both cycling and pregnant endometrium (414), and is significantly increased in the midsecretory phase, the time of implantation (215). Interestingly, reduced midsecretory endometrium expression of ghrelin and GHSR1a, at the stage when implantation is likely to occur, is associated with reproductive dysfunction and infertility (215). Ghrelin mRNA and immunolocalization is also strongly detected in first trimester human placenta (414, 416), with negligible to no expression of GHSR transcripts in first trimester and at term placentae (381, 414, 417). In rat placenta, ghrelin mRNA detection is minimal during early gestation, peaking at gestational day 16, and followed by decreasing expression at the latest stage of gestation (416). Ghrelin immunolocalization has also been detected in ovine placentae throughout gestation, with maximal expression occurring toward mid gestation, as well as persistent GHSR1a immunoexpression, with no effect of gestational age on its placental levels (404). Ghrelin and GHSR1 amRNA and protein levels are also detected in in vitro preimplantation sheep embryos (418). These patterns of expression indicate differences between species in the pregnancy-related time course of the ghrelin system expression, likely to reflect species differences in gestation length and energy demands (419).

Ghrelin mRNA expression has been found to be significantly increased in endometrial stromal cell coculture with first trimester human placenta, as compared with endometrial stromal cell primary culture alone. Moreover, addition of AG to the incubation media increases decidualization of endometrial stromal cells induced by cyclic adenosine monophosphate in vitro (414, 420), suggesting ghrelin may play role in the remodeling of the endometrium in preparation for pregnancy. Ghrelin immunostaining is also detected in human blastocysts, and ghrelin levels are present in blastocyst culture medium, suggesting ghrelin is secreted by the blastocysts and may be involved in the blastocyst–endometrium interaction (215). Low and medium, but not high doses of AG, have also been shown to stimulate proliferation and decrease apoptosis in human choriocarcinoma cell line JEG-3, common processes in placental formation (421). AG can also promote the rate of blastocyst formation in vitro (422, 423), but to exert inhibitory effects on the inner cell mass and trophectoderm cell numbers in blastocysts, negatively affecting potential embryo viability in sheep and mice (415, 422, 423). Corresponding to the latter inhibitory effects, AG has been shown to diminish the rate of meiotic maturation of the porcine oocytes in vitro (424). Importantly, the effects of AG on blastocyst formation and embryo quality are not directly dose-dependent, with some doses improving and others inhibiting blastocyst formation rate (425). These nonlinear effects of AG may reflect the differences between the exogenous doses to naturally circulating ghrelin under basal, overfed, fasting, or stressed conditions (256). Stress conditions, when glucocorticoid and AG levels are high (253, 426), have been reported to produce a negative effect on blastocyst formation and implantation (427).

Circulating ghrelin levels fluctuate during the course of pregnancy, with a substantial increase in AG and total ghrelin levels at midgestation in human pregnancy compared with nonpregnant women, followed by declining levels in the third trimester (428, 429), as opposed to a peak in total (430), AG (236), and DAG (405) levels toward the end of gestation in rats, followed by a decline after parturition (430). Generally, circulating AG levels are lower in human pregnancy than postpartum (431), suggesting that AG may contribute to the adaptation to a positive energy balance in pregnancy. The decrease in AG cannot be attributed to its increased deacylation, because the activity of butyrylcholinesterase, the enzyme that deacylates circulating ghrelin (432), is also significantly reduced in pregnancy (431). The peripartum period is also associated with stress-hyporesponsiveness, in humans and nonhuman animals, vital for fetal development and for maternal mental health and wellbeing (433439).

Although one study has shown a negative association between circulating and follicular fluid total ghrelin levels and embryo development (440), others have found no effect of ghrelin on human embryo quality and pregnancy success (441, 442). Circulating total ghrelin levels in early pregnancy have also failed to predict pregnancy viability in women undergoing in vitro fertilization procedures (443). However, in vivo studies in rodents have demonstrated the ability of exogenous ghrelin to interfere with pregnancy success. In mice, hyperghrelinemia induced by a high dose of AG, as well as GHSR1a antagonism during the peri-implantation and early gestation periods, led to adverse effects on pregnancy outcomes, including diminished fertilization rate, and delayed embryo development (216). Similarly, in rats, chronic administration of AG during the first half of gestation results in reduced litter size at birth (394). Therefore, although variations in fluctuation of ghrelin in pregnancy may not be sufficient to predict pregnancy outcomes, disruption of this natural process may be detrimental to pregnancy viability and success. Similar to CRH or glucocorticoid excess, that may contribute to the risk of early miscarriage (13, 444), evidence presented previously suggests that stress-induced hyperghrelinemia is likely to lead to negative pregnancy outcomes. It is important to note that some studies have not found an association between stress and/or excess glucocorticoids and pregnancy failure (445, 446), thus the causal link between stress-induced changes in the availability of ghrelin and pregnancy outcomes need to be assessed in future studies. It is also important to specifically investigate the acylation status of circulating ghrelin in its relation to pregnancy, because some studies provide measures of total ghrelin, which include both AG and DAG (430, 440, 441, 443).

Ghrelin’s role in fetal development

AG and DAG are present in the fetal circulation, with decreasing levels of both peptides at the end of gestation in rats, whereas amniotic levels of DAG remain high at this time (405). Nakahara et al. (405) has also demonstrated that maternal ghrelin (AG) rapidly crosses the placenta and enters fetal circulation, and that chronic treatment with AG, but not DAG at the end of pregnancy significantly and dose-dependently increases neonatal body weight at birth. On the other hand, exposure to maternal AG deficiency has been shown to reduce body weight at birth in rats (405), as well as to affect the fertility of the offspring, leading to abnormal endometrial function in these animals (447). Circulating total ghrelin levels in term infants negatively correlates with birth weight and body length. These correlations are absent in preterm infants (born between 23 and 36 weeks of gestation) (448, 449). Furthermore, total ghrelin levels are significantly increased in the circulation of intrauterine growth-restricted fetuses (450), suggesting that ghrelin may potentially begin to regulate neonatal growth and metabolism at a late gestational stage and may play a role in fetal adaptation to an adverse environment.

“We propose that elevated maternal AG is likely to influence the reproductive potential of the offspring.”

Ghrelin, GHSR1a, GOAT and prohormone convertase 1/3, that is responsible for the conversion of proghrelin to ghrelin (451), are all expressed in the human myometrium, indicative of its potential for autocrine and paracrine effects in this tissue (452). O’Brien et al. (452) has also shown downregulation of ghrelin mRNA and protein expression, along with a substantial decrease in GHSR1, GOAT, and prohormone convertase 1/3 protein expression during labor. Because ghrelin has been shown to inhibit human myometrial contractility in vitro (453), this decrease in the myometrial expression of the ghrelin system at labor may potentially be necessary to allow successful parturition. This decrease in ghrelin expression coincides with stress-hyporesponsiveness and suppression of the HPA axis activity at parturition, driven by endogenous opioids, prolactin, and oxytocin that act together to suppress stress-responsivity in the peripartum period (433, 454, 455).

Maternal total ghrelin levels subside immediately after delivery and are lower in lactating rats (430), and in both breastfeeding and nonbreastfeeding women postpartum (456), than in nonpregnant controls. Another study that assessed the acylation status of ghrelin in human mothers has shown that although total ghrelin concentrations begin to increase between days 4 to 180 of lactation, circulating AG levels continue to decrease (457). Fasting ghrelin levels, that are lower in postpartum than nonpregnant women, are negatively correlated with body mass index and fat mass in the mother (456). Interestingly, ghrelin levels in lactating rats do not appear to be dependent on prolactin and oxytocin, the hormonal regulators of lactation (430). Administration of dopamine agonist decreases circulating prolactin, and treatment with the dopamine antagonist increases prolactin levels, with no effect on the concentrations of ghrelin. Oxytocin antagonist has also been shown to inhibit the levels of oxytocin, without affecting circulating ghrelin levels (430). AG and total ghrelin are also present in human breast milk (457460) and their levels correlate with circulating ghrelin concentrations in breastfed infants (457, 460). Ghrelin mRNA is present in the mammary gland, suggesting the source of ghrelin in breast milk may be both the mammary gland and the circulation (459, 461). Similarly, glucocorticoids are present in mother’s milk, positively correlating with maternal circulating concentrations, and elevated levels of glucocorticoids in the milk have been found to influence offspring behavioral phenotype, including increased fear and negative emotionality in human infants and nonhuman primates (462464).

Recent discoveries, including our own, have identified the role of AG and DAG in fetal and neonatal development. Both AG and DAG are involved in fetal neurogenesis within the hypothalamus and the spinal cord, with the proliferative effects of AG, but not DAG continuing after birth in rats (406). In the neonatal period, AG regulates hypothalamic development by limiting the leptin-induced growth of hypothalamic connectivity (465). Changes to circulating AG and DAG have been shown in neonatally overfed mice (466) and rats (467), respectively. These changes are associated with altered central responsiveness to exogenous ghrelin, and may in part explain the development of an obese phenotype in this model (466, 467). Although the role of ghrelin in human fetal and neonatal development has not yet been established, it is highly plausible that maternal ghrelin similarly influences critical developmental processes in infants. Alterations to ghrelin’s naturally fluctuating circulating and breast milk levels during the peri- and postpartum periods may thus differentially program brain development. In this regard, a study in pregnant mice has demonstrated that chronic administration of AG during pregnancy increases an anxietylike phenotype, basal circulating CRH and fasting AG levels in adult offspring, along with a reduction in hypothalamic GHSR1a and neuropeptide Y gene expression (468). Interestingly, the authors have also demonstrated that exposure to chronic stress in pregnancy induces an increase in endogenous maternal AG and DAG, and this effect is then reflected in increased fetal AG levels (468). These data indicate that maternal ghrelin has a direct effect on the fetus, and these early alterations in the maternal–fetal ghrelin milieu induce long-lasting neuroendocrine and behavioral changes in the offspring. Although fetal development obviously has complex inputs from many factors additional to ghrelin and more research is required to establish the link between maternal AG, DAG, and neurogenesis in humans, the findings we presented previously are consistent with our hypothesis that stress-induced increases in maternal ghrelin levels may affect pregnancy success and program fetal and neonatal development. Because maternal AG can be transferred to the offspring during the peripartum period (405, 457, 460), and increased AG has mostly inhibitory effects on the reproductive axis (375, 392, 469, 470), we propose that elevated maternal AG is likely to influence the reproductive potential of the offspring.

The role of ghrelin in pregnancy-associated disorders

Clinical perspectives on ghrelin dysregulation in pregnancy: Who is at risk?

Overweight and obesity significantly increase the risk of pregnancy complications, including preeclampsia and gestational diabetes mellitus (GDM). Both AG and DAG are significantly decreased in obese individuals (266, 471) and high-fat feeding in mice has been shown to induce hypothalamic resistance to ghrelin (472, 473) that is reversed by calorie-restricted diet (474). As discussed in more detail in Preeclampsia and Gestational Diabetes sections, dysregulation of ghrelin signaling is associated with preeclampsia, GDM, intrauterine growth-restriction, and other complications, and is also independently implicated in adverse pregnancy outcomes. Although the pathophysiological relationship between metabolic disorders and pregnancy is well recognized, the influence of psychological stress and stress-related disorders on pregnancy outcomes is often overlooked (475). Chronic stress during pregnancy and higher levels of maternal CRH has been shown to predict preterm delivery (476). Depression and anxiety have been similarly demonstrated to be important risk factors for pregnancy loss, preterm delivery, and low birth weight (477479). Chronic stress has also been suggested to increase the risk of preeclampsia (480). The role of perinatal stress in developmental programming of the brain, cognition, and behavior, as well as in developmental programming of metabolic dysfunction has also been established [reviewed in (481483)]. As discussed earlier, chronic stress also contributes to the development of metabolic disorders (484), and is associated with elevated ghrelin levels (121, 280282). Therefore, assessment of AG and DAG, cortisol, along with body mass index, blood pressure, glucose tolerance, and other stress and metabolic biomarkers, may help to predict the risk of pregnancy complications. Later we present the existing evidence for the role of ghrelin in preeclampsia and GDM, the common complications of pregnancy.

Preeclampsia

Gestational hypertension and preeclampsia affect 3% to 6% of pregnancies, with a higher incidence in first pregnancies (485, 486). Preeclampsia is associated with the risks of intrauterine growth-restriction, preterm delivery, and perinatal mortality (487489). Although the underlying etiology remains uncertain and likely involves many contributing factors, obesity is considered to be a major risk factor for hypertension and preeclampsia (490). Leptin levels are increased in pregnancy, and are particularly increased in women with preeclampsia compared with women with a healthy pregnancy (491), and these levels are positively correlated with body mass index (492). In contrast, total ghrelin levels have been found to be significantly decreased in pregnant women with preeclampsia as compared with healthy pregnant women, and to negatively correlate with blood pressure (493495). This inverse relationship between circulating ghrelin and blood pressure is also typically present in nonpregnant healthy and hypertensive individuals, as well as animals with hypertension [reviewed in (496, 497)]. Makino et al. (498), however, has shown that although total ghrelin levels negatively correlate with blood pressure, these levels are increased in preeclamptic as compared with healthy pregnant women. It is important to note the potential differences in the gestational stage at the time of ghrelin assessment between the studies, as well as in the methods of sample analysis and processing. Because rapid deacylation of ghrelin occurs in circulation (432), protection of ghrelin from deacylation upon blood sample collection is essential to evaluate physiological concentrations of AG and DAG (251, 499). Therefore, further investigation into the specific roles of AG and DAG in preeclampsia is required.

Psychological stress is also a substantial contributor to preeclampsia in women (500504). Animal studies have shown chronic stress in pregnancy leads to hypertension accompanied by increased adrenal weight, as well as an increase in circulating catecholamines, CRH, and insulin levels in pregnant rats (505507). Increased blood pressure and adrenal weight in the dam are associated with lower fetal weight and increased fetal adrenal weight and blood pressure (507), similar to changes that occur in human preeclampsia. The development of preeclampsia in conditions of chronic distress has been proposed to be mediated by increased levels of glucocorticoids that are independently associated with hypertension and endothelial dysfunction, common features of preeclampsia (480). With respect to hypertension and ghrelin levels, spontaneously hypertensive pregnant rats have significantly increased circulating total ghrelin levels compared with normotensive controls at the last day of gestation. However, placental expression of the ghrelin gene is significantly lower in hypertensive than in normotensive rats (508). In contrast, a study using Dahl salt-sensitive rats as a model of hypertension and intrauterine growth-restriction, has found that the levels of placental ghrelin are significantly higher in Dahl salt-fed pregnant rats than in Dahl pregnant rats fed a control chow diet (509). Although both these studies indicate a potential for an independent placental synthesis of ghrelin, the discrepancies in the direction of change between hypertensive to normotensive pregnancies may be related to specific strain differences, emphasizing the need for further research of ghrelin’s role in human pregnancy, particularly in mediating the deleterious effects of psychological stress.

“Prolonged exposure to increased AG has overall negative impacts on psycho-physiological state.”

Gestational diabetes

Another common complication in pregnancy is GDM. GDM affects 2% to 10% of pregnancies and its prevalence has significantly increased over the past 20 years worldwide (510, 511) and is a substantial risk for the development of type 2 diabetes (512). Overweight and obesity prior to and during pregnancy predispose to GDM (513, 514). GDM exposes the fetus to maternal hyperglycemia, leading to fetal macrosomia and hyperinsulinemia (515, 516). Several studies in women with GDM have identified an association with psychological stress and mood disorders (517, 518); however, the causality of this direction is not clear, because the diagnosis of pregnancy complications is itself a stressful experience. Nevertheless, mood disorders are known to contribute to hyperinsulinemia and insulin resistance (519, 520), and exposure to perinatal stress has been shown to promote insulin resistance in the offspring and induce diabetes-related autoimmunity, independently of other risk factors (521, 522), suggesting psychological stress during pregnancy may contribute to the development of insulin resistance and GDM, with long-term developmental implications. Psychological stress has also been shown to increase the production of inflammatory markers in pregnant women (523), consistent with increased risk of pregnancy complications, including GDM (524, 525).

Several studies have assessed the role of ghrelin as one of several likely players in the development of GDM, with some inconsistencies regarding the change of total ghrelin vs specific changes in AG and DAG in women with GDM. AG has been found to be decreased (429), whereas others found no changes in AG, but an increase in DAG in women with GDM both during pregnancy and postpartum (431). Another study in a small sample of diabetic pregnant women, using glucose and insulin clamp techniques, has demonstrated that acute increases in glucose and insulin do not affect AG, but significantly decrease circulating DAG levels (526), suggesting DAG may be involved in the regulation of energy balance in GDM. Although no differences have been found in circulating total ghrelin levels at baseline or after a glucose load between women with GDM and pregnant women with normal glucose tolerance (527, 528), ghrelin mRNA expression is significantly increased in the placental tissue of GDM women (528). Interestingly, a follow-up study for a period of 8 to 10 years of 98 women with GDM has revealed that 22.5% of the participants developed type 2 diabetes, and low total ghrelin levels at 12 weeks postpartum was a substantial risk factor for this complication (529).

Linking Ghrelin Imbalance and Stress in Reproductive Dysfunction: A Role for an Endogenous Hormone

The role of ghrelin signaling in PCOS

Our previous discussion has demonstrated that ghrelin and stress are both intimately and interactively involved in regulating all aspects of reproductive function, and their interaction typically leads to detrimental outcomes (see Fig. 6). One of the more researched reproductive disorders in the context of ghrelin signaling is PCOS. PCOS is typically characterized by polycystic ovarian morphology, and often accompanied by insulin resistance, hyperandrogenism, and chronic anovulation (530, 531). Women with PCOS demonstrate increased sympathetic nerve activity (298), increased sympathetic innervation of the ovaries (299), and an impairment in noradrenaline reuptake or deamination (532). Obesity significantly increases the risk of PCOS and contributes to other comorbidities of PCOS, including insulin resistance (533). As previously discussed, both stress and ghrelin activate the sympathetic nervous system (296, 297), contributing to the development of PCOS (202, 203, 298, 299). Altered cortisol metabolism has also been attributed to the pathophysiology of PCOS (534536).

Ghrelin regulates the effects of stress on fertility. Exposure to stress involves the coordinated interaction between the sympathomedullary system, the HPA axis, the HPG axis, and the ghrelin system. Ghrelin regulates the stress response by acting indirectly on CRH neurons in the PVN and directly at the anterior pituitary gland to facilitate ACTH release. Ghrelin also binds to catecholaminergic neurons in locus coeruleus (LC) increasing noradrenaline production, and further influencing the HPA axis. The increased HPA axis and sympathetic activity exert negative effects along the HPG axis. Ghrelin has also an inhibitory and indirect effect on kisspeptin-stimulated LH release, and is able to act directly at the level of the pituitary influencing gonadotropin secretion. Within the ovary and the testis, ghrelin has predominantly an inhibitory effect on steroidogenesis. Adapted from Servier Medical Art under Creative Commons CC-BY license.
Figure 6.

Ghrelin regulates the effects of stress on fertility. Exposure to stress involves the coordinated interaction between the sympathomedullary system, the HPA axis, the HPG axis, and the ghrelin system. Ghrelin regulates the stress response by acting indirectly on CRH neurons in the PVN and directly at the anterior pituitary gland to facilitate ACTH release. Ghrelin also binds to catecholaminergic neurons in locus coeruleus (LC) increasing noradrenaline production, and further influencing the HPA axis. The increased HPA axis and sympathetic activity exert negative effects along the HPG axis. Ghrelin has also an inhibitory and indirect effect on kisspeptin-stimulated LH release, and is able to act directly at the level of the pituitary influencing gonadotropin secretion. Within the ovary and the testis, ghrelin has predominantly an inhibitory effect on steroidogenesis. Adapted from Servier Medical Art under Creative Commons CC-BY license.

Total ghrelin levels that are typically reduced in obese individuals (273, 471, 537) are also reduced in women with PCOS, as has been demonstrated in several studies (538541). There is a further reduction in ghrelin levels in obese PCOS as compared with lean PCOS patients (542, 543), or obese controls (544, 545). Some studies, however, have not found these differences (546, 547). A negative correlation has been demonstrated between total ghrelin levels and insulin in obese women with PCOS (544, 545, 548). However, Schöfl et al. (549) has found this relationship to be present only in patients with insulin-sensitive, but not in patients with insulin-resistant PCOS who displayed very low fasting ghrelin levels (549), similar to obese insulin-resistant individuals (550). Metformin treatment, a common therapy of choice in insulin-resistant women with PCOS, significantly increases circulating ghrelin levels (549). An inverse correlation between ghrelin and androgen levels has also been demonstrated (538, 541, 544, 545, 551), as well as evidence of a negative relationship between hirsutism and ghrelin levels (544). Treatment with antiandrogen drugs or oral contraceptives increases plasma ghrelin (552, 553). Although normalization of androgen levels also improves insulin sensitivity, changes in plasma ghrelin concentrations are mainly dependent on the declining androgen levels and not on changes in insulin (552), suggesting that androgens may play role in the regulation of ghrelin production in PCOS. Because increased ghrelin levels typically inhibit LH release (217, 219, 220), it is also possible that lower levels of ghrelin in patients with PCOS promote hypersecretion of LH, a characteristic feature of PCOS (554), leading to an increase in ovarian androgen production (470). Interestingly, one study has examined the psychological parameters and emotional state in patients with PCOS and their relation to total ghrelin levels (555). Unlike in other studies, this group has demonstrated that lean women with PCOS exhibit higher ghrelin levels as compared with their weight-matched controls (555, 556), whereas in obese patients with PCOS ghrelin is reduced (555). Moreover, Komarowska et al. (555) has shown that lean patients with PCOS exhibit poor resistance to stress and significantly increased ACTH levels, compared with obese patients with PCOS, and that overall anxiety state is positively correlated with total ghrelin levels in patients with PCOS. These latter data emphasize the complexity and the heterogeneous phenotype of this disorder, and the need for further evaluation of the role that ghrelin may play in regulating both the etiology of PCOS and the patients’ emotional state.

Ghrelin’s role in energy deficiency and reproduction

Sufficient and adequate energy reserves are required for reproduction. It is therefore not surprising that different states of metabolic imbalance, ranging from energy insufficiency to obesity, often lead to reproductive dysfunction. As a major orexigenic hormone that regulates energy homeostasis, ghrelin, has been suggested to mediate the influences of altered nutritional states on reproductive health (557). Although circulating ghrelin levels are decreased in obese patients and increased in patients with anorexia nervosa (558), both conditions contribute to ghrelin resistance (559).

As we discussed previously, obesity is an important contributing risk factor to the development of PCOS, as well as to the development of pregnancy complications. Anorexia nervosa is also associated with reproductive issues, such as amenorrhea (560). Although despite menstrual irregularities women with anorexia nervosa may become pregnant, the physiological and psychological demands of pregnancy and motherhood present a substantial challenge for women who struggle with anorexia (561). Animal models of undernutrition have also shown a decrease in the expression of hypothalamic kisspeptin and delayed puberty onset (562), as well as a diminished ovarian reserve, when an exposure to suboptimal nutritional environment occurs early in life (563, 564).

Ghrelin levels are increased in women with anorexia nervosa and exercise-induced amenorrhea (335, 337, 565). In both anorexia and obesity, ghrelin levels are inversely correlated with body mass index (273, 566, 567). Therefore, increased ghrelin levels in the state of chronic energy deficiency may act as a compensatory mechanism in an attempt to increase food intake and normalize energy homeostasis, as well as to inhibit HPG axis signaling, both centrally and peripherally, until energy reserves are restored.

In rats, food deprivation has also been shown to be associated with increased noradrenergic release in the PVN and elevated plasma ACTH levels, whereas intracerebroventricular administration of AG further enhanced these effects (568). These findings further support the role of ghrelin in mediating neuroendocrine responses to stress, and suggest that increased ghrelin levels in the state of energy deficiency enhance stress responsivity. Psychological stress, in turn, induces an increase in circulating ghrelin, specifically in AG, but not DAG, and this increase has been shown to have both anxiolytic and anxiogenic effects [reviewed in (250)]. These differences are likely to be related to the duration of stress (212, 569). AG release in situations of acute stress is likely to play an adaptive role, helping an individual to cope with stress (113). However, prolonged exposure to increased AG has overall negative impacts on psycho-physiological state (121, 266, 268, 280), and as we propose herein, this stress-induced increase in AG is detrimental to reproductive function.

Ghrelin’s role in stress-induced reproductive dysfunction

As has been demonstrated with ghr−/− animals and with cases of infertility despite normal ghrelin activity, ghrelin is not strictly necessary (under nonstressed conditions) nor sufficient for reproduction. However, ghrelin modulates the stress response at almost all levels of the HPA and SAM axes, strongly influences reward, and is also an important player in successful functioning of the HPG axis, ovarian follicle maturation, and spermatogenesis. We therefore conclude ghrelin plays a substantial role linking stress with infertility.

The evidence we presented previously describes ghrelin’s role in integrating stress responsivity, as well as its regulatory role in reproductive function. The role of ghrelin signaling in both of these neuroendocrine systems is undoubtedly complex and is intertwined with its role in energy homeostasis. Interestingly, although the hypothalamus is the main site of ghrelin’s metabolic, stress-related, and reproductive actions, these effects appear to be regionally dissociated. Both the arcuate nucleus and PVN mediate the orexigenic effects of centrally administered AG (115, 297, 570), and these regions are also responsive to its anxiogenic effects (571). However, although intact arcuate nucleus signaling is essential for the metabolic actions of peripheral ghrelin (297, 572), ghrelin-induced activation of PVN CRH neurons is independent of the arcuate nucleus (114, 266, 268, 571). CRH neuronal signaling is also implicated in the central reproductive actions of ghrelin, because CRH antagonist has been shown to prevent the inhibitory effects of AG on LH pulsatility in nonhuman primates (261). However, because PVN CRH neurons are not directly involved in the suppression of LH pulsatility (109, 110, 112), these CRH-mediated effects of AG are likely be conveyed by CRH-GnRH connectivity in the mPOA (107), where AG exerts inhibitory (indirect) effects on Kiss1 neurons (116). In the periphery, administration of AG induces an increase in ACTH and glucocorticoid levels in animals and humans (258261), and these inhibit reproductive function at all sites of the reproductive axis as we have discussed. It is therefore plausible that in response to stress, both AG and HPA axis hormones act synergistically to suppress reproduction. It is important to incorporate in this regard the role of ghrelin in reward, including in the rewarding aspects of mating. Acutely AG has been demonstrated to enhance sexual motivation and behavior in male mice (303, 329). However, in female mice, chronic calorie restriction, typically associated with increased AG, reduces sexual receptivity and this is reversed by administration of the GHSR antagonist (573), suggesting that persistent elevation of AG induced by chronic stress may also inhibit sexual motivation and behavior.

The ghrelin system is also critically involved in the regulation of pregnancy and fetal development, and although the evidence is limited, it appears that increased levels of AG, including those induced by chronic stress, can be transferred to the fetus, programming an anxietylike phenotype in the offspring (468). Ghrelin deficiency during pregnancy, however, also produces detrimental developmental outcomes (405), suggesting that balanced ghrelin levels are required to maintain healthy pregnancy and improve fetal outcomes. Altered levels of ghrelin have also been associated with complications of pregnancy and other reproductive disorders in humans [reviewed in (469, 470)], and although further research using experimental conditions that induce alterations in ghrelin levels is required, we propose that chronic stress may significantly contribute to these reproductive disorders, via an imbalance in the availability of ghrelin.

Current Limitations and Future Directions

It is important to note that although the evidence that we presented in this review strongly supports our hypothesis that stress negatively impacts fertility and pregnancy by stimulating dysregulation in ghrelin signaling, a direct role for ghrelin as a mediator between stress and fertility has not yet been shown. Therefore, much remains to be elucidated in regards to the mechanisms involved in the integration of stress, ghrelin, and reproductive function, and with respect to potential therapeutic implications of these discoveries.

Future experiments addressing this role will need to be mindful of the potentially independent, complementary, and antagonistic role of AG and DAG, as indicated by the research looking into the distinct roles of AG and DAG in metabolism (249, 574577), neuroprotection, and cerebrovascular function (207, 211, 578, 579), as well as stress and anxiety (113, 251). Unfortunately, the vast majority of studies investigating the role of ghrelin in reproduction that we have presented in this review do not specify the acylation status of ghrelin.

Future studies will also need to assess the potential for commercially available pharmacological compounds targeting the availability of AG and DAG to remedy stress-induced infertility. Encouragingly, recently developed bioactive DAG analog, AZP-531 (Alizé Pharma, France), is a potent inhibitor of circulating AG (574, 580). This compound is currently undergoing clinical trials in patients with type 2 diabetes and in patients with Prader-Willi syndrome, who suffer from elevated AG and dysregulation of the ghrelin system. Thus far, AZP-531 has been shown to be safe, well tolerated, and its improved pharmacokinetic profile differentiates it from existing ghrelin antagonists (581). This compound may therefore provide a useful therapeutic in the context chronic stress-induced infertility. However, due to the involvement of ghrelin in multiple functions, extensive research is required to address the possibility of safely utilizing this compound in improving fertility. Nonetheless, the clear evidence of ghrelin’s role in stress and fertility suggest targeting its action may provide a useful therapeutic to remedy infertility in some stress-susceptible couples.

Abbreviations:

     
  • ACTH

    adrenocorticotropic hormone

  •  
  • AG

    acylated ghrelin

  •  
  • CDGP

    constitutional delay of growth and puberty

  •  
  • CRH

    corticotropin-releasing hormone

  •  
  • DAG

    des-acylated ghrelin

  •  
  • FSH

    follicle-stimulating hormone

  •  
  • GABA

    γ-aminobutyric acid

  •  
  • GDM

    gestational diabetes mellitus

  •  
  • GHSR

    growth hormone secretagogue receptor

  •  
  • GnRH

    gonadotropin-releasing hormone

  •  
  • GOAT

    ghrelin-O-acyltransferase

  •  
  • HPA

    hypothalamic-pituitary-adrenal

  •  
  • HPG

    hypothalamic-pituitary-gonadal

  •  
  • LH

    luteinizing hormone

  •  
  • mPOA

    medial preoptic area

  •  
  • PCOS

    polycystic ovarian syndrome

  •  
  • PVN

    paraventricular nucleus of the hypothalamus.

Acknowledgments

Financial Support: This work is supported by a Discovery Project Grant from the Australian Research Council (ARC) to S.J.S. (DP130100508). S.J.S. is an ARC Future Fellow (FT110100084) and an RMIT University VC Senior Research Fellow.

Disclosure Summary: The authors have nothing to disclose.

References

1.

Mascarenhas
MN
,
Flaxman
SR
,
Boerma
T
,
Vanderpoel
S
,
Stevens
GA
.
National, regional, and global trends in infertility prevalence since 1990: a systematic analysis of 277 health surveys
.
PLoS Med
.
2012
;
9
(
12
):
e1001356
.

2.

Inhorn
MC
,
Patrizio
P
.
Infertility around the globe: new thinking on gender, reproductive technologies and global movements in the 21st century
.
Hum Reprod Update
.
2015
;
21
(
4
):
411
426
.

3.

Brugo-Olmedo
S
,
Chillik
C
,
Kopelman
S
.
Definition and causes of infertility
.
Reprod Biomed Online
.
2001
;
2
(
1
):
41
53
.

4.

Gelbaya
TA
,
Potdar
N
,
Jeve
YB
,
Nardo
LG
.
Definition and epidemiology of unexplained infertility
.
Obstet Gynecol Surv
.
2014
;
69
(
2
):
109
115
.

5.

Brugh
III
VM
,
Lipshultz
LI
.
Male factor infertility: evaluation and management
.
Med Clin North Am
.
2004
;
88
(
2
):
367
385
.

6.

Walker
AK
,
Hiles
SA
,
Sominsky
L
,
McLaughlin
EA
,
Hodgson
DM
.
Neonatal lipopolysaccharide exposure impairs sexual development and reproductive success in the Wistar rat
.
Brain Behav Immun
.
2011
;
25
(
4
):
674
684
.

7.

Gao
Y
,
Chen
F
,
Kong
QQ
,
Ning
SF
,
Yuan
HJ
,
Lian
HY
,
Luo
MJ
,
Tan
JH
.
Stresses on female mice impair oocyte developmental potential: effects of stress severity and duration on oocytes at the growing follicle stage
.
Reprod Sci
.
2016
;
23
(
9
):
1148
1157
.

8.

Wu
LM
,
Hu
MH
,
Tong
XH
,
Han
H
,
Shen
N
,
Jin
RT
,
Wang
W
,
Zhou
GX
,
He
GP
,
Liu
YS
.
Chronic unpredictable stress decreases expression of brain-derived neurotrophic factor (BDNF) in mouse ovaries: relationship to oocytes developmental potential
.
PLoS One
.
2012
;
7
(
12
):
e52331
.

9.

Wu
LM
,
Liu
YS
,
Tong
XH
,
Shen
N
,
Jin
RT
,
Han
H
,
Hu
MH
,
Wang
W
,
Zhou
GX
.
Inhibition of follicular development induced by chronic unpredictable stress is associated with growth and differentiation factor 9 and gonadotropin in mice
.
Biol Reprod
.
2012
;
86
(
4
):
121
.

10.

Hou
G
,
Xiong
W
,
Wang
M
,
Chen
X
,
Yuan
TF
.
Chronic stress influences sexual motivation and causes damage to testicular cells in male rats
.
J Sex Med
.
2014
;
11
(
3
):
653
663
.

11.

von Borell
E
,
Dobson
H
,
Prunier
A
.
Stress, behaviour and reproductive performance in female cattle and pigs
.
Horm Behav
.
2007
;
52
(
1
):
130
138
.

12.

Michopoulos
V
,
Berga
SL
,
Kaplan
JR
,
Wilson
ME
.
Social subordination and polymorphisms in the gene encoding the serotonin transporter enhance estradiol inhibition of luteinizing hormone secretion in female rhesus monkeys
.
Biol Reprod
.
2009
;
81
(
6
):
1154
1163
.

13.

Nepomnaschy
PA
,
Welch
KB
,
McConnell
DS
,
Low
BS
,
Strassmann
BI
,
England
BG
.
Cortisol levels and very early pregnancy loss in humans
.
Proc Natl Acad Sci USA
.
2006
;
103
(
10
):
3938
3942
.

14.

Pfaus
JG
.
Pathways of sexual desire
.
J Sex Med
.
2009
;
6
(
6
):
1506
1533
.

15.

Gollenberg
AL
,
Liu
F
,
Brazil
C
,
Drobnis
EZ
,
Guzick
D
,
Overstreet
JW
,
Redmon
JB
,
Sparks
A
,
Wang
C
,
Swan
SH
.
Semen quality in fertile men in relation to psychosocial stress
.
Fertil Steril
.
2010
;
93
(
4
):
1104
1111
.

16.

Young
AJ
,
Carlson
AA
,
Monfort
SL
,
Russell
AF
,
Bennett
NC
,
Clutton-Brock
T
.
Stress and the suppression of subordinate reproduction in cooperatively breeding meerkats
.
Proc Natl Acad Sci USA
.
2006
;
103
(
32
):
12005
12010
.

17.

Wischmann
TH
.
Psychogenic infertility--myths and facts
.
J Assist Reprod Genet
.
2003
;
20
(
12
):
485
494
.

18.

Greil
AL
,
Slauson-Blevins
K
,
McQuillan
J
.
The experience of infertility: a review of recent literature
.
Sociol Health Illn
.
2010
;
32
(
1
):
140
162
.

19.

Anderson
K
,
Nisenblat
V
,
Norman
R
.
Lifestyle factors in people seeking infertility treatment - a review
.
Aust N Z J Obstet Gynaecol
.
2010
;
50
(
1
):
8
20
.

20.

Nepomnaschy
PA
,
Sheiner
E
,
Mastorakos
G
,
Arck
PC
.
Stress, immune function, and women’s reproduction
.
Ann N Y Acad Sci
.
2007
;
1113
:
350
364
.

21.

Domar
AD
,
Clapp
D
,
Slawsby
EA
,
Dusek
J
,
Kessel
B
,
Freizinger
M
.
Impact of group psychological interventions on pregnancy rates in infertile women
.
Fertil Steril
.
2000
;
73
(
4
):
805
811
.

22.

Zorn
B
,
Auger
J
,
Velikonja
V
,
Kolbezen
M
,
Meden-Vrtovec
H
.
Psychological factors in male partners of infertile couples: relationship with semen quality and early miscarriage
.
Int J Androl
.
2008
;
31
(
6
):
557
564
.

23.

Gourounti
K
,
Anagnostopoulos
F
,
Vaslamatzis
G
.
The relation of psychological stress to pregnancy outcome among women undergoing in-vitro fertilization and intracytoplasmic sperm injection
.
Women Health
.
2011
;
51
(
4
):
321
339
.

24.

Klonoff-Cohen
H
,
Chu
E
,
Natarajan
L
,
Sieber
W
.
A prospective study of stress among women undergoing in vitro fertilization or gamete intrafallopian transfer
.
Fertil Steril
.
2001
;
76
(
4
):
675
687
.

25.

Catherino
WH
.
Stress relief to augment fertility: the pressure mounts
.
Fertil Steril
.
2011
;
95
(
8
):
2462
2463
.

26.

Louis
GM
,
Lum
KJ
,
Sundaram
R
,
Chen
Z
,
Kim
S
,
Lynch
CD
,
Schisterman
EF
,
Pyper
C
.
Stress reduces conception probabilities across the fertile window: evidence in support of relaxation
.
Fertil Steril
.
2011
;
95
(
7
):
2184
2189
.

27.

Lynch
CD
,
Sundaram
R
,
Maisog
JM
,
Sweeney
AM
,
Buck Louis
GM
.
Preconception stress increases the risk of infertility: results from a couple-based prospective cohort study--the LIFE study
.
Hum Reprod
.
2014
;
29
(
5
):
1067
1075
.

28.

Boivin
J
,
Schmidt
L
.
Infertility-related stress in men and women predicts treatment outcome 1 year later
.
Fertil Steril
.
2005
;
83
(
6
):
1745
1752
.

29.

Schliep
KC
,
Mumford
SL
,
Vladutiu
CJ
,
Ahrens
KA
,
Perkins
NJ
,
Sjaarda
LA
,
Kissell
KA
,
Prasad
A
,
Wactawski-Wende
J
,
Schisterman
EF
.
Perceived stress, reproductive hormones, and ovulatory function: a prospective cohort study
.
Epidemiology
.
2015
;
26
(
2
):
177
184
.

30.

Ebbesen
SM
,
Zachariae
R
,
Mehlsen
MY
,
Thomsen
D
,
Højgaard
A
,
Ottosen
L
,
Petersen
T
,
Ingerslev
HJ
.
Stressful life events are associated with a poor in-vitro fertilization (IVF) outcome: a prospective study
.
Hum Reprod
.
2009
;
24
(
9
):
2173
2182
.

31.

Nepomnaschy
PA
,
Welch
K
,
McConnell
D
,
Strassmann
BI
,
England
BG
.
Stress and female reproductive function: a study of daily variations in cortisol, gonadotrophins, and gonadal steroids in a rural Mayan population
.
Am J Hum Biol
.
2004
;
16
(
5
):
523
532
.

32.

Donarelli
Z
,
Lo Coco
G
,
Gullo
S
,
Marino
A
,
Volpes
A
,
Salerno
L
,
Allegra
A
.
Infertility-related stress, anxiety and ovarian stimulation: can couples be reassured about the effects of psychological factors on biological responses to assisted reproductive technology?
Reprod Biomed Soc Online
.
2016
;
3
:
16
23
.

33.

Matthiesen
SM
,
Frederiksen
Y
,
Ingerslev
HJ
,
Zachariae
R
.
Stress, distress and outcome of assisted reproductive technology (ART): a meta-analysis
.
Hum Reprod
.
2011
;
26
(
10
):
2763
2776
.

34.

Boivin
J
,
Griffiths
E
,
Venetis
CA
.
Emotional distress in infertile women and failure of assisted reproductive technologies: meta-analysis of prospective psychosocial studies
.
BMJ
.
2011
;
342
:
d223
.

35.

Frederiksen
Y
,
Farver-Vestergaard
I
,
Skovgård
NG
,
Ingerslev
HJ
,
Zachariae
R
.
Efficacy of psychosocial interventions for psychological and pregnancy outcomes in infertile women and men: a systematic review and meta-analysis
.
BMJ Open
.
2015
;
5
(
1
):
e006592
.

36.

Lintsen
AM
,
Verhaak
CM
,
Eijkemans
MJ
,
Smeenk
JM
,
Braat
DD
.
Anxiety and depression have no influence on the cancellation and pregnancy rates of a first IVF or ICSI treatment
.
Hum Reprod
.
2009
;
24
(
5
):
1092
1098
.

37.

Nouri
K
,
Litschauer
B
,
Huber
JC
,
Buerkle
B
,
Tiringer
D
,
Tempfer
CB
.
Saliva cortisol levels and subjective stress are not associated with number of oocytes after controlled ovarian hyperstimulation in patients undergoing in vitro fertilization
.
Fertil Steril
.
2011
;
96
(
1
):
69
72
.

38.

Hjollund
NH
,
Bonde
JP
,
Henriksen
TB
,
Giwercman
A
,
Olsen
J
;
Danish First Pregnancy Planner Study Team
.
Reproductive effects of male psychologic stress
.
Epidemiology
.
2004
;
15
(
1
):
21
27
.

39.

Campagne
DM
.
Should fertilization treatment start with reducing stress?
Hum Reprod
.
2006
;
21
(
7
):
1651
1658
.

40.

Quant
HS
,
Zapantis
A
,
Nihsen
M
,
Bevilacqua
K
,
Jindal
S
,
Pal
L
.
Reproductive implications of psychological distress for couples undergoing IVF
.
J Assist Reprod Genet
.
2013
;
30
(
11
):
1451
1458
.

41.

Turner
K
,
Reynolds-May
MF
,
Zitek
EM
,
Tisdale
RL
,
Carlisle
AB
,
Westphal
LM
.
Stress and anxiety scores in first and repeat IVF cycles: a pilot study
.
PLoS One
.
2013
;
8
(
5
):
e63743
.

42.

Olivius
C
,
Friden
B
,
Borg
G
,
Bergh
C
.
Why do couples discontinue in vitro fertilization treatment? A cohort study
.
Fertil Steril
.
2004
;
81
(
2
):
258
261
.

43.

Smeenk
JM
,
Verhaak
CM
,
Stolwijk
AM
,
Kremer
JA
,
Braat
DD
.
Reasons for dropout in an in vitro fertilization/intracytoplasmic sperm injection program
.
Fertil Steril
.
2004
;
81
(
2
):
262
268
.

44.

Rajkhowa
M
,
McConnell
A
,
Thomas
GE
.
Reasons for discontinuation of IVF treatment: a questionnaire study
.
Hum Reprod
.
2006
;
21
(
2
):
358
363
.

45.

Maheshwari
A
,
Hamilton
M
,
Bhattacharya
S
.
Effect of female age on the diagnostic categories of infertility
.
Hum Reprod
.
2008
;
23
(
3
):
538
542
.

46.

Demyttenaere
K
,
Nijs
P
,
Evers-Kiebooms
G
,
Koninckx
PR
.
Coping and the ineffectiveness of coping influence the outcome of in vitro fertilization through stress responses
.
Psychoneuroendocrinology
.
1992
;
17
(
6
):
655
665
.

47.

Thiering
P
,
Beaurepaire
J
,
Jones
M
,
Saunders
D
,
Tennant
C
.
Mood state as a predictor of treatment outcome after in vitro fertilization/embryo transfer technology (IVF/ET)
.
J Psychosom Res
.
1993
;
37
(
5
):
481
491
.

48.

Smeenk
JM
,
Verhaak
CM
,
Eugster
A
,
van Minnen
A
,
Zielhuis
GA
,
Braat
DD
.
The effect of anxiety and depression on the outcome of in-vitro fertilization
.
Hum Reprod
.
2001
;
16
(
7
):
1420
1423
.

49.

Verhaak
CM
,
Smeenk
JM
,
Eugster
A
,
van Minnen
A
,
Kremer
JA
,
Kraaimaat
FW
.
Stress and marital satisfaction among women before and after their first cycle of in vitro fertilization and intracytoplasmic sperm injection
.
Fertil Steril
.
2001
;
76
(
3
):
525
531
.

50.

Eugster
A
,
Vingerhoets
AJ
,
van Heck
GL
,
Merkus
JM
.
The effect of episodic anxiety on an in vitro fertilization and intracytoplasmic sperm injection treatment outcome: a pilot study
.
J Psychosom Obstet Gynaecol
.
2004
;
25
(
1
):
57
65
.

51.

Merari
D
,
Feldberg
D
,
Elizur
A
,
Goldman
J
,
Modan
B
.
Psychological and hormonal changes in the course of in vitro fertilization
.
J Assist Reprod Genet
.
1992
;
9
(
2
):
161
169
.

52.

Boivin
J
,
Takefman
JE
.
Stress level across stages of in vitro fertilization in subsequently pregnant and nonpregnant women
.
Fertil Steril
.
1995
;
64
(
4
):
802
810
.

53.

Anderheim
L
,
Holter
H
,
Bergh
C
,
Möller
A
.
Does psychological stress affect the outcome of in vitro fertilization?
Hum Reprod
.
2005
;
20
(
10
):
2969
2975
.

54.

de Klerk
C
,
Hunfeld
JA
,
Heijnen
EM
,
Eijkemans
MJ
,
Fauser
BC
,
Passchier
J
,
Macklon
NS
.
Low negative affect prior to treatment is associated with a decreased chance of live birth from a first IVF cycle
.
Hum Reprod
.
2008
;
23
(
1
):
112
116
.

55.

Hämmerli
K
,
Znoj
H
,
Barth
J
.
The efficacy of psychological interventions for infertile patients: a meta-analysis examining mental health and pregnancy rate
.
Hum Reprod Update
.
2009
;
15
(
3
):
279
295
.

56.

de Liz
TM
,
Strauss
B
.
Differential efficacy of group and individual/couple psychotherapy with infertile patients
.
Hum Reprod
.
2005
;
20
(
5
):
1324
1332
.

58.

Gameiro
S
,
Boivin
J
,
Dancet
E
,
de Klerk
C
,
Emery
M
,
Lewis-Jones
C
,
Thorn
P
,
Van den Broeck
U
,
Venetis
C
,
Verhaak
CM
,
Wischmann
T
,
Vermeulen
N
.
ESHRE guideline: routine psychosocial care in infertility and medically assisted reproduction-a guide for fertility staff
.
Hum Reprod
.
2015
;
30
(
11
):
2476
2485
.

59.

Boivin
J
.
A review of psychosocial interventions in infertility
.
Soc Sci Med
.
2003
;
57
(
12
):
2325
2341
.

60.

Domar
AD
,
Gross
J
,
Rooney
K
,
Boivin
J
.
Exploratory randomized trial on the effect of a brief psychological intervention on emotions, quality of life, discontinuation, and pregnancy rates in in vitro fertilization patients
.
Fertil Steril
.
2015
;
104
(
2
):
440
451
.

61.

Cannon
WB
.
Stresses and strains of homeostasis
.
Am J Med Sci
.
1935
;
189
(
1
):
13
14
.

62.

Cannon
WB
.
Bodily Changes in Pain, Hunger, Fear and Rage: An Account of Recent Researches Into the Function of Emotional Excitement
. 2nd ed.
New York, NY, and London, England
:
D. Appleton and Company
;
1929
.

63.

Selye
H
.
A syndrome produced by diverse nocuous agents. 1936
.
J Neuropsychiatry Clin Neurosci
.
1998
;
10
(
2
):
230
231
.

64.

Selye
H
.
The general adaptation syndrome and the diseases of adaptation
.
J Clin Endocrinol Metab
.
1946
;
6
:
117
230
.

65.

Day
TA
.
Defining stress as a prelude to mapping its neurocircuitry: no help from allostasis
.
Prog Neuropsychopharmacol Biol Psychiatry
.
2005
;
29
(
8
):
1195
1200
.

66.

Dayas
CV
,
Buller
KM
,
Day
TA
.
Neuroendocrine responses to an emotional stressor: evidence for involvement of the medial but not the central amygdala
.
Eur J Neurosci
.
1999
;
11
(
7
):
2312
2322
.

67.

Dayas
CV
,
Buller
KM
,
Crane
JW
,
Xu
Y
,
Day
TA
.
Stressor categorization: acute physical and psychological stressors elicit distinctive recruitment patterns in the amygdala and in medullary noradrenergic cell groups
.
Eur J Neurosci
.
2001
;
14
(
7
):
1143
1152
.

68.

Herman
JP
,
Figueiredo
H
,
Mueller
NK
,
Ulrich-Lai
Y
,
Ostrander
MM
,
Choi
DC
,
Cullinan
WE
.
Central mechanisms of stress integration: hierarchical circuitry controlling hypothalamo-pituitary-adrenocortical responsiveness
.
Front Neuroendocrinol
.
2003
;
24
(
3
):
151
180
.

69.

Sapolsky
RM
,
Romero
LM
,
Munck
AU
.
How do glucocorticoids influence stress responses? Integrating permissive, suppressive, stimulatory, and preparative actions
.
Endocr Rev
.
2000
;
21
(
1
):
55
89
.

70.

Windle
RJ
,
Wood
SA
,
Shanks
N
,
Lightman
SL
,
Ingram
CD
.
Ultradian rhythm of basal corticosterone release in the female rat: dynamic interaction with the response to acute stress
.
Endocrinology
.
1998
;
139
(
2
):
443
450
.

71.

Smith
R
,
Grossman
A
,
Gaillard
R
,
Clement-Jones
V
,
Ratter
S
,
Mallinson
J
,
Lowry
PJ
,
Besser
GM
,
Rees
LH
.
Studies on circulating met-enkephalin and beta-endorphin: normal subjects and patients with renal and adrenal disease
.
Clin Endocrinol (Oxf)
.
1981
;
15
(
3
):
291
300
.

72.

Cai
G
,
Ziko
I
,
Barwood
J
,
Soch
A
,
Sominsky
L
,
Molero
JC
,
Spencer
SJ
.
Overfeeding during a critical postnatal period exacerbates hypothalamic-pituitary-adrenal axis responses to immune challenge: a role for adrenal melanocortin 2 receptors
.
Sci Rep
.
2016
;
6
:
21097
.

73.

Spencer
SJ
,
Tilbrook
A
.
The glucocorticoid contribution to obesity
.
Stress
.
2011
;
14
(
3
):
233
246
.

74.

Papadimitriou
A
,
Priftis
KN
.
Regulation of the hypothalamic-pituitary-adrenal axis
.
Neuroimmunomodulation
.
2009
;
16
(
5
):
265
271
.

75.

Sapolsky
RM
.
Glucocorticoids and hippocampal atrophy in neuropsychiatric disorders
.
Arch Gen Psychiatry
.
2000
;
57
(
10
):
925
935
.

76.

Walker
JJ
,
Terry
JR
,
Lightman
SL
.
Origin of ultradian pulsatility in the hypothalamic-pituitary-adrenal axis
.
Proc Biol Sci
.
2010
;
277
(
1688
):
1627
1633
.

77.

Sominsky
L
,
Meehan
CL
,
Walker
AK
,
Bobrovskaya
L
,
McLaughlin
EA
,
Hodgson
DM
.
Neonatal immune challenge alters reproductive development in the female rat
.
Horm Behav
.
2012
;
62
(
3
):
345
355
.

78.

Krsmanovic
LZ
,
Hu
L
,
Leung
P-K
,
Feng
H
,
Catt
KJ
.
The hypothalamic GnRH pulse generator: multiple regulatory mechanisms
.
Trends Endocrinol Metab
.
2009
;
20
(
8
):
402
408
.

79.

Ohkura
S
,
Uenoyama
Y
,
Yamada
S
,
Homma
T
,
Takase
K
,
Inoue
N
,
Maeda
K
,
Tsukamura
H
.
Physiological role of metastin/kisspeptin in regulating gonadotropin-releasing hormone (GnRH) secretion in female rats
.
Peptides
.
2009
;
30
(
1
):
49
56
.

80.

Krsmanovic
LZ
,
Stojilkovic
SS
,
Catt
KJ
.
Pulsatile gonadotropin-releasing hormone release and its regulation
.
Trends Endocrinol Metab
.
1996
;
7
(
2
):
56
59
.

81.

Vitalis
EA
,
Costantin
JL
,
Tsai
PS
,
Sakakibara
H
,
Paruthiyil
S
,
Iiri
T
,
Martini
JF
,
Taga
M
,
Choi
AL
,
Charles
AC
,
Weiner
RI
.
Role of the cAMP signaling pathway in the regulation of gonadotropin-releasing hormone secretion in GT1 cells
.
Proc Natl Acad Sci USA
.
2000
;
97
(
4
):
1861
1866
.

82.

Knobil
E
.
The hypothalamic gonadotrophic hormone releasing hormone (GnRH) pulse generator in the rhesus monkey and its neuroendocrine control
.
Hum Reprod
.
1988
;
3
(
1
):
29
31
.

83.

Krsmanović
LZ
,
Stojilković
SS
,
Mertz
LM
,
Tomić
M
,
Catt
KJ
.
Expression of gonadotropin-releasing hormone receptors and autocrine regulation of neuropeptide release in immortalized hypothalamic neurons
.
Proc Natl Acad Sci USA
.
1993
;
90
(
9
):
3908
3912
.

84.

Krsmanovic
LZ
,
Mores
N
,
Navarro
CE
,
Arora
KK
,
Catt
KJ
.
An agonist-induced switch in G protein coupling of the gonadotropin-releasing hormone receptor regulates pulsatile neuropeptide secretion
.
Proc Natl Acad Sci USA
.
2003
;
100
(
5
):
2969
2974
.

85.

Irwig
MS
,
Fraley
GS
,
Smith
JT
,
Acohido
BV
,
Popa
SM
,
Cunningham
MJ
,
Gottsch
ML
,
Clifton
DK
,
Steiner
RA
.
Kisspeptin activation of gonadotropin releasing hormone neurons and regulation of KiSS-1 mRNA in the male rat
.
Neuroendocrinology
.
2004
;
80
(
4
):
264
272
.

86.

Hu
L
,
Gustofson
RL
,
Feng
H
,
Leung
PK
,
Mores
N
,
Krsmanovic
LZ
,
Catt
KJ
.
Converse regulatory functions of estrogen receptor-alpha and -beta subtypes expressed in hypothalamic gonadotropin-releasing hormone neurons
.
Mol Endocrinol
.
2008
;
22
(
10
):
2250
2259
.

87.

Millar
RP
.
GnRHs and GnRH receptors
.
Anim Reprod Sci
.
2005
;
88
(
1-2
):
5
28
.

88.

Vadakkadath Meethal
S
,
Atwood
CS
.
The role of hypothalamic-pituitary-gonadal hormones in the normal structure and functioning of the brain
.
Cell Mol Life Sci
.
2005
;
62
(
3
):
257
270
.

89.

Rivier
C
,
Rivest
S
.
Effect of stress on the activity of the hypothalamic-pituitary-gonadal axis: peripheral and central mechanisms
.
Biol Reprod
.
1991
;
45
(
4
):
523
532
.

90.

Tilbrook
AJ
,
Turner
AI
,
Clarke
IJ
.
Effects of stress on reproduction in non-rodent mammals: the role of glucocorticoids and sex differences
.
Rev Reprod
.
2000
;
5
(
2
):
105
113
.

91.

Tilbrook
AJ
,
Turner
AI
,
Clarke
IJ
.
Stress and reproduction: central mechanisms and sex differences in non-rodent species
.
Stress
.
2002
;
5
(
2
):
83
100
.

92.

Liu
JH
.
Hypothalamic amenorrhea: clinical perspectives, pathophysiology, and management
.
Am J Obstet Gynecol
.
1990
;
163
(
5 Pt 2
):
1732
1736
.

93.

Rivier
C
,
Vale
W
.
Influence of corticotropin-releasing factor on reproductive functions in the rat
.
Endocrinology
.
1984
;
114
(
3
):
914
921
.

94.

Tilbrook
AJ
,
Canny
BJ
,
Serapiglia
MD
,
Ambrose
TJ
,
Clarke
IJ
.
Suppression of the secretion of luteinizing hormone due to isolation/restraint stress in gonadectomised rams and ewes is influenced by sex steroids
.
J Endocrinol
.
1999
;
160
(
3
):
469
481
.

95.

Williams
CL
,
Nishihara
M
,
Thalabard
JC
,
Grosser
PM
,
Hotchkiss
J
,
Knobil
E
.
Corticotropin-releasing factor and gonadotropin-releasing hormone pulse generator activity in the rhesus monkey. Electrophysiological studies
.
Neuroendocrinology
.
1990
;
52
(
2
):
133
137
.

96.

Barbarino
A
,
De Marinis
L
,
Folli
G
,
Tofani
A
,
Della Casa
S
,
D’Amico
C
,
Mancini
A
,
Corsello
SM
,
Sambo
P
,
Barini
A
.
Corticotropin-releasing hormone inhibition of gonadotropin secretion during the menstrual cycle
.
Metabolism
.
1989
;
38
(
6
):
504
506
.

97.

Cates
PS
,
Li
XF
,
O’Byrne
KT
.
The influence of 17beta-oestradiol on corticotrophin-releasing hormone induced suppression of luteinising hormone pulses and the role of CRH in hypoglycaemic stress-induced suppression of pulsatile LH secretion in the female rat
.
Stress
.
2004
;
7
(
2
):
113
118
.

98.

Li
XF
,
Bowe
JE
,
Lightman
SL
,
O’Byrne
KT
.
Role of corticotropin-releasing factor receptor-2 in stress-induced suppression of pulsatile luteinizing hormone secretion in the rat
.
Endocrinology
.
2005
;
146
(
1
):
318
322
.

99.

Li
XF
,
Bowe
JE
,
Kinsey-Jones
JS
,
Brain
SD
,
Lightman
SL
,
O’Byrne
KT
.
Differential role of corticotrophin-releasing factor receptor types 1 and 2 in stress-induced suppression of pulsatile luteinising hormone secretion in the female rat
.
J Neuroendocrinol
.
2006
;
18
(
8
):
602
610
.

100.

Herod
SM
,
Pohl
CR
,
Cameron
JL
.
Treatment with a CRH-R1 antagonist prevents stress-induced suppression of the central neural drive to the reproductive axis in female macaques
.
Am J Physiol Endocrinol Metab
.
2011
;
300
(
1
):
E19
E27
.

101.

Xiao
E
,
Xia-Zhang
L
,
Vulliemoz
N
,
Rivier
J
,
Ferin
M
.
Astressin B, a corticotropin-releasing hormone receptor antagonist, accelerates the return to normal luteal function after an inflammatory-like stress challenge in the rhesus monkey
.
Endocrinology
.
2007
;
148
(
2
):
841
848
.

102.

Perrin
M
,
Donaldson
C
,
Chen
R
,
Blount
A
,
Berggren
T
,
Bilezikjian
L
,
Sawchenko
P
,
Vale
W
.
Identification of a second corticotropin-releasing factor receptor gene and characterization of a cDNA expressed in heart
.
Proc Natl Acad Sci USA
.
1995
;
92
(
7
):
2969
2973
.

103.

Van Pett
K
,
Viau
V
,
Bittencourt
JC
,
Chan
RK
,
Li
HY
,
Arias
C
,
Prins
GS
,
Perrin
M
,
Vale
W
,
Sawchenko
PE
.
Distribution of mRNAs encoding CRF receptors in brain and pituitary of rat and mouse
.
J Comp Neurol
.
2000
;
428
(
2
):
191
212
.

104.

Vale
W
,
Spiess
J
,
Rivier
C
,
Rivier
J
.
Characterization of a 41-residue ovine hypothalamic peptide that stimulates secretion of corticotropin and beta-endorphin
.
Science
.
1981
;
213
(
4514
):
1394
1397
.

105.

Traslaviña
GA
,
Franci
CR
.
Divergent roles of the CRH receptors in the control of gonadotropin secretion induced by acute restraint stress at proestrus
.
Endocrinology
.
2012
;
153
(
10
):
4838
4848
.

106.

MacLusky
NJ
,
Naftolin
F
,
Leranth
C
.
Immunocytochemical evidence for direct synaptic connections between corticotrophin-releasing factor (CRF) and gonadotrophin-releasing hormone (GnRH)-containing neurons in the preoptic area of the rat
.
Brain Res
.
1988
;
439
(
1-2
):
391
395
.

107.

Jasoni
CL
,
Todman
MG
,
Han
SK
,
Herbison
AE
.
Expression of mRNAs encoding receptors that mediate stress signals in gonadotropin-releasing hormone neurons of the mouse
.
Neuroendocrinology
.
2005
;
82
(
5-6
):
320
328
.

108.

Ferguson
AV
,
Latchford
KJ
,
Samson
WK
.
The paraventricular nucleus of the hypothalamus - a potential target for integrative treatment of autonomic dysfunction
.
Expert Opin Ther Targets
.
2008
;
12
(
6
):
717
727
.

109.

Rivest
S
,
Rivier
C
.
Influence of the paraventricular nucleus of the hypothalamus in the alteration of neuroendocrine functions induced by intermittent footshock or interleukin
.
Endocrinology
.
1991
;
129
(
4
):
2049
2057
.

110.

Hahn
JD
,
Kalamatianos
T
,
Coen
CW
.
Studies on the neuroanatomical basis for stress-induced oestrogen-potentiated suppression of reproductive function: evidence against direct corticotropin-releasing hormone projections to the vicinity of luteinizing hormone-releasing hormone cell bodies in female rats
.
J Neuroendocrinol
.
2003
;
15
(
8
):
732
742
.

111.

Li
XF
,
Bowe
JE
,
Mitchell
JC
,
Brain
SD
,
Lightman
SL
,
O’Byrne
KT
.
Stress-induced suppression of the gonadotropin-releasing hormone pulse generator in the female rat: a novel neural action for calcitonin gene-related peptide
.
Endocrinology
.
2004
;
145
(
4
):
1556
1563
.

112.

Li
XF
,
Kinsey-Jones
JS
,
Bowe
JE
,
Wilkinson
ES
,
Brain
SD
,
Lightman
SL
,
O’Byrne
KT
.
A role for the medial preoptic area in CGRP-induced suppression of pulsatile LH secretion in the female rat
.
Stress
.
2009
;
12
(
3
):
259
267
.

113.

Spencer
SJ
,
Xu
L
,
Clarke
MA
,
Lemus
M
,
Reichenbach
A
,
Geenen
B
,
Kozicz
T
,
Andrews
ZB
.
Ghrelin regulates the hypothalamic-pituitary-adrenal axis and restricts anxiety after acute stress
.
Biol Psychiatry
.
2012
;
72
(
6
):
457
465
.

114.

Cabral
A
,
Portiansky
E
,
Sánchez-Jaramillo
E
,
Zigman
JM
,
Perello
M
.
Ghrelin activates hypophysiotropic corticotropin-releasing factor neurons independently of the arcuate nucleus
.
Psychoneuroendocrinology
.
2016
;
67
:
27
39
.

115.

Currie
PJ
,
Mirza
A
,
Fuld
R
,
Park
D
,
Vasselli
JR
.
Ghrelin is an orexigenic and metabolic signaling peptide in the arcuate and paraventricular nuclei
.
Am J Physiol Regul Integr Comp Physiol
.
2005
;
289
(
2
):
R353
R358
.

116.

Forbes
S
,
Li
XF
,
Kinsey-Jones
J
,
O’Byrne
K
.
Effects of ghrelin on Kisspeptin mRNA expression in the hypothalamic medial preoptic area and pulsatile luteinising hormone secretion in the female rat
.
Neurosci Lett
.
2009
;
460
(
2
):
143
147
.

117.

Frazao
R
,
Dungan Lemko
HM
,
da Silva
RP
,
Ratra
DV
,
Lee
CE
,
Williams
KW
,
Zigman
JM
,
Elias
CF
.
Estradiol modulates Kiss1 neuronal response to ghrelin
.
Am J Physiol Endocrinol Metab
.
2014
;
306
(
6
):
E606
E614
.

118.

Shepard
JD
,
Barron
KW
,
Myers
DA
.
Corticosterone delivery to the amygdala increases corticotropin-releasing factor mRNA in the central amygdaloid nucleus and anxiety-like behavior
.
Brain Res
.
2000
;
861
(
2
):
288
295
.

119.

Keen-Rhinehart
E
,
Michopoulos
V
,
Toufexis
DJ
,
Martin
EI
,
Nair
H
,
Ressler
KJ
,
Davis
M
,
Owens
MJ
,
Nemeroff
CB
,
Wilson
ME
.
Continuous expression of corticotropin-releasing factor in the central nucleus of the amygdala emulates the dysregulation of the stress and reproductive axes
.
Mol Psychiatry
.
2009
;
14
(
1
):
37
50
.

120.

Lin
Y
,
Li
X
,
Lupi
M
,
Kinsey-Jones
JS
,
Shao
B
,
Lightman
SL
,
O’Byrne
KT
.
The role of the medial and central amygdala in stress-induced suppression of pulsatile LH secretion in female rats
.
Endocrinology
.
2011
;
152
(
2
):
545
555
.

121.

Meyer
RM
,
Burgos-Robles
A
,
Liu
E
,
Correia
SS
,
Goosens
KA
.
A ghrelin-growth hormone axis drives stress-induced vulnerability to enhanced fear
.
Mol Psychiatry
.
2014
;
19
(
12
):
1284
1294
.

122.

Herbison
AE
,
Chapman
C
,
Dyer
RG
.
Role of medial preoptic GABA neurones in regulating luteinising hormone secretion in the ovariectomised rat
.
Exp Brain Res
.
1991
;
87
(
2
):
345
352
.

123.

Bilger
M
,
Heger
S
,
Brann
DW
,
Paredes
A
,
Ojeda
SR
.
A conditional tetracycline-regulated increase in Gamma amino butyric acid production near luteinizing hormone-releasing hormone nerve terminals disrupts estrous cyclicity in the rat
.
Endocrinology
.
2001
;
142
(
5
):
2102
2114
.

124.

Scott
CJ
,
Clarke
IJ
.
Evidence that changes in the function of the subtypes of the receptors for gamma-amino butyric acid may be involved in the seasonal changes in the negative-feedback effects of estrogen on gonadotropin-releasing hormone secretion and plasma luteinizing hormone levels in the ewe
.
Endocrinology
.
1993
;
133
(
6
):
2904
2912
.

125.

Jarry
H
,
Perschl
A
,
Wuttke
W
.
Further evidence that preoptic anterior hypothalamic GABAergic neurons are part of the GnRH pulse and surge generator
.
Acta Endocrinol (Copenh)
.
1988
;
118
(
4
):
573
579
.

126.

Han
SK
,
Abraham
IM
,
Herbison
AE
.
Effect of GABA on GnRH neurons switches from depolarization to hyperpolarization at puberty in the female mouse
.
Endocrinology
.
2002
;
143
(
4
):
1459
1466
.

127.

Keen
KL
,
Burich
AJ
,
Mitsushima
D
,
Kasuya
E
,
Terasawa
E
.
Effects of pulsatile infusion of the GABA(A) receptor blocker bicuculline on the onset of puberty in female rhesus monkeys
.
Endocrinology
.
1999
;
140
(
11
):
5257
5266
.

128.

Li
XF
,
Lin
YS
,
Kinsey-Jones
JS
,
Milligan
SR
,
Lightman
SL
,
O’Byrne
KT
.
The role of the bed nucleus of the stria terminalis in stress-induced inhibition of pulsatile luteinising hormone secretion in the female rat
.
J Neuroendocrinol
.
2011
;
23
(
1
):
3
11
.

129.

Akema
T
,
He
D
,
Sugiyama
H
.
Lipopolysaccharide increases gamma-aminobutyric acid synthesis in medial preoptic neurones in association with inhibition of steroid-induced luteinising hormone surge in female rats
.
J Neuroendocrinol
.
2005
;
17
(
10
):
672
678
.

130.

Lin
YS
,
Li
XF
,
Shao
B
,
Hu
MH
,
Goundry
AL
,
Jeyaram
A
,
Lightman
SL
,
O’Byrne
KT
.
The role of GABAergic signalling in stress-induced suppression of gonadotrophin-releasing hormone pulse generator frequency in female rats
.
J Neuroendocrinol
.
2012
;
24
(
3
):
477
488
.

131.

Li
X
,
Shao
B
,
Lin
C
,
O’Byrne
KT
,
Lin
Y
.
Stress-induced inhibition of LH pulses in female rats: role of GABA in arcuate nucleus
.
J Mol Endocrinol
.
2015
;
55
(
1
):
9
19
.

132.

Gottsch
ML
,
Popa
SM
,
Lawhorn
JK
,
Qiu
J
,
Tonsfeldt
KJ
,
Bosch
MA
,
Kelly
MJ
,
Rønnekleiv
OK
,
Sanz
E
,
McKnight
GS
,
Clifton
DK
,
Palmiter
RD
,
Steiner
RA
.
Molecular properties of Kiss1 neurons in the arcuate nucleus of the mouse
.
Endocrinology
.
2011
;
152
(
11
):
4298
4309
.

133.

Li
XF
,
Kinsey-Jones
JS
,
Cheng
Y
,
Knox
AM
,
Lin
Y
,
Petrou
NA
,
Roseweir
A
,
Lightman
SL
,
Milligan
SR
,
Millar
RP
,
O’Byrne
KT
.
Kisspeptin signalling in the hypothalamic arcuate nucleus regulates GnRH pulse generator frequency in the rat
.
PLoS One
.
2009
;
4
(
12
):
e8334
.

134.

Smith
JT
.
Sex steroid regulation of kisspeptin circuits
.
Adv Exp Med Biol
.
2013
;
784
:
275
295
.

135.

Seminara
SB
,
Messager
S
,
Chatzidaki
EE
,
Thresher
RR
,
Acierno
JS
, Jr
,
Shagoury
JK
,
Bo-Abbas
Y
,
Kuohung
W
,
Schwinof
KM
,
Hendrick
AG
,
Zahn
D
,
Dixon
J
,
Kaiser
UB
,
Slaugenhaupt
SA
,
Gusella
JF
,
O’Rahilly
S
,
Carlton
MB
,
Crowley
WF
, Jr
,
Aparicio
SA
,
Colledge
WH
.
The GPR54 gene as a regulator of puberty
.
N Engl J Med
.
2003
;
349
(
17
):
1614
1627
.

136.

Smith
JT
,
Clay
CM
,
Caraty
A
,
Clarke
IJ
.
KiSS-1 messenger ribonucleic acid expression in the hypothalamus of the ewe is regulated by sex steroids and season
.
Endocrinology
.
2007
;
148
(
3
):
1150
1157
.

137.

Clarkson
J
,
d’Anglemont de Tassigny
X
,
Colledge
WH
,
Caraty
A
,
Herbison
AE
.
Distribution of kisspeptin neurones in the adult female mouse brain
.
J Neuroendocrinol
.
2009
;
21
(
8
):
673
682
.

138.

Herman
JP
,
Ostrander
MM
,
Mueller
NK
,
Figueiredo
H
.
Limbic system mechanisms of stress regulation: hypothalamo-pituitary-adrenocortical axis
.
Prog Neuropsychopharmacol Biol Psychiatry
.
2005
;
29
(
8
):
1201
1213
.

139.

Kinsey-Jones
JS
,
Li
XF
,
Knox
AM
,
Wilkinson
ES
,
Zhu
XL
,
Chaudhary
AA
,
Milligan
SR
,
Lightman
SL
,
O’Byrne
KT
.
Down-regulation of hypothalamic kisspeptin and its receptor, Kiss1r, mRNA expression is associated with stress-induced suppression of luteinising hormone secretion in the female rat
.
J Neuroendocrinol
.
2009
;
21
(
1
):
20
29
.

140.

Iwasa
T
,
Matsuzaki
T
,
Tungalagsuvd
A
,
Munkhzaya
M
,
Kawami
T
,
Niki
H
,
Kato
T
,
Kuwahara
A
,
Uemura
H
,
Yasui
T
,
Irahara
M
.
Hypothalamic Kiss1 and RFRP gene expressions are changed by a high dose of lipopolysaccharide in female rats
.
Horm Behav
.
2014
;
66
(
2
):
309
316
.

141.

Martini
AC
,
Fernández-Fernández
R
,
Tovar
S
,
Navarro
VM
,
Vigo
E
,
Vazquez
MJ
,
Davies
JS
,
Thompson
NM
,
Aguilar
E
,
Pinilla
L
,
Wells
T
,
Dieguez
C
,
Tena-Sempere
M
.
Comparative analysis of the effects of ghrelin and unacylated ghrelin on luteinizing hormone secretion in male rats
.
Endocrinology
.
2006
;
147
(
5
):
2374
2382
.

142.

Ubuka
T
,
Morgan
K
,
Pawson
AJ
,
Osugi
T
,
Chowdhury
VS
,
Minakata
H
,
Tsutsui
K
,
Millar
RP
,
Bentley
GE
.
Identification of human GnIH homologs, RFRP-1 and RFRP-3, and the cognate receptor, GPR147 in the human hypothalamic pituitary axis
.
PLoS One
.
2009
;
4
(
12
):
e8400
.

143.

Kirby
ED
,
Geraghty
AC
,
Ubuka
T
,
Bentley
GE
,
Kaufer
D
.
Stress increases putative gonadotropin inhibitory hormone and decreases luteinizing hormone in male rats
.
Proc Natl Acad Sci USA
.
2009
;
106
(
27
):
11324
11329
.

144.

Geraghty
AC
,
Muroy
SE
,
Zhao
S
,
Bentley
GE
,
Kriegsfeld
LJ
,
Kaufer
D
.
Knockdown of hypothalamic RFRP3 prevents chronic stress-induced infertility and embryo resorption
.
eLife
.
2015
;
4
:
4
.

145.

Wahab
F
,
Shahab
M
,
Behr
R
.
The involvement of gonadotropin inhibitory hormone and kisspeptin in the metabolic regulation of reproduction
.
J Endocrinol
.
2015
;
225
(
2
):
R49
R66
.

146.

Smith
JT
,
Reichenbach
A
,
Lemus
M
,
Mani
BK
,
Zigman
JM
,
Andrews
ZB
.
An eGFP-expressing subpopulation of growth hormone secretagogue receptor cells are distinct from kisspeptin, tyrosine hydroxylase, and RFamide-related peptide neurons in mice
.
Peptides
.
2013
;
47
:
45
53
.

147.

Kageyama
K
,
Hasegawa
G
,
Akimoto
K
,
Yamagata
S
,
Tamasawa
N
,
Suda
T
.
Differential regulation of gonadotropin-releasing hormone by corticotropin-releasing factor family peptides in hypothalamic N39 cells
.
Peptides
.
2012
;
33
(
1
):
149
155
.

148.

Rizwan
MZ
,
Poling
MC
,
Corr
M
,
Cornes
PA
,
Augustine
RA
,
Quennell
JH
,
Kauffman
AS
,
Anderson
GM
.
RFamide-related peptide-3 receptor gene expression in GnRH and kisspeptin neurons and GnRH-dependent mechanism of action
.
Endocrinology
.
2012
;
153
(
8
):
3770
3779
.

149.

Wu
M
,
Dumalska
I
,
Morozova
E
,
van den Pol
AN
,
Alreja
M
.
Gonadotropin inhibitory hormone inhibits basal forebrain vGluT2-gonadotropin-releasing hormone neurons via a direct postsynaptic mechanism
.
J Physiol
.
2009
;
587
(
Pt 7
):
1401
1411
.

150.

Li
PS
,
Wagner
WC
.
In vivo and in vitro studies on the effect of adrenocorticotropic hormone or cortisol on the pituitary response to gonadotropin releasing hormone
.
Biol Reprod
.
1983
;
29
(
1
):
25
37
.

151.

Melis
GB
,
Mais
V
,
Gambacciani
M
,
Paoletti
AM
,
Antinori
D
,
Fioretti
P
.
Dexamethasone reduces the postcastration gonadotropin rise in women
.
J Clin Endocrinol Metab
.
1987
;
65
(
2
):
237
241
.

152.

Pearce
GP
,
Paterson
AM
,
Hughes
PE
.
Effect of short-term elevations in plasma cortisol concentration on LH secretion in prepubertal gilts
.
J Reprod Fertil
.
1988
;
83
(
1
):
413
418
.

153.

Suter
DE
,
Schwartz
NB
,
Ringstrom
SJ
.
Dual role of glucocorticoids in regulation of pituitary content and secretion of gonadotropins
.
Am J Physiol
.
1988
;
254
(
5 Pt 1
):
E595
E600
.

154.

Suter
DE
,
Orosz
G
.
Effect of treatment with cortisol in vivo on secretion of gonadotropins in vitro
.
Biol Reprod
.
1989
;
41
(
6
):
1091
1096
.

155.

Fernández-Fernández
R
,
Tena-Sempere
M
,
Navarro
VM
,
Barreiro
ML
,
Castellano
JM
,
Aguilar
E
,
Pinilla
L
.
Effects of ghrelin upon gonadotropin-releasing hormone and gonadotropin secretion in adult female rats: in vivo and in vitro studies
.
Neuroendocrinology
.
2005
;
82
(
5-6
):
245
255
.

156.

Fernández-Fernández
R
,
Tena-Sempere
M
,
Roa
J
,
Castellano
JM
,
Navarro
VM
,
Aguilar
E
,
Pinilla
L
.
Direct stimulatory effect of ghrelin on pituitary release of LH through a nitric oxide-dependent mechanism that is modulated by estrogen
.
Reproduction
.
2007
;
133
(
6
):
1223
1232
.

157.

Suter
DE
,
Schwartz
NB
.
Effects of glucocorticoids on secretion of luteinizing hormone and follicle-stimulating hormone by female rat pituitary cells in vitro
.
Endocrinology
.
1985
;
117
(
3
):
849
854
.

158.

Kononen
J
,
Honkaniemi
J
,
Gustafsson
JA
,
Pelto-Huikko
M
.
Glucocorticoid receptor colocalization with pituitary hormones in the rat pituitary gland
.
Mol Cell Endocrinol
.
1993
;
93
(
1
):
97
103
.

159.

Breen
KM
,
Thackray
VG
,
Hsu
T
,
Mak-McCully
RA
,
Coss
D
,
Mellon
PL
.
Stress levels of glucocorticoids inhibit LHβ-subunit gene expression in gonadotrope cells
.
Mol Endocrinol
.
2012
;
26
(
10
):
1716
1731
.

160.

Evans
JJ
.
Modulation of gonadotropin levels by peptides acting at the anterior pituitary gland
.
Endocr Rev
.
1999
;
20
(
1
):
46
67
.

161.

Breen
KM
,
Karsch
FJ
.
Does cortisol inhibit pulsatile luteinizing hormone secretion at the hypothalamic or pituitary level?
Endocrinology
.
2004
;
145
(
2
):
692
698
.

162.

Breen
KM
,
Stackpole
CA
,
Clarke
IJ
,
Pytiak
AV
,
Tilbrook
AJ
,
Wagenmaker
ER
,
Young
EA
,
Karsch
FJ
.
Does the type II glucocorticoid receptor mediate cortisol-induced suppression in pituitary responsiveness to gonadotropin-releasing hormone?
Endocrinology
.
2004
;
145
(
6
):
2739
2746
.

163.

Breen
KM
,
Oakley
AE
,
Pytiak
AV
,
Tilbrook
AJ
,
Wagenmaker
ER
,
Karsch
FJ
.
Does cortisol acting via the type II glucocorticoid receptor mediate suppression of pulsatile luteinizing hormone secretion in response to psychosocial stress?
Endocrinology
.
2007
;
148
(
4
):
1882
1890
.

164.

Roney
JR
,
Simmons
ZL
.
Elevated psychological stress predicts reduced estradiol concentrations in young women
.
Adaptive Hum Behav Physiol
.
2015
;
1
(
1
):
30
40
.

165.

Norman
RL
,
Smith
CJ
.
Restraint inhibits luteinizing hormone and testosterone secretion in intact male rhesus macaques: effects of concurrent naloxone administration
.
Neuroendocrinology
.
1992
;
55
(
4
):
405
415
.

166.

Rivier
C
.
Inhibitory effect of neurogenic and immune stressors on testosterone secretion in rats
.
Neuroimmunomodulation
.
2002
;
10
(
1
):
17
29
.

167.

Macfarlane
MS
,
Breen
KM
,
Sakurai
H
,
Adams
BM
,
Adams
TE
.
Effect of duration of infusion of stress-like concentrations of cortisol on follicular development and the preovulatory surge of LH in sheep
.
Anim Reprod Sci
.
2000
;
63
(
3-4
):
167
175
.

168.

Yazawa
H
,
Sasagawa
I
,
Ishigooka
M
,
Nakada
T
.
Effect of immobilization stress on testicular germ cell apoptosis in rats
.
Hum Reprod
.
1999
;
14
(
7
):
1806
1810
.

169.

Yazawa
H
,
Sasagawa
I
,
Nakada
T
.
Apoptosis of testicular germ cells induced by exogenous glucocorticoid in rats
.
Hum Reprod
.
2000
;
15
(
9
):
1917
1920
.

170.

Cumming
DC
,
Quigley
ME
,
Yen
SS
.
Acute suppression of circulating testosterone levels by cortisol in men
.
J Clin Endocrinol Metab
.
1983
;
57
(
3
):
671
673
.

171.

Bhongade
MB
,
Prasad
S
,
Jiloha
RC
,
Ray
PC
,
Mohapatra
S
,
Koner
BC
.
Effect of psychological stress on fertility hormones and seminal quality in male partners of infertile couples
.
Andrologia
.
2015
;
47
(
3
):
336
342
.

172.

Yap
BK
,
Kazlauskas
R
,
Elghazi
K
,
Johnston
GA
,
Weatherby
RP
.
Profiling of urinary testosterone and luteinizing hormone in exercise-stressed male athletes, using gas chromatography-mass spectrometry and enzyme immunoassay techniques
.
J Chromatogr B Biomed Appl
.
1996
;
687
(
1
):
117
125
.

173.

Fenster
L
,
Katz
DF
,
Wyrobek
AJ
,
Pieper
C
,
Rempel
DM
,
Oman
D
,
Swan
SH
.
Effects of psychological stress on human semen quality
.
J Androl
.
1997
;
18
(
2
):
194
202
.

174.

Zorn
B
,
Sucur
V
,
Stare
J
,
Meden-Vrtovec
H
.
Decline in sex ratio at birth after 10-day war in Slovenia: brief communication
.
Hum Reprod
.
2002
;
17
(
12
):
3173
3177
.

175.

Abu-Musa
AA
,
Nassar
AH
,
Hannoun
AB
,
Usta
IM
.
Effect of the Lebanese civil war on sperm parameters
.
Fertil Steril
.
2007
;
88
(
6
):
1579
1582
.

176.

Hjollund
NH
,
Bonde
JP
,
Henriksen
TB
,
Giwercman
A
,
Olsen
J
;
Danish First Pregnancy Planner Study Team
.
Job strain and male fertility
.
Epidemiology
.
2004
;
15
(
1
):
114
117
.

177.

Janevic
T
,
Kahn
LG
,
Landsbergis
P
,
Cirillo
PM
,
Cohn
BA
,
Liu
X
,
Factor-Litvak
P
.
Effects of work and life stress on semen quality
.
Fertil Steril
.
2014
;
102
(
2
):
530
538
.

178.

Bigelow
PL
,
Jarrell
J
,
Young
MR
,
Keefe
TJ
,
Love
EJ
.
Association of semen quality and occupational factors: comparison of case-control analysis and analysis of continuous variables
.
Fertil Steril
.
1998
;
69
(
1
):
11
18
.

179.

Sheiner
EK
,
Sheiner
E
,
Carel
R
,
Potashnik
G
,
Shoham-Vardi
I
.
Potential association between male infertility and occupational psychological stress
.
J Occup Environ Med
.
2002
;
44
(
12
):
1093
1099
.

180.

Theorell
T
,
Karasek
RA
,
Eneroth
P
.
Job strain variations in relation to plasma testosterone fluctuations in working men--a longitudinal study
.
J Intern Med
.
1990
;
227
(
1
):
31
36
.

181.

de Jonge
J
,
Bosma
H
,
Peter
R
,
Siegrist
J
.
Job strain, effort-reward imbalance and employee well-being: a large-scale cross-sectional study
.
Soc Sci Med
.
2000
;
50
(
9
):
1317
1327
.

182.

Martin
SA
,
Atlantis
E
,
Lange
K
,
Taylor
AW
,
O’Loughlin
P
,
Wittert
GA
;
Florey Adelaide Male Ageing Study
.
Predictors of sexual dysfunction incidence and remission in men
.
J Sex Med
.
2014
;
11
(
5
):
1136
1147
.

183.

Barbazanges
A
,
Piazza
PV
,
Le Moal
M
,
Maccari
S
.
Maternal glucocorticoid secretion mediates long-term effects of prenatal stress
.
J Neurosci
.
1996
;
16
(
12
):
3943
3949
.

184.

Stalker
A
,
Hermo
L
,
Antakly
T
.
Covalent affinity labeling, radioautography, and immunocytochemistry localize the glucocorticoid receptor in rat testicular Leydig cells
.
Am J Anat
.
1989
;
186
(
4
):
369
377
.

185.

Hu
GX
,
Lian
QQ
,
Lin
H
,
Latif
SA
,
Morris
DJ
,
Hardy
MP
,
Ge
RS
.
Rapid mechanisms of glucocorticoid signaling in the Leydig cell
.
Steroids
.
2008
;
73
(
9-10
):
1018
1024
.

186.

Barreiro
ML
,
Gaytan
F
,
Castellano
JM
,
Suominen
JS
,
Roa
J
,
Gaytan
M
,
Aguilar
E
,
Dieguez
C
,
Toppari
J
,
Tena-Sempere
M
.
Ghrelin inhibits the proliferative activity of immature Leydig cells in vivo and regulates stem cell factor messenger ribonucleic acid expression in rat testis
.
Endocrinology
.
2004
;
145
(
11
):
4825
4834
.

187.

Kheradmand
A
,
Roshangar
L
,
Taati
M
.
The role of ghrelin on the morphometry and intracellular changes in the rat testis
.
Tissue Cell
.
2009
;
41
(
2
):
105
111
.

188.

Schreiber
JR
,
Nakamura
K
,
Erickson
GF
.
Rat ovary glucocorticoid receptor: identification and characterization
.
Steroids
.
1982
;
39
(
5
):
569
584
.

189.

Tetsuka
M
,
Milne
M
,
Simpson
GE
,
Hillier
SG
.
Expression of 11beta-hydroxysteroid dehydrogenase, glucocorticoid receptor, and mineralocorticoid receptor genes in rat ovary
.
Biol Reprod
.
1999
;
60
(
2
):
330
335
.

190.

Michael
AE
,
Pester
LA
,
Curtis
P
,
Shaw
RW
,
Edwards
CR
,
Cooke
BA
.
Direct inhibition of ovarian steroidogenesis by cortisol and the modulatory role of 11 beta-hydroxysteroid dehydrogenase
.
Clin Endocrinol (Oxf)
.
1993
;
38
(
6
):
641
644
.

191.

Viani
I
,
Vottero
A
,
Tassi
F
,
Cremonini
G
,
Sartori
C
,
Bernasconi
S
,
Ferrari
B
,
Ghizzoni
L
.
Ghrelin inhibits steroid biosynthesis by cultured granulosa-lutein cells
.
J Clin Endocrinol Metab
.
2008
;
93
(
4
):
1476
1481
.

192.

Rak-Mardyła
A
,
Wróbel
A
,
Gregoraszczuk
EL
.
Ghrelin negatively affects the function of ovarian follicles in mature pigs by direct action on basal and gonadotropin-stimulated steroidogenesis
.
Reprod Sci
.
2015
;
22
(
4
):
469
475
.

193.

Yang
JG
,
Chen
WY
,
Li
PS
.
Effects of glucocorticoids on maturation of pig oocytes and their subsequent fertilizing capacity in vitro
.
Biol Reprod
.
1999
;
60
(
4
):
929
936
.

194.

Van Merris
V
,
Van Wemmel
K
,
Cortvrindt
R
.
In vitro effects of dexamethasone on mouse ovarian function and pre-implantation embryo development
.
Reprod Toxicol
.
2007
;
23
(
1
):
32
41
.

195.

González
R
,
Ruiz-León
Y
,
Gomendio
M
,
Roldan
ER
.
The effect of glucocorticoids on ERK-1/2 phosphorylation during maturation of lamb oocytes and their subsequent fertilization and cleavage ability in vitro
.
Reprod Toxicol
.
2010
;
29
(
2
):
198
205
.

196.

Jimena
P
,
Castilla
JA
,
Peran
F
,
Ramirez
JP
,
Vergara
F
, Jr
,
Molina
R
,
Vergara
F
,
Herruzo
A
.
Adrenal hormones in human follicular fluid
.
Acta Endocrinol (Copenh)
.
1992
;
127
(
5
):
403
406
.

197.

Lewicka
S
,
von Hagens
C
,
Hettinger
U
,
Grunwald
K
,
Vecsei
P
,
Runnebaum
B
,
Rabe
T
.
Cortisol and cortisone in human follicular fluid and serum and the outcome of IVF treatment
.
Hum Reprod
.
2003
;
18
(
8
):
1613
1617
.

198.

Lara
HE
,
McDonald
JK
,
Ojeda
SR
.
Involvement of nerve growth factor in female sexual development
.
Endocrinology
.
1990
;
126
(
1
):
364
375
.

199.

Aguado
LI
,
Petrovic
SL
,
Ojeda
SR
.
Ovarian beta-adrenergic receptors during the onset of puberty: characterization, distribution, and coupling to steroidogenic responses
.
Endocrinology
.
1982
;
110
(
4
):
1124
1132
.

200.

Aguado
LI
,
Ojeda
SR
.
Prepubertal ovarian function is finely regulated by direct adrenergic influences. Role of noradrenergic innervation
.
Endocrinology
.
1984
;
114
(
5
):
1845
1853
.

201.

Hernandez
ER
,
Jimenez
JL
,
Payne
DW
,
Adashi
EY
.
Adrenergic regulation of ovarian androgen biosynthesis is mediated via beta 2-adrenergic theca-interstitial cell recognition sites
.
Endocrinology
.
1988
;
122
(
4
):
1592
1602
.

202.

Paredes
A
,
Gálvez
A
,
Leyton
V
,
Aravena
G
,
Fiedler
JL
,
Bustamante
D
,
Lara
HE
.
Stress promotes development of ovarian cysts in rats: the possible role of sympathetic nerve activation
.
Endocrine
.
1998
;
8
(
3
):
309
315
.

203.

Dorfman
M
,
Arancibia
S
,
Fiedler
JL
,
Lara
HE
.
Chronic intermittent cold stress activates ovarian sympathetic nerves and modifies ovarian follicular development in the rat
.
Biol Reprod
.
2003
;
68
(
6
):
2038
2043
.

204.

Bernuci
MP
,
Szawka
RE
,
Helena
CV
,
Leite
CM
,
Lara
HE
,
Anselmo-Franci
JA
.
Locus coeruleus mediates cold stress-induced polycystic ovary in rats
.
Endocrinology
.
2008
;
149
(
6
):
2907
2916
.

205.

Tschöp
M
,
Smiley
DL
,
Heiman
ML
.
Ghrelin induces adiposity in rodents
.
Nature
.
2000
;
407
(
6806
):
908
913
.

206.

Nakazato
M
,
Murakami
N
,
Date
Y
,
Kojima
M
,
Matsuo
H
,
Kangawa
K
,
Matsukura
S
.
A role for ghrelin in the central regulation of feeding
.
Nature
.
2001
;
409
(
6817
):
194
198
.

207.

Ku
JM
,
Andrews
ZB
,
Barsby
T
,
Reichenbach
A
,
Lemus
MB
,
Drummond
GR
,
Sleeman
MW
,
Spencer
SJ
,
Sobey
CG
,
Miller
AA
.
Ghrelin-related peptides exert protective effects in the cerebral circulation of male mice through a nonclassical ghrelin receptor(s)
.
Endocrinology
.
2015
;
156
(
1
):
280
290
.

208.

Abizaid
A
,
Liu
ZW
,
Andrews
ZB
,
Shanabrough
M
,
Borok
E
,
Elsworth
JD
,
Roth
RH
,
Sleeman
MW
,
Picciotto
MR
,
Tschöp
MH
,
Gao
XB
,
Horvath
TL
.
Ghrelin modulates the activity and synaptic input organization of midbrain dopamine neurons while promoting appetite
.
J Clin Invest
.
2006
;
116
(
12
):
3229
3239
.

209.

Naleid
AM
,
Grace
MK
,
Cummings
DE
,
Levine
AS
.
Ghrelin induces feeding in the mesolimbic reward pathway between the ventral tegmental area and the nucleus accumbens
.
Peptides
.
2005
;
26
(
11
):
2274
2279
.

210.

Diano
S
,
Farr
SA
,
Benoit
SC
,
McNay
EC
,
da Silva
I
,
Horvath
B
,
Gaskin
FS
,
Nonaka
N
,
Jaeger
LB
,
Banks
WA
,
Morley
JE
,
Pinto
S
,
Sherwin
RS
,
Xu
L
,
Yamada
KA
,
Sleeman
MW
,
Tschöp
MH
,
Horvath
TL
.
Ghrelin controls hippocampal spine synapse density and memory performance
.
Nat Neurosci
.
2006
;
9
(
3
):
381
388
.

211.

Ku
JM
,
Taher
M
,
Chin
KY
,
Barsby
T
,
Austin
V
,
Wong
CH
,
Andrews
ZB
,
Spencer
SJ
,
Miller
AA
.
Protective actions of des-acylated ghrelin on brain injury and blood-brain barrier disruption after stroke in mice
.
Clin Sci (Lond)
.
2016
;
130
(
17
):
1545
1558
.

212.

Spencer
SJ
,
Emmerzaal
TL
,
Kozicz
T
,
Andrews
ZB
.
Ghrelin’s role in the hypothalamic-pituitary-adrenal axis stress response: implications for mood disorders
.
Biol Psychiatry
.
2015
;
78
(
1
):
19
27
.

213.

Kheradmand
A
,
Alirezaei
M
,
Asadian
P
,
Rafiei Alavi
E
,
Joorabi
S
.
Antioxidant enzyme activity and MDA level in the rat testis following chronic administration of ghrelin
.
Andrologia
.
2009
;
41
(
6
):
335
340
.

214.

Kheradmand
A
,
Roshangar
L
,
Taati
M
,
Sirotkin
AV
.
Morphometrical and intracellular changes in rat ovaries following chronic administration of ghrelin
.
Tissue Cell
.
2009
;
41
(
5
):
311
317
.

215.

Aghajanova
L
,
Rumman
A
,
Altmäe
S
,
Wånggren
K
,
Stavreus-Evers
A
.
Diminished endometrial expression of ghrelin and ghrelin receptor contributes to infertility
.
Reprod Sci
.
2010
;
17
(
9
):
823
832
.

216.

Luque
EM
,
Torres
PJ
,
de Loredo
N
,
Vincenti
LM
,
Stutz
G
,
Santillán
ME
,
Ruiz
RD
,
de Cuneo
MF
,
Martini
AC
.
Role of ghrelin in fertilization, early embryo development, and implantation periods
.
Reproduction
.
2014
;
148
(
2
):
159
167
.

217.

Kluge
M
,
Schüssler
P
,
Uhr
M
,
Yassouridis
A
,
Steiger
A
.
Ghrelin suppresses secretion of luteinizing hormone in humans
.
J Clin Endocrinol Metab
.
2007
;
92
(
8
):
3202
3205
.

218.

Kluge
M
,
Uhr
M
,
Bleninger
P
,
Yassouridis
A
,
Steiger
A
.
Ghrelin suppresses secretion of FSH in males
.
Clin Endocrinol (Oxf)
.
2009
;
70
(
6
):
920
923
.

219.

Kluge
M
,
Schüssler
P
,
Schmidt
D
,
Uhr
M
,
Steiger
A
.
Ghrelin suppresses secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in women
.
J Clin Endocrinol Metab
.
2012
;
97
(
3
):
E448
E451
.

220.

Kluge
M
.
Ghrelin suppresses secretion of gonadotropins in women
.
Reprod Sci
.
2012
;
19
(
12
):
NP3
.

221.

Kluge
M
,
Schmidt
D
,
Uhr
M
,
Steiger
A
.
Ghrelin suppresses nocturnal secretion of luteinizing hormone (LH) and thyroid stimulating hormone (TSH) in patients with major depression
.
J Psychiatr Res
.
2013
;
47
(
9
):
1236
1239
.

222.

Kluge
M
.
Evidence of a suppressive effect of ghrelin on FSH secretion in vivo: comment on the review article “Ghrelin: a metabolic signal affecting the reproductive system”
.
Cytokine Growth Factor Rev
.
2009
;
20
(
4
):
339
340
.

223.

Kojima
M
,
Hosoda
H
,
Date
Y
,
Nakazato
M
,
Matsuo
H
,
Kangawa
K
.
Ghrelin is a growth-hormone-releasing acylated peptide from stomach
.
Nature
.
1999
;
402
(
6762
):
656
660
.

224.

Yang
J
,
Brown
MS
,
Liang
G
,
Grishin
NV
,
Goldstein
JL
.
Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone
.
Cell
.
2008
;
132
(
3
):
387
396
.

225.

Gutierrez
JA
,
Solenberg
PJ
,
Perkins
DR
,
Willency
JA
,
Knierman
MD
,
Jin
Z
,
Witcher
DR
,
Luo
S
,
Onyia
JE
,
Hale
JE
.
Ghrelin octanoylation mediated by an orphan lipid transferase
.
Proc Natl Acad Sci USA
.
2008
;
105
(
17
):
6320
6325
.

226.

Caminos
JE
,
Tena-Sempere
M
,
Gaytán
F
,
Sanchez-Criado
JE
,
Barreiro
ML
,
Nogueiras
R
,
Casanueva
FF
,
Aguilar
E
,
Diéguez
C
.
Expression of ghrelin in the cyclic and pregnant rat ovary
.
Endocrinology
.
2003
;
144
(
4
):
1594
1602
.

227.

Caminos
JE
,
Nogueiras
R
,
Blanco
M
,
Seoane
LM
,
Bravo
S
,
Alvarez
CV
,
García-Caballero
T
,
Casanueva
FF
,
Diéguez
C
.
Cellular distribution and regulation of ghrelin messenger ribonucleic acid in the rat pituitary gland
.
Endocrinology
.
2003
;
144
(
11
):
5089
5097
.

228.

Gaytan
F
,
Barreiro
ML
,
Caminos
JE
,
Chopin
LK
,
Herington
AC
,
Morales
C
,
Pinilla
L
,
Paniagua
R
,
Nistal
M
,
Casanueva
FF
,
Aguilar
E
,
Diéguez
C
,
Tena-Sempere
M
.
Expression of ghrelin and its functional receptor, the type 1a growth hormone secretagogue receptor, in normal human testis and testicular tumors
.
J Clin Endocrinol Metab
.
2004
;
89
(
1
):
400
409
.

229.

Gaytan
F
,
Morales
C
,
Barreiro
ML
,
Jeffery
P
,
Chopin
LK
,
Herington
AC
,
Casanueva
FF
,
Aguilar
E
,
Dieguez
C
,
Tena-Sempere
M
.
Expression of growth hormone secretagogue receptor type 1a, the functional ghrelin receptor, in human ovarian surface epithelium, mullerian duct derivatives, and ovarian tumors
.
J Clin Endocrinol Metab
.
2005
;
90
(
3
):
1798
1804
.

230.

Sakata
I
,
Nakano
Y
,
Osborne-Lawrence
S
,
Rovinsky
SA
,
Lee
CE
,
Perello
M
,
Anderson
JG
,
Coppari
R
,
Xiao
G
,
Lowell
BB
,
Elmquist
JK
,
Zigman
JM
.
Characterization of a novel ghrelin cell reporter mouse
.
Regul Pept
.
2009
;
155
(
1-3
):
91
98
.

231.

Furness
JB
,
Hunne
B
,
Matsuda
N
,
Yin
L
,
Russo
D
,
Kato
I
,
Fujimiya
M
,
Patterson
M
,
McLeod
J
,
Andrews
ZB
,
Bron
R
.
Investigation of the presence of ghrelin in the central nervous system of the rat and mouse
.
Neuroscience
.
2011
;
193
:
1
9
.

232.

Banks
WA
,
Tschöp
M
,
Robinson
SM
,
Heiman
ML
.
Extent and direction of ghrelin transport across the blood-brain barrier is determined by its unique primary structure
.
J Pharmacol Exp Ther
.
2002
;
302
(
2
):
822
827
.

233.

Rak
A
,
Szczepankiewicz
D
,
Gregoraszczuk
EL
.
Expression of ghrelin receptor, GHSR-1a, and its functional role in the porcine ovarian follicles
.
Growth Horm IGF Res
.
2009
;
19
(
1
):
68
76
.

234.

Zigman
JM
,
Jones
JE
,
Lee
CE
,
Saper
CB
,
Elmquist
JK
.
Expression of ghrelin receptor mRNA in the rat and the mouse brain
.
J Comp Neurol
.
2006
;
494
(
3
):
528
548
.

235.

Reichenbach
A
,
Steyn
FJ
,
Sleeman
MW
,
Andrews
ZB
.
Ghrelin receptor expression and colocalization with anterior pituitary hormones using a GHSR-GFP mouse line
.
Endocrinology
.
2012
;
153
(
11
):
5452
5466
.

236.

Szczepankiewicz
D
,
Skrzypski
M
,
Pruszynska-Oszmalek
E
,
Zimmermann
D
,
Andralojc
K
,
Kaczmarek
P
,
Wojciechowicz
T
,
Sassek
M
,
Nowak
KW
.
Importance of ghrelin in hypothalamus-pituitary axis on growth hormone release during normal pregnancy in the rat
.
J Physiol Pharmacol
.
2010
;
61
(
4
):
443
449
.

237.

Miller
DW
,
Harrison
JL
,
Brown
YA
,
Doyle
U
,
Lindsay
A
,
Adam
CL
,
Lea
RG
.
Immunohistochemical evidence for an endocrine/paracrine role for ghrelin in the reproductive tissues of sheep
.
Reprod Biol Endocrinol
.
2005
;
3
:
60
.

238.

Wang
J
,
Guo
S
,
Han
L
,
Fang
M
,
Wang
L
,
Bartsch
JW
,
Li
J
.
Correlation of ghrelin and growth hormone secretagogue receptor expression with clinical features in human pituitary adenomas
.
Exp Ther Med
.
2015
;
9
(
5
):
1909
1914
.

239.

El-Magd
MA
,
Saleh
AA
,
Abdel-Hamid
TM
,
Saleh
RM
,
Afifi
MA
.
Is really endogenous ghrelin a hunger signal in chickens? Association of GHSR SNPs with increase appetite, growth traits, expression and serum level of GHRL, and GH
.
Gen Comp Endocrinol
.
2016
;
237
:
131
139
.

240.

Chen
T
,
Tang
Z
,
Yan
A
,
Li
W
,
Lin
H
.
Molecular cloning and mRNA expression analysis of two GH secretagogue receptor transcripts in orange-spotted grouper (Epinephelus coioides)
.
J Endocrinol
.
2008
;
199
(
2
):
253
265
.

241.

Chan
CB
,
Leung
PK
,
Wise
H
,
Cheng
CH
.
Signal transduction mechanism of the seabream growth hormone secretagogue receptor
.
FEBS Lett
.
2004
;
577
(
1-2
):
147
153
.

242.

Camiña
JP
.
Cell biology of the ghrelin receptor
.
J Neuroendocrinol
.
2006
;
18
(
1
):
65
76
.

243.

Howard
AD
,
Feighner
SD
,
Cully
DF
,
Arena
JP
,
Liberator
PA
,
Rosenblum
CI
,
Hamelin
M
,
Hreniuk
DL
,
Palyha
OC
,
Anderson
J
,
Paress
PS
,
Diaz
C
,
Chou
M
,
Liu
KK
,
McKee
KK
,
Pong
SS
,
Chaung
LY
,
Elbrecht
A
,
Dashkevicz
M
,
Heavens
R
,
Rigby
M
,
Sirinathsinghji
DJ
,
Dean
DC
,
Melillo
DG
,
Patchett
AA
,
Nargund
R
,
Griffin
PR
,
DeMartino
JA
,
Gupta
SK
,
Schaeffer
JM
,
Smith
RG
,
Van der Ploeg
LH
.
A receptor in pituitary and hypothalamus that functions in growth hormone release
.
Science
.
1996
;
273
(
5277
):
974
977
.

244.

McKee
KK
,
Palyha
OC
,
Feighner
SD
,
Hreniuk
DL
,
Tan
CP
,
Phillips
MS
,
Smith
RG
,
Van der Ploeg
LH
,
Howard
AD
.
Molecular analysis of rat pituitary and hypothalamic growth hormone secretagogue receptors
.
Mol Endocrinol
.
1997
;
11
(
4
):
415
423
.

245.

Petersen
PS
,
Woldbye
DP
,
Madsen
AN
,
Egerod
KL
,
Jin
C
,
Lang
M
,
Rasmussen
M
,
Beck-Sickinger
AG
,
Holst
B
.
In vivo characterization of high Basal signaling from the ghrelin receptor
.
Endocrinology
.
2009
;
150
(
11
):
4920
4930
.

246.

Kern
A
,
Albarran-Zeckler
R
,
Walsh
HE
,
Smith
RG
.
Apo-ghrelin receptor forms heteromers with DRD2 in hypothalamic neurons and is essential for anorexigenic effects of DRD2 agonism
.
Neuron
.
2012
;
73
(
2
):
317
332
.

247.

Rediger
A
,
Piechowski
CL
,
Yi
CX
,
Tarnow
P
,
Strotmann
R
,
Grüters
A
,
Krude
H
,
Schöneberg
T
,
Tschöp
MH
,
Kleinau
G
,
Biebermann
H
.
Mutually opposite signal modulation by hypothalamic heterodimerization of ghrelin and melanocortin-3 receptors
.
J Biol Chem
.
2011
;
286
(
45
):
39623
39631
.

248.

Schellekens
H
,
van Oeffelen
WE
,
Dinan
TG
,
Cryan
JF
.
Promiscuous dimerization of the growth hormone secretagogue receptor (GHS-R1a) attenuates ghrelin-mediated signaling
.
J Biol Chem
.
2013
;
288
(
1
):
181
191
.

249.

Delhanty
PJ
,
Neggers
SJ
,
van der Lely
AJ
.
Should we consider des-acyl ghrelin as a separate hormone and if so, what does it do?
Front Horm Res
.
2014
;
42
:
163
174
.

250.

Stengel
A
,
Wang
L
,
Taché
Y
.
Stress-related alterations of acyl and desacyl ghrelin circulating levels: mechanisms and functional implications
.
Peptides
.
2011
;
32
(
11
):
2208
2217
.

251.

Stark
R
,
Santos
VV
,
Geenen
B
,
Cabral
A
,
Dinan
T
,
Bayliss
JA
,
Lockie
SH
,
Reichenbach
A
,
Lemus
MB
,
Perello
M
,
Spencer
SJ
,
Kozicz
T
,
Andrews
ZB
.
Des-acyl ghrelin and ghrelin o-acyltransferase regulate hypothalamic-pituitary-adrenal axis activation and anxiety in response to acute stress
.
Endocrinology
.
2016
;
157
(
10
):
3946
3957
.

252.

Jaremka
LM
,
Belury
MA
,
Andridge
RR
,
Malarkey
WB
,
Glaser
R
,
Christian
L
,
Emery
CF
,
Kiecolt-Glaser
JK
.
Interpersonal stressors predict ghrelin and leptin levels in women
.
Psychoneuroendocrinology
.
2014
;
48
:
178
188
.

253.

Rouach
V
,
Bloch
M
,
Rosenberg
N
,
Gilad
S
,
Limor
R
,
Stern
N
,
Greenman
Y
.
The acute ghrelin response to a psychological stress challenge does not predict the post-stress urge to eat
.
Psychoneuroendocrinology
.
2007
;
32
(
6
):
693
702
.

254.

Raspopow
K
,
Abizaid
A
,
Matheson
K
,
Anisman
H
.
Psychosocial stressor effects on cortisol and ghrelin in emotional and non-emotional eaters: influence of anger and shame
.
Horm Behav
.
2010
;
58
(
4
):
677
684
.

255.

Monteleone
P
,
Tortorella
A
,
Scognamiglio
P
,
Serino
I
,
Monteleone
AM
,
Maj
M
.
The acute salivary ghrelin response to a psychosocial stress is enhanced in symptomatic patients with bulimia nervosa: a pilot study
.
Neuropsychobiology
.
2012
;
66
(
4
):
230
236
.

256.

Raspopow
K
,
Abizaid
A
,
Matheson
K
,
Anisman
H
.
Anticipation of a psychosocial stressor differentially influences ghrelin, cortisol and food intake among emotional and non-emotional eaters
.
Appetite
.
2014
;
74
:
35
43
.

257.

Takaya
K
,
Ariyasu
H
,
Kanamoto
N
,
Iwakura
H
,
Yoshimoto
A
,
Harada
M
,
Mori
K
,
Komatsu
Y
,
Usui
T
,
Shimatsu
A
,
Ogawa
Y
,
Hosoda
K
,
Akamizu
T
,
Kojima
M
,
Kangawa
K
,
Nakao
K
.
Ghrelin strongly stimulates growth hormone release in humans
.
J Clin Endocrinol Metab
.
2000
;
85
(
12
):
4908
4911
.

258.

Arvat
E
,
Maccario
M
,
Di Vito
L
,
Broglio
F
,
Benso
A
,
Gottero
C
,
Papotti
M
,
Muccioli
G
,
Dieguez
C
,
Casanueva
FF
,
Deghenghi
R
,
Camanni
F
,
Ghigo
E
.
Endocrine activities of ghrelin, a natural growth hormone secretagogue (GHS), in humans: comparison and interactions with hexarelin, a nonnatural peptidyl GHS, and GH-releasing hormone
.
J Clin Endocrinol Metab
.
2001
;
86
(
3
):
1169
1174
.

259.

Schmid
DA
,
Held
K
,
Ising
M
,
Uhr
M
,
Weikel
JC
,
Steiger
A
.
Ghrelin stimulates appetite, imagination of food, GH, ACTH, and cortisol, but does not affect leptin in normal controls
.
Neuropsychopharmacology
.
2005
;
30
(
6
):
1187
1192
.

260.

Vulliémoz
NR
,
Xiao
E
,
Xia-Zhang
L
,
Germond
M
,
Rivier
J
,
Ferin
M
.
Decrease in luteinizing hormone pulse frequency during a five-hour peripheral ghrelin infusion in the ovariectomized rhesus monkey
.
J Clin Endocrinol Metab
.
2004
;
89
(
11
):
5718
5723
.

261.

Vulliémoz
NR
,
Xiao
E
,
Xia-Zhang
L
,
Rivier
J
,
Ferin
M
.
Astressin B, a nonselective corticotropin-releasing hormone receptor antagonist, prevents the inhibitory effect of ghrelin on luteinizing hormone pulse frequency in the ovariectomized rhesus monkey
.
Endocrinology
.
2008
;
149
(
3
):
869
874
.

262.

Michopoulos
V
,
Loucks
T
,
Berga
SL
,
Rivier
J
,
Wilson
ME
.
Increased ghrelin sensitivity and calorie consumption in subordinate monkeys is affected by short-term astressin B administration
.
Endocrine
.
2010
;
38
(
2
):
227
234
.

263.

Jarrell
H
,
Hoffman
JB
,
Kaplan
JR
,
Berga
S
,
Kinkead
B
,
Wilson
ME
.
Polymorphisms in the serotonin reuptake transporter gene modify the consequences of social status on metabolic health in female rhesus monkeys
.
Physiol Behav
.
2008
;
93
(
4-5
):
807
819
.

264.

Hansson
C
,
Annerbrink
K
,
Nilsson
S
,
Bah
J
,
Olsson
M
,
Allgulander
C
,
Andersch
S
,
Sjödin
I
,
Eriksson
E
,
Dickson
SL
.
A possible association between panic disorder and a polymorphism in the preproghrelingene
.
Psychiatry Res
.
2013
;
206
(
1
):
22
25
.

265.

Nakashima
K
,
Akiyoshi
J
,
Hatano
K
,
Hanada
H
,
Tanaka
Y
,
Tsuru
J
,
Matsushita
H
,
Kodama
K
,
Isogawa
K
.
Ghrelin gene polymorphism is associated with depression, but not panic disorder
.
Psychiatr Genet
.
2008
;
18
(
5
):
257
.

266.

Ishitobi
Y
,
Kohno
K
,
Kanehisa
M
,
Inoue
A
,
Imanaga
J
,
Maruyama
Y
,
Ninomiya
T
,
Higuma
H
,
Okamoto
S
,
Tanaka
Y
,
Tsuru
J
,
Hanada
H
,
Isogawa
K
,
Akiyoshi
J
.
Serum ghrelin levels and the effects of antidepressants in major depressive disorder and panic disorder
.
Neuropsychobiology
.
2012
;
66
(
3
):
185
192
.

267.

Kurt
E
,
Guler
O
,
Serteser
M
,
Cansel
N
,
Ozbulut
O
,
Altinbaş
K
,
Alataş
G
,
Savaş
H
,
Gecici
O
.
The effects of electroconvulsive therapy on ghrelin, leptin and cholesterol levels in patients with mood disorders
.
Neurosci Lett
.
2007
;
426
(
1
):
49
53
.

268.

Ozsoy
S
,
Besirli
A
,
Abdulrezzak
U
,
Basturk
M
.
Serum ghrelin and leptin levels in patients with depression and the effects of treatment
.
Psychiatry Investig
.
2014
;
11
(
2
):
167
172
.

269.

Tunçel
OK
,
Akbaş
S
,
Bilgici
B
.
Increased ghrelin levels and unchanged adipocytokine levels in major depressive disorder
.
J Child Adolesc Psychopharmacol
.
2016
;
26
(
8
):
733
739
.

270.

Schanze
A
,
Reulbach
U
,
Scheuchenzuber
M
,
Groschl
M
,
Kornhuber
J
,
Kraus
T
.
Ghrelin and eating disturbances in psychiatric disorders
.
Neuropsychobiology
.
2008
;
57
(
3
):
126
130
.

271.

Kluge
M
,
Schussler
P
,
Schmid
D
,
Uhr
M
,
Kleyer
S
,
Yassouridis
A
,
Steiger
A
.
Ghrelin plasma levels are not altered in major depression
.
Neuropsychobiology
.
2009
;
59
(
4
):
199
204
.

272.

Matsuo
K
,
Nakano
M
,
Nakashima
M
,
Watanuki
T
,
Egashira
K
,
Matsubara
T
,
Watanabe
Y
.
Neural correlates of plasma acylated ghrelin level in individuals with major depressive disorder
.
Brain Res
.
2012
;
1473
:
185
192
.

273.

Tschöp
M
,
Weyer
C
,
Tataranni
PA
,
Devanarayan
V
,
Ravussin
E
,
Heiman
ML
.
Circulating ghrelin levels are decreased in human obesity
.
Diabetes
.
2001
;
50
(
4
):
707
709
.

274.

Williams
DL
,
Cummings
DE
.
Regulation of ghrelin in physiologic and pathophysiologic states
.
J Nutr
.
2005
;
135
(
5
):
1320
1325
.

275.

Barim
AO
,
Aydin
S
,
Colak
R
,
Dag
E
,
Deniz
O
,
Sahin
I
.
Ghrelin, paraoxonase and arylesterase levels in depressive patients before and after citalopram treatment
.
Clin Biochem
.
2009
;
42
(
10-11
):
1076
1081
.

276.

Schmid
DA
,
Wichniak
A
,
Uhr
M
,
Ising
M
,
Brunner
H
,
Held
K
,
Weikel
JC
,
Sonntag
A
,
Steiger
A
.
Changes of sleep architecture, spectral composition of sleep EEG, the nocturnal secretion of cortisol, ACTH, GH, prolactin, melatonin, ghrelin, and leptin, and the DEX-CRH test in depressed patients during treatment with mirtazapine
.
Neuropsychopharmacology
.
2006
;
31
(
4
):
832
844
.

277.

Pinar
M
,
Gulsun
M
,
Tasci
I
,
Erdil
A
,
Bolu
E
,
Acikel
C
,
Doruk
A
.
Maprotiline induced weight gain in depressive disorder: changes in circulating ghrelin and adiponectin levels and insulin sensitivity
.
Prog Neuropsychopharmacol Biol Psychiatry
.
2008
;
32
(
1
):
135
139
.

278.

Kluge
M
,
Schüssler
P
,
Dresler
M
,
Schmidt
D
,
Yassouridis
A
,
Uhr
M
,
Steiger
A
.
Effects of ghrelin on psychopathology, sleep and secretion of cortisol and growth hormone in patients with major depression
.
J Psychiatr Res
.
2011
;
45
(
3
):
421
426
.

279.

Homan
P
,
Grob
S
,
Milos
G
,
Schnyder
U
,
Hasler
G
.
Reduction in total plasma ghrelin levels following catecholamine depletion: relation to bulimic and depressive symptoms
.
Psychoneuroendocrinology
.
2013
;
38
(
9
):
1545
1552
.

280.

Asakawa
A
,
Inui
A
,
Kaga
T
,
Yuzuriha
H
,
Nagata
T
,
Fujimiya
M
,
Katsuura
G
,
Makino
S
,
Fujino
MA
,
Kasuga
M
.
A role of ghrelin in neuroendocrine and behavioral responses to stress in mice
.
Neuroendocrinology
.
2001
;
74
(
3
):
143
147
.

281.

Kristenssson
E
,
Sundqvist
M
,
Astin
M
,
Kjerling
M
,
Mattsson
H
,
Dornonville de la Cour
C
,
Håkanson
R
,
Lindström
E
.
Acute psychological stress raises plasma ghrelin in the rat
.
Regul Pept
.
2006
;
134
(
2-3
):
114
117
.

282.

Ochi
M
,
Tominaga
K
,
Tanaka
F
,
Tanigawa
T
,
Shiba
M
,
Watanabe
T
,
Fujiwara
Y
,
Oshitani
N
,
Higuchi
K
,
Arakawa
T
.
Effect of chronic stress on gastric emptying and plasma ghrelin levels in rats
.
Life Sci
.
2008
;
82
(
15-16
):
862
868
.

283.

Patterson
ZR
,
Ducharme
R
,
Anisman
H
,
Abizaid
A
.
Altered metabolic and neurochemical responses to chronic unpredictable stressors in ghrelin receptor-deficient mice
.
Eur J Neurosci
.
2010
;
32
(
4
):
632
639
.

284.

Berton
O
,
McClung
CA
,
Dileone
RJ
,
Krishnan
V
,
Renthal
W
,
Russo
SJ
,
Graham
D
,
Tsankova
NM
,
Bolanos
CA
,
Rios
M
,
Monteggia
LM
,
Self
DW
,
Nestler
EJ
.
Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress
.
Science
.
2006
;
311
(
5762
):
864
868
.

285.

Nestler
EJ
,
Hyman
SE
.
Animal models of neuropsychiatric disorders
.
Nat Neurosci
.
2010
;
13
(
10
):
1161
1169
.

286.

Lutter
M
,
Sakata
I
,
Osborne-Lawrence
S
,
Rovinsky
SA
,
Anderson
JG
,
Jung
S
,
Birnbaum
S
,
Yanagisawa
M
,
Elmquist
JK
,
Nestler
EJ
,
Zigman
JM
.
The orexigenic hormone ghrelin defends against depressive symptoms of chronic stress
.
Nat Neurosci
.
2008
;
11
(
7
):
752
753
.

287.

Cabral
A
,
Suescun
O
,
Zigman
JM
,
Perello
M
.
Ghrelin indirectly activates hypophysiotropic CRF neurons in rodents
.
PLoS One
.
2012
;
7
(
2
):
e31462
.

288.

Sominsky
L
,
Ziko
I
,
Spencer
SJ
.
Neonatal overfeeding disrupts pituitary ghrelin signalling in female rats long-term; Implications for the stress response
.
PLoS One
.
2017
;
12
(
3
):
e0173498
.

289.

Johnstone
LE
,
Srisawat
R
,
Kumarnsit
E
,
Leng
G
.
Hypothalamic expression of NPY mRNA, vasopressin mRNA and CRF mRNA in response to food restriction and central administration of the orexigenic peptide GHRP-6
.
Stress
.
2005
;
8
(
1
):
59
67
.

290.

Schellekens
H
,
Finger
BC
,
Dinan
TG
,
Cryan
JF
.
Ghrelin signalling and obesity: at the interface of stress, mood and food reward
.
Pharmacol Ther
.
2012
;
135
(
3
):
316
326
.

291.

Sacher
J
,
Neumann
J
,
Fünfstück
T
,
Soliman
A
,
Villringer
A
,
Schroeter
ML
.
Mapping the depressed brain: a meta-analysis of structural and functional alterations in major depressive disorder
.
J Affect Disord
.
2012
;
140
(
2
):
142
148
.

292.

Goldstone
AP
,
Prechtl de Hernandez
CG
,
Beaver
JD
,
Muhammed
K
,
Croese
C
,
Bell
G
,
Durighel
G
,
Hughes
E
,
Waldman
AD
,
Frost
G
,
Bell
JD
.
Fasting biases brain reward systems towards high-calorie foods
.
Eur J Neurosci
.
2009
;
30
(
8
):
1625
1635
.

293.

Malik
S
,
McGlone
F
,
Bedrossian
D
,
Dagher
A
.
Ghrelin modulates brain activity in areas that control appetitive behavior
.
Cell Metab
.
2008
;
7
(
5
):
400
409
.

294.

Hansson
C
,
Haage
D
,
Taube
M
,
Egecioglu
E
,
Salomé
N
,
Dickson
SL
.
Central administration of ghrelin alters emotional responses in rats: behavioural, electrophysiological and molecular evidence
.
Neuroscience
.
2011
;
180
:
201
211
.

295.

Alvarez-Crespo
M
,
Skibicka
KP
,
Farkas
I
,
Molnár
CS
,
Egecioglu
E
,
Hrabovszky
E
,
Liposits
Z
,
Dickson
SL
.
The amygdala as a neurobiological target for ghrelin in rats: neuroanatomical, electrophysiological and behavioral evidence
.
PLoS One
.
2012
;
7
(
10
):
e46321
.

296.

Dunn
AJ
,
Swiergiel
AH
,
Palamarchouk
V
.
Brain circuits involved in corticotropin-releasing factor-norepinephrine interactions during stress
.
Ann N Y Acad Sci
.
2004
;
1018
:
25
34
.

297.

Cabral
A
,
Valdivia
S
,
Fernandez
G
,
Reynaldo
M
,
Perello
M
.
Divergent neuronal circuitries underlying acute orexigenic effects of peripheral or central ghrelin: critical role of brain accessibility
.
J Neuroendocrinol
.
2014
;
26
(
8
):
542
554
.

298.

Sverrisdóttir
YB
,
Mogren
T
,
Kataoka
J
,
Janson
PO
,
Stener-Victorin
E
.
Is polycystic ovary syndrome associated with high sympathetic nerve activity and size at birth?
Am J Physiol Endocrinol Metab
.
2008
;
294
(
3
):
E576
E581
.

299.

Heider
U
,
Pedal
I
,
Spanel-Borowski
K
.
Increase in nerve fibers and loss of mast cells in polycystic and postmenopausal ovaries
.
Fertil Steril
.
2001
;
75
(
6
):
1141
1147
.

300.

Buller
K
,
Xu
Y
,
Dayas
C
,
Day
T
.
Dorsal and ventral medullary catecholamine cell groups contribute differentially to systemic interleukin-1beta-induced hypothalamic pituitary adrenal axis responses
.
Neuroendocrinology
.
2001
;
73
(
2
):
129
138
.

301.

Buller
KM
.
Neuroimmune stress responses: reciprocal connections between the hypothalamus and the brainstem
.
Stress
.
2003
;
6
(
1
):
11
17
.

302.

Lockie
SH
,
Dinan
T
,
Lawrence
AJ
,
Spencer
SJ
,
Andrews
ZB
.
Diet-induced obesity causes ghrelin resistance in reward processing tasks
.
Psychoneuroendocrinology
.
2015
;
62
:
114
120
.

303.

Egecioglu
E
,
Prieto-Garcia
L
,
Studer
E
,
Westberg
L
,
Jerlhag
E
.
The role of ghrelin signalling for sexual behaviour in male mice
.
Addict Biol
.
2016
;
21
(
2
):
348
359
.

304.

Schellekens
H
,
Dinan
TG
,
Cryan
JF
.
Ghrelin at the interface of obesity and reward
.
Vitam Horm
.
2013
;
91
:
285
323
.

305.

Chuang
JC
,
Perello
M
,
Sakata
I
,
Osborne-Lawrence
S
,
Savitt
JM
,
Lutter
M
,
Zigman
JM
.
Ghrelin mediates stress-induced food-reward behavior in mice
.
J Clin Invest
.
2011
;
121
(
7
):
2684
2692
.

306.

Egecioglu
E
,
Jerlhag
E
,
Salomé
N
,
Skibicka
KP
,
Haage
D
,
Bohlooly-Y
M
,
Andersson
D
,
Bjursell
M
,
Perrissoud
D
,
Engel
JA
,
Dickson
SL
.
Ghrelin increases intake of rewarding food in rodents
.
Addict Biol
.
2010
;
15
(
3
):
304
311
.

307.

Shimbara
T
,
Mondal
MS
,
Kawagoe
T
,
Toshinai
K
,
Koda
S
,
Yamaguchi
H
,
Date
Y
,
Nakazato
M
.
Central administration of ghrelin preferentially enhances fat ingestion
.
Neurosci Lett
.
2004
;
369
(
1
):
75
79
.

308.

Disse
E
,
Bussier
AL
,
Veyrat-Durebex
C
,
Deblon
N
,
Pfluger
PT
,
Tschöp
MH
,
Laville
M
,
Rohner-Jeanrenaud
F
.
Peripheral ghrelin enhances sweet taste food consumption and preference, regardless of its caloric content
.
Physiol Behav
.
2010
;
101
(
2
):
277
281
.

309.

Skibicka
KP
,
Hansson
C
,
Alvarez-Crespo
M
,
Friberg
PA
,
Dickson
SL
.
Ghrelin directly targets the ventral tegmental area to increase food motivation
.
Neuroscience
.
2011
;
180
:
129
137
.

310.

Andrews
ZB
,
Erion
D
,
Beiler
R
,
Liu
ZW
,
Abizaid
A
,
Zigman
J
,
Elsworth
JD
,
Savitt
JM
,
DiMarchi
R
,
Tschoep
M
,
Roth
RH
,
Gao
XB
,
Horvath
TL
.
Ghrelin promotes and protects nigrostriatal dopamine function via a UCP2-dependent mitochondrial mechanism
.
J Neurosci
.
2009
;
29
(
45
):
14057
14065
.

311.

Hull
EM
,
Muschamp
JW
,
Sato
S
.
Dopamine and serotonin: influences on male sexual behavior
.
Physiol Behav
.
2004
;
83
(
2
):
291
307
.

312.

Blackburn
JR
,
Pfaus
JG
,
Phillips
AG
.
Dopamine functions in appetitive and defensive behaviours
.
Prog Neurobiol
.
1992
;
39
(
3
):
247
279
.

313.

Jerlhag
E
,
Egecioglu
E
,
Dickson
SL
,
Douhan
A
,
Svensson
L
,
Engel
JA
.
Ghrelin administration into tegmental areas stimulates locomotor activity and increases extracellular concentration of dopamine in the nucleus accumbens
.
Addict Biol
.
2007
;
12
(
1
):
6
16
.

314.

Skibicka
KP
,
Hansson
C
,
Egecioglu
E
,
Dickson
SL
.
Role of ghrelin in food reward: impact of ghrelin on sucrose self-administration and mesolimbic dopamine and acetylcholine receptor gene expression
.
Addict Biol
.
2012
;
17
(
1
):
95
107
.

315.

Jerlhag
E
,
Egecioglu
E
,
Landgren
S
,
Salomé
N
,
Heilig
M
,
Moechars
D
,
Datta
R
,
Perrissoud
D
,
Dickson
SL
,
Engel
JA
.
Requirement of central ghrelin signaling for alcohol reward
.
Proc Natl Acad Sci USA
.
2009
;
106
(
27
):
11318
11323
.

316.

Jerlhag
E
,
Egecioglu
E
,
Dickson
SL
,
Engel
JA
.
Ghrelin receptor antagonism attenuates cocaine- and amphetamine-induced locomotor stimulation, accumbal dopamine release, and conditioned place preference
.
Psychopharmacology (Berl)
.
2010
;
211
(
4
):
415
422
.

317.

Jerlhag
E
,
Engel
JA
.
Ghrelin receptor antagonism attenuates nicotine-induced locomotor stimulation, accumbal dopamine release and conditioned place preference in mice
.
Drug Alcohol Depend
.
2011
;
117
(
2-3
):
126
131
.

318.

Wellman
PJ
,
Hollas
CN
,
Elliott
AE
.
Systemic ghrelin sensitizes cocaine-induced hyperlocomotion in rats
.
Regul Pept
.
2008
;
146
(
1-3
):
33
37
.

319.

Wellman
PJ
,
Clifford
PS
,
Rodriguez
J
,
Hughes
S
,
Eitan
S
,
Brunel
L
,
Fehrentz
JA
,
Martinez
J
.
Pharmacologic antagonism of ghrelin receptors attenuates development of nicotine induced locomotor sensitization in rats
.
Regul Pept
.
2011
;
172
(
1-3
):
77
80
.

320.

Abizaid
A
,
Mineur
YS
,
Roth
RH
,
Elsworth
JD
,
Sleeman
MW
,
Picciotto
MR
,
Horvath
TL
.
Reduced locomotor responses to cocaine in ghrelin-deficient mice
.
Neuroscience
.
2011
;
192
:
500
506
.

321.

Clifford
PS
,
Rodriguez
J
,
Schul
D
,
Hughes
S
,
Kniffin
T
,
Hart
N
,
Eitan
S
,
Brunel
L
,
Fehrentz
JA
,
Martinez
J
,
Wellman
PJ
.
Attenuation of cocaine-induced locomotor sensitization in rats sustaining genetic or pharmacologic antagonism of ghrelin receptors
.
Addict Biol
.
2012
;
17
(
6
):
956
963
.

322.

Dickson
SL
,
Hrabovszky
E
,
Hansson
C
,
Jerlhag
E
,
Alvarez-Crespo
M
,
Skibicka
KP
,
Molnar
CS
,
Liposits
Z
,
Engel
JA
,
Egecioglu
E
.
Blockade of central nicotine acetylcholine receptor signaling attenuate ghrelin-induced food intake in rodents
.
Neuroscience
.
2010
;
171
(
4
):
1180
1186
.

323.

Kaur
S
,
Ryabinin
AE
.
Ghrelin receptor antagonism decreases alcohol consumption and activation of perioculomotor urocortin-containing neurons
.
Alcohol Clin Exp Res
.
2010
;
34
(
9
):
1525
1534
.

324.

Burkett
JP
,
Young
LJ
.
The behavioral, anatomical and pharmacological parallels between social attachment, love and addiction
.
Psychopharmacology (Berl)
.
2012
;
224
(
1
):
1
26
.

325.

Pitchers
KK
,
Balfour
ME
,
Lehman
MN
,
Richtand
NM
,
Yu
L
,
Coolen
LM
.
Neuroplasticity in the mesolimbic system induced by natural reward and subsequent reward abstinence
.
Biol Psychiatry
.
2010
;
67
(
9
):
872
879
.

326.

Pitchers
KK
,
Schmid
S
,
Di Sebastiano
AR
,
Wang
X
,
Laviolette
SR
,
Lehman
MN
,
Coolen
LM
.
Natural reward experience alters AMPA and NMDA receptor distribution and function in the nucleus accumbens
.
PLoS One
.
2012
;
7
(
4
):
e34700
.

327.

Pitchers
KK
,
Frohmader
KS
,
Vialou
V
,
Mouzon
E
,
Nestler
EJ
,
Lehman
MN
,
Coolen
LM
.
ΔFosB in the nucleus accumbens is critical for reinforcing effects of sexual reward
.
Genes Brain Behav
.
2010
;
9
(
7
):
831
840
.

328.

Shah
SN
,
Nyby
JG
.
Ghrelin’s quick inhibition of androgen-dependent behaviors of male house mice (Mus musculus)
.
Horm Behav
.
2010
;
57
(
3
):
291
296
.

329.

Prieto-Garcia
L
,
Egecioglu
E
,
Studer
E
,
Westberg
L
,
Jerlhag
E
.
Ghrelin and GHS-R1A signaling within the ventral and laterodorsal tegmental area regulate sexual behavior in sexually naïve male mice
.
Psychoneuroendocrinology
.
2015
;
62
:
392
402
.

330.

Govic
A
,
Levay
EA
,
Hazi
A
,
Penman
J
,
Kent
S
,
Paolini
AG
.
Alterations in male sexual behaviour, attractiveness and testosterone levels induced by an adult-onset calorie restriction regimen
.
Behav Brain Res
.
2008
;
190
(
1
):
140
146
.

331.

García
MC
,
López
M
,
Alvarez
CV
,
Casanueva
F
,
Tena-Sempere
M
,
Diéguez
C
.
Role of ghrelin in reproduction
.
Reproduction
.
2007
;
133
(
3
):
531
540
.

332.

Furuta
M
,
Funabashi
T
,
Kimura
F
.
Intracerebroventricular administration of ghrelin rapidly suppresses pulsatile luteinizing hormone secretion in ovariectomized rats
.
Biochem Biophys Res Commun
.
2001
;
288
(
4
):
780
785
.

333.

Fernández-Fernández
R
,
Tena-Sempere
M
,
Aguilar
E
,
Pinilla
L
.
Ghrelin effects on gonadotropin secretion in male and female rats
.
Neurosci Lett
.
2004
;
362
(
2
):
103
107
.

334.

Inhoff
T
,
Mönnikes
H
,
Noetzel
S
,
Stengel
A
,
Goebel
M
,
Dinh
QT
,
Riedl
A
,
Bannert
N
,
Wisser
AS
,
Wiedenmann
B
,
Klapp
BF
,
Taché
Y
,
Kobelt
P
.
Desacyl ghrelin inhibits the orexigenic effect of peripherally injected ghrelin in rats
.
Peptides
.
2008
;
29
(
12
):
2159
2168
.

335.

De Souza
MJ
,
Leidy
HJ
,
O’Donnell
E
,
Lasley
B
,
Williams
NI
.
Fasting ghrelin levels in physically active women: relationship with menstrual disturbances and metabolic hormones
.
J Clin Endocrinol Metab
.
2004
;
89
(
7
):
3536
3542
.

336.

Schneider
LF
,
Monaco
SE
,
Warren
MP
.
Elevated ghrelin level in women of normal weight with amenorrhea is related to disordered eating
.
Fertil Steril
.
2008
;
90
(
1
):
121
128
.

337.

Christo
K
,
Cord
J
,
Mendes
N
,
Miller
KK
,
Goldstein
MA
,
Klibanski
A
,
Misra
M
.
Acylated ghrelin and leptin in adolescent athletes with amenorrhea, eumenorrheic athletes and controls: a cross-sectional study
.
Clin Endocrinol (Oxf)
.
2008
;
69
(
4
):
628
633
.

338.

Wahab
F
,
Aziz
F
,
Irfan
S
,
Zaman
WU
,
Shahab
M
.
Short-term fasting attenuates the response of the HPG axis to kisspeptin challenge in the adult male rhesus monkey (Macaca mulatta)
.
Life Sci
.
2008
;
83
(
19-20
):
633
637
.

339.

Mauvais-Jarvis
F
,
Clegg
DJ
,
Hevener
AL
.
The role of estrogens in control of energy balance and glucose homeostasis
.
Endocr Rev
.
2013
;
34
(
3
):
309
338
.

340.

Shansky
RM
,
Hamo
C
,
Hof
PR
,
Lou
W
,
McEwen
BS
,
Morrison
JH
.
Estrogen promotes stress sensitivity in a prefrontal cortex-amygdala pathway
.
Cereb Cortex
.
2010
;
20
(
11
):
2560
2567
.

341.

Komesaroff
PA
,
Esler
MD
,
Sudhir
K
.
Estrogen supplementation attenuates glucocorticoid and catecholamine responses to mental stress in perimenopausal women
.
J Clin Endocrinol Metab
.
1999
;
84
(
2
):
606
610
.

342.

Hlatky
MA
,
Boothroyd
D
,
Vittinghoff
E
,
Sharp
P
,
Whooley
MA
;
Heart and Estrogen/Progestin Replacement Study (HERS) Research Group
.
Quality-of-life and depressive symptoms in postmenopausal women after receiving hormone therapy: results from the Heart and Estrogen/Progestin Replacement Study (HERS) trial
.
JAMA
.
2002
;
287
(
5
):
591
597
.

343.

Michopoulos
V
,
Wilson
ME
.
Body weight decreases induced by estradiol in female rhesus monkeys are dependent upon social status
.
Physiol Behav
.
2011
;
102
(
3-4
):
382
388
.

344.

Reding
K
,
Michopoulos
V
,
Wallen
K
,
Sanchez
M
,
Wilson
ME
,
Toufexis
D
.
Social status modifies estradiol activation of sociosexual behavior in female rhesus monkeys
.
Horm Behav
.
2012
;
62
(
5
):
612
620
.

345.

Michopoulos
V
,
Embree
M
,
Reding
K
,
Sanchez
MM
,
Toufexis
D
,
Votaw
JR
,
Voll
RJ
,
Goodman
MM
,
Rivier
J
,
Wilson
ME
,
Berga
SL
.
CRH receptor antagonism reverses the effect of social subordination upon central GABAA receptor binding in estradiol-treated ovariectomized female rhesus monkeys
.
Neuroscience
.
2013
;
250
:
300
308
.

346.

Shansky
RM
,
Glavis-Bloom
C
,
Lerman
D
,
McRae
P
,
Benson
C
,
Miller
K
,
Cosand
L
,
Horvath
TL
,
Arnsten
AF
.
Estrogen mediates sex differences in stress-induced prefrontal cortex dysfunction
.
Mol Psychiatry
.
2004
;
9
(
5
):
531
538
.

347.

Wood
GE
,
Shors
TJ
.
Stress facilitates classical conditioning in males, but impairs classical conditioning in females through activational effects of ovarian hormones
.
Proc Natl Acad Sci USA
.
1998
;
95
(
7
):
4066
4071
.

348.

Bowman
RE
,
Ferguson
D
,
Luine
VN
.
Effects of chronic restraint stress and estradiol on open field activity, spatial memory, and monoaminergic neurotransmitters in ovariectomized rats
.
Neuroscience
.
2002
;
113
(
2
):
401
410
.

349.

Walf
AA
,
Frye
CA
.
A review and update of mechanisms of estrogen in the hippocampus and amygdala for anxiety and depression behavior
.
Neuropsychopharmacology
.
2006
;
31
(
6
):
1097
1111
.

350.

Brunner
RL
,
Gass
M
,
Aragaki
A
,
Hays
J
,
Granek
I
,
Woods
N
,
Mason
E
,
Brzyski
R
,
Ockene
J
,
Assaf
A
,
LaCroix
A
,
Matthews
K
,
Wallace
R
;
Women’s Health Initiative Investigators
.
Effects of conjugated equine estrogen on health-related quality of life in postmenopausal women with hysterectomy: results from the Women’s Health Initiative Randomized Clinical Trial
.
Arch Intern Med
.
2005
;
165
(
17
):
1976
1986
.

351.

Hays
J
,
Ockene
JK
,
Brunner
RL
,
Kotchen
JM
,
Manson
JE
,
Patterson
RE
,
Aragaki
AK
,
Shumaker
SA
,
Brzyski
RG
,
LaCroix
AZ
,
Granek
IA
,
Valanis
BG
;
Women’s Health Initiative Investigators
.
Effects of estrogen plus progestin on health-related quality of life
.
N Engl J Med
.
2003
;
348
(
19
):
1839
1854
.

352.

Handa
RJ
,
Mani
SK
,
Uht
RM
.
Estrogen receptors and the regulation of neural stress responses
.
Neuroendocrinology
.
2012
;
96
(
2
):
111
118
.

353.

Heine
PA
,
Taylor
JA
,
Iwamoto
GA
,
Lubahn
DB
,
Cooke
PS
.
Increased adipose tissue in male and female estrogen receptor-alpha knockout mice
.
Proc Natl Acad Sci USA
.
2000
;
97
(
23
):
12729
12734
.

354.

Smith
JT
,
Cunningham
MJ
,
Rissman
EF
,
Clifton
DK
,
Steiner
RA
.
Regulation of Kiss1 gene expression in the brain of the female mouse
.
Endocrinology
.
2005
;
146
(
9
):
3686
3692
.

355.

Lanfranco
F
,
Bonelli
L
,
Baldi
M
,
Me
E
,
Broglio
F
,
Ghigo
E
.
Acylated ghrelin inhibits spontaneous luteinizing hormone pulsatility and responsiveness to naloxone but not that to gonadotropin-releasing hormone in young men: evidence for a central inhibitory action of ghrelin on the gonadal axis
.
J Clin Endocrinol Metab
.
2008
;
93
(
9
):
3633
3639
.

356.

Grossman
A
,
Moult
PJ
,
Gaillard
RC
,
Delitala
G
,
Toff
WD
,
Rees
LH
,
Besser
GM
.
The opioid control of LH and FSH release: effects of a met-enkephalin analogue and naloxone
.
Clin Endocrinol (Oxf)
.
1981
;
14
(
1
):
41
47
.

357.

Ben-Jonathan
N
.
Dopamine: a prolactin-inhibiting hormone
.
Endocr Rev
.
1985
;
6
(
4
):
564
589
.

358.

Torner
L
.
Actions of prolactin in the brain: from physiological adaptations to stress and neurogenesis to psychopathology
.
Front Endocrinol (Lausanne)
.
2016
;
7
:
25
.

359.

Grattan
DR
,
Kokay
IC
.
Prolactin: a pleiotropic neuroendocrine hormone
.
J Neuroendocrinol
.
2008
;
20
(
6
):
752
763
.

360.

Torner
L
,
Toschi
N
,
Nava
G
,
Clapp
C
,
Neumann
ID
.
Increased hypothalamic expression of prolactin in lactation: involvement in behavioural and neuroendocrine stress responses
.
Eur J Neurosci
.
2002
;
15
(
8
):
1381
1389
.

361.

Torner
L
,
Neumann
ID
.
The brain prolactin system: involvement in stress response adaptations in lactation
.
Stress
.
2002
;
5
(
4
):
249
257
.

362.

Faron-Górecka
A
,
Kuśmider
M
,
Kolasa
M
,
Zurawek
D
,
Gruca
P
,
Papp
M
,
Szafran
K
,
Solich
J
,
Pabian
P
,
Romańska
I
,
Antkiewicz-Michaluk
L
,
Dziedzicka-Wasylewska
M
.
Prolactin and its receptors in the chronic mild stress rat model of depression
.
Brain Res
.
2014
;
1555
:
48
59
.

363.

Koike
K
,
Miyake
A
,
Aono
T
,
Sakumoto
T
,
Ohmichi
M
,
Yamaguchi
M
,
Tanizawa
O
.
Effect of prolactin on the secretion of hypothalamic GnRH and pituitary gonadotropins
.
Horm Res
.
1991
;
35
(
Suppl 1
):
5
12
.

364.

Matsuzaki
T
,
Azuma
K
,
Irahara
M
,
Yasui
T
,
Aono
T
.
Mechanism of anovulation in hyperprolactinemic amenorrhea determined by pulsatile gonadotropin-releasing hormone injection combined with human chorionic gonadotropin
.
Fertil Steril
.
1994
;
62
(
6
):
1143
1149
.

365.

Cohen-Becker
IR
,
Selmanoff
M
,
Wise
PM
.
Hyperprolactinemia alters the frequency and amplitude of pulsatile luteinizing hormone secretion in the ovariectomized rat
.
Neuroendocrinology
.
1986
;
42
(
4
):
328
333
.

366.

Patel
SS
,
Bamigboye
V
.
Hyperprolactinaemia
.
J Obstet Gynaecol
.
2007
;
27
(
5
):
455
459
.

367.

Messini
CI
,
Dafopoulos
K
,
Chalvatzas
N
,
Georgoulias
P
,
Anifandis
G
,
Messinis
IE
.
Effect of ghrelin and metoclopramide on prolactin secretion in normal women
.
J Endocrinol Invest
.
2011
;
34
(
4
):
276
279
.

368.

Rubinfeld
H
,
Hadani
M
,
Taylor
JE
,
Dong
JZ
,
Comstock
J
,
Shen
Y
,
DeOliveira
D
,
Datta
R
,
Culler
MD
,
Shimon
I
.
Novel ghrelin analogs with improved affinity for the GH secretagogue receptor stimulate GH and prolactin release from human pituitary cells
.
Eur J Endocrinol
.
2004
;
151
(
6
):
787
795
.

369.

Broglio
F
,
Gottero
C
,
Prodam
F
,
Gauna
C
,
Muccioli
G
,
Papotti
M
,
Abribat
T
,
Van Der Lely
AJ
,
Ghigo
E
.
Non-acylated ghrelin counteracts the metabolic but not the neuroendocrine response to acylated ghrelin in humans
.
J Clin Endocrinol Metab
.
2004
;
89
(
6
):
3062
3065
.

370.

Iqbal
J
,
Kurose
Y
,
Canny
B
,
Clarke
IJ
.
Effects of central infusion of ghrelin on food intake and plasma levels of growth hormone, luteinizing hormone, prolactin, and cortisol secretion in sheep
.
Endocrinology
.
2006
;
147
(
1
):
510
519
.

371.

Tena-Sempere
M
,
Aguilar
E
,
Fernandez-Fernandez
R
,
Pinilla
L
.
Ghrelin inhibits prolactin secretion in prepubertal rats
.
Neuroendocrinology
.
2004
;
79
(
3
):
133
141
.

372.

Smith
RG
,
Van der Ploeg
LH
,
Howard
AD
,
Feighner
SD
,
Cheng
K
,
Hickey
GJ
,
Wyvratt
MJ
, Jr
,
Fisher
MH
,
Nargund
RP
,
Patchett
AA
.
Peptidomimetic regulation of growth hormone secretion
.
Endocr Rev
.
1997
;
18
(
5
):
621
645
.

373.

Córdoba-Chacón
J
,
Gahete
MD
,
Pozo-Salas
AI
,
Martínez-Fuentes
AJ
,
de Lecea
L
,
Gracia-Navarro
F
,
Kineman
RD
,
Castaño
JP
,
Luque
RM
.
Cortistatin is not a somatostatin analogue but stimulates prolactin release and inhibits GH and ACTH in a gender-dependent fashion: potential role of ghrelin
.
Endocrinology
.
2011
;
152
(
12
):
4800
4812
.

374.

Yang
H
,
Dixit
VD
,
Patel
K
,
Vandanmagsar
B
,
Collins
G
,
Sun
Y
,
Smith
RG
,
Taub
DD
.
Reduction in hypophyseal growth hormone and prolactin expression due to deficiency in ghrelin receptor signaling is associated with Pit-1 suppression: relevance to the immune system
.
Brain Behav Immun
.
2008
;
22
(
8
):
1138
1145
.

375.

Lorenzi
T
,
Meli
R
,
Marzioni
D
,
Morroni
M
,
Baragli
A
,
Castellucci
M
,
Gualillo
O
,
Muccioli
G
.
Ghrelin: a metabolic signal affecting the reproductive system
.
Cytokine Growth Factor Rev
.
2009
;
20
(
2
):
137
152
.

376.

Muccioli
G
,
Lorenzi
T
,
Lorenzi
M
,
Ghè
C
,
Arnoletti
E
,
Raso
GM
,
Castellucci
M
,
Gualillo
O
,
Meli
R
.
Beyond the metabolic role of ghrelin: a new player in the regulation of reproductive function
.
Peptides
.
2011
;
32
(
12
):
2514
2521
.

377.

Barreiro
ML
,
Suominen
JS
,
Gaytán
F
,
Pinilla
L
,
Chopin
LK
,
Casanueva
FF
,
Diéguez
C
,
Aguilar
E
,
Toppari
J
,
Tena-Sempere
M
.
Developmental, stage-specific, and hormonally regulated expression of growth hormone secretagogue receptor messenger RNA in rat testis
.
Biol Reprod
.
2003
;
68
(
5
):
1631
1640
.

378.

Tena-Sempere
M
,
Barreiro
ML
,
González
LC
,
Gaytán
F
,
Zhang
FP
,
Caminos
JE
,
Pinilla
L
,
Casanueva
FF
,
Diéguez
C
,
Aguilar
E
.
Novel expression and functional role of ghrelin in rat testis
.
Endocrinology
.
2002
;
143
(
2
):
717
725
.

379.

Rak-Mardyła
A
,
Gregoraszczuk
EL
,
Karpeta
A
,
Duda
M
.
Expression of ghrelin and the ghrelin receptor in different stages of porcine corpus luteum development and the inhibitory effects of ghrelin on progesterone secretion, 3β-hydroxysteroid dehydrogenase (3β-honestly significant difference (HSD)) activity and protein expression
.
Theriogenology
.
2012
;
77
(
8
):
1505
1512
.

380.

Gaytan
F
,
Barreiro
ML
,
Chopin
LK
,
Herington
AC
,
Morales
C
,
Pinilla
L
,
Casanueva
FF
,
Aguilar
E
,
Diéguez
C
,
Tena-Sempere
M
.
Immunolocalization of ghrelin and its functional receptor, the type 1a growth hormone secretagogue receptor, in the cyclic human ovary
.
J Clin Endocrinol Metab
.
2003
;
88
(
2
):
879
887
.

381.

Gnanapavan
S
,
Kola
B
,
Bustin
SA
,
Morris
DG
,
McGee
P
,
Fairclough
P
,
Bhattacharya
S
,
Carpenter
R
,
Grossman
AB
,
Korbonits
M
.
The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans
.
J Clin Endocrinol Metab
.
2002
;
87
(
6
):
2988
.

382.

Rannikko
A
,
Penttilä
TL
,
Zhang
FP
,
Toppari
J
,
Parvinen
M
,
Huhtaniemi
I
.
Stage-specific expression of the FSH receptor gene in the prepubertal and adult rat seminiferous epithelium
.
J Endocrinol
.
1996
;
151
(
1
):
29
35
.

383.

Heckert
L
,
Griswold
MD
.
Expression of the FSH receptor in the testis
.
Recent Prog Horm Res
.
1993
;
48
:
61
77
.

384.

Zhu
CC
,
Zhang
H
,
Zhang
JS
,
Li
Z
,
Zhao
J
,
Li
W
,
Zhang
YQ
.
Inhibition of ghrelin signaling improves the reproductive phenotype of male ob/ob mouse
.
Fertil Steril
.
2013
;
99
(
3
):
918
926
.

385.

Ishikawa
T
,
Fujioka
H
,
Ishimura
T
,
Takenaka
A
,
Fujisawa
M
.
Ghrelin expression in human testis and serum testosterone level
.
J Androl
.
2007
;
28
(
2
):
320
324
.

386.

Barreiro
ML
,
Gaytán
F
,
Caminos
JE
,
Pinilla
L
,
Casanueva
FF
,
Aguilar
E
,
Diéguez
C
,
Tena-Sempere
M
.
Cellular location and hormonal regulation of ghrelin expression in rat testis
.
Biol Reprod
.
2002
;
67
(
6
):
1768
1776
.

387.

Garcia
JM
,
Chen
JA
,
Guillory
B
,
Donehower
LA
,
Smith
RG
,
Lamb
DJ
.
Ghrelin prevents cisplatin-induced testicular damage by facilitating repair of DNA double strand breaks through activation of p53 in mice
.
Biol Reprod
.
2015
;
93
(
1
):
24
.

388.

Li
W
,
Zeng
Y
,
Zhao
J
,
Zhu
CJ
,
Hou
WG
,
Zhang
S
.
Upregulation and nuclear translocation of testicular ghrelin protects differentiating spermatogonia from ionizing radiation injury
.
Cell Death Dis
.
2014
;
5
:
e1248
.

389.

Taati
M
,
Moghadasi
M
,
Dezfoulian
O
,
Asadian
P
,
Kheradmand
A
,
Abbasi
M
,
Zendehdel
M
.
The effect of ghrelin pretreatment on epididymal sperm quality and tissue antioxidant enzyme activities after testicular ischemia/reperfusion in rats
.
J Physiol Biochem
.
2012
;
68
(
1
):
91
97
.

390.

Kheradmand
A
,
Dezfoulian
O
,
Tarrahi
MJ
.
Ghrelin attenuates heat-induced degenerative effects in the rat testis
.
Regul Pept
.
2011
;
167
(
1
):
97
104
.

391.

Moretti
E
,
Vindigni
C
,
Tripodi
SA
,
Mazzi
L
,
Nuti
R
,
Figura
N
,
Collodel
G
.
Immunolocalisation of ghrelin and obestatin in human testis, seminal vesicles, prostate and spermatozoa
.
Andrologia
.
2014
;
46
(
9
):
979
985
.

392.

Roa
J
,
García-Galiano
D
,
Castellano
JM
,
Gaytan
F
,
Pinilla
L
,
Tena-Sempere
M
.
Metabolic control of puberty onset: new players, new mechanisms
.
Mol Cell Endocrinol
.
2010
;
324
(
1-2
):
87
94
.

393.

Shalitin
S
,
Phillip
M
.
Role of obesity and leptin in the pubertal process and pubertal growth--a review
.
Int J Obes Relat Metab Disord
.
2003
;
27
(
8
):
869
874
.

394.

Fernández-Fernández
R
,
Navarro
VM
,
Barreiro
ML
,
Vigo
EM
,
Tovar
S
,
Sirotkin
AV
,
Casanueva
FF
,
Aguilar
E
,
Dieguez
C
,
Pinilla
L
,
Tena-Sempere
M
.
Effects of chronic hyperghrelinemia on puberty onset and pregnancy outcome in the rat
.
Endocrinology
.
2005
;
146
(
7
):
3018
3025
.

395.

Soriano-Guillén
L
,
Barrios
V
,
Chowen
JA
,
Sánchez
I
,
Vila
S
,
Quero
J
,
Argente
J
.
Ghrelin levels from fetal life through early adulthood: relationship with endocrine and metabolic and anthropometric measures
.
J Pediatr
.
2004
;
144
(
1
):
30
35
.

396.

Whatmore
AJ
,
Hall
CM
,
Jones
J
,
Westwood
M
,
Clayton
PE
.
Ghrelin concentrations in healthy children and adolescents
.
Clin Endocrinol (Oxf)
.
2003
;
59
(
5
):
649
654
.

397.

Divall
SA
,
Williams
TR
,
Carver
SE
,
Koch
L
,
Brüning
JC
,
Kahn
CR
,
Wondisford
F
,
Radovick
S
,
Wolfe
A
.
Divergent roles of growth factors in the GnRH regulation of puberty in mice
.
J Clin Invest
.
2010
;
120
(
8
):
2900
2909
.

398.

Prader
A
.
Delayed adolescence
.
Clin Endocrinol Metab
.
1975
;
4
(
1
):
143
155
.

399.

Gordon
M
,
Crouthamel
C
,
Post
EM
,
Richman
RA
.
Psychosocial aspects of constitutional short stature: social competence, behavior problems, self-esteem, and family functioning
.
J Pediatr
.
1982
;
101
(
3
):
477
480
.

400.

Sandberg
DE
,
Voss
LD
.
The psychosocial consequences of short stature: a review of the evidence
.
Best Pract Res Clin Endocrinol Metab
.
2002
;
16
(
3
):
449
463
.

401.

Visser-van Balen
H
,
Sinnema
G
,
Geenen
R
.
Growing up with idiopathic short stature: psychosocial development and hormone treatment; a critical review
.
Arch Dis Child
.
2006
;
91
(
5
):
433
439
.

402.

Sen
TA
,
Şimşek
DG
,
Darcan
S
,
Coker
M
.
Ghrelin levels in children with constitutional delay of growth and puberty
.
J Clin Res Pediatr Endocrinol
.
2010
;
2
(
3
):
117
121
.

403.

El-Eshmawy
MM
,
Abdel Aal
IA
,
El Hawary
AK
.
Association of ghrelin and leptin with reproductive hormones in constitutional delay of growth and puberty
.
Reprod Biol Endocrinol
.
2010
;
8
:
153
.

404.

Harrison
JL
,
Adam
CL
,
Brown
YA
,
Wallace
JM
,
Aitken
RP
,
Lea
RG
,
Miller
DW
.
An immunohistochemical study of the localization and developmental expression of ghrelin and its functional receptor in the ovine placenta
.
Reprod Biol Endocrinol
.
2007
;
5
:
25
.

405.

Nakahara
K
,
Nakagawa
M
,
Baba
Y
,
Sato
M
,
Toshinai
K
,
Date
Y
,
Nakazato
M
,
Kojima
M
,
Miyazato
M
,
Kaiya
H
,
Hosoda
H
,
Kangawa
K
,
Murakami
N
.
Maternal ghrelin plays an important role in rat fetal development during pregnancy
.
Endocrinology
.
2006
;
147
(
3
):
1333
1342
.

406.

Inoue
Y
,
Nakahara
K
,
Kangawa
K
,
Murakami
N
.
Transitional change in rat fetal cell proliferation in response to ghrelin and des-acyl ghrelin during the last stage of pregnancy
.
Biochem Biophys Res Commun
.
2010
;
393
(
3
):
455
460
.

407.

Diedrich
K
,
Fauser
BC
,
Devroey
P
,
Griesinger
G
;
Evian Annual Reproduction (EVAR) Workshop Group
.
The role of the endometrium and embryo in human implantation
.
Hum Reprod Update
.
2007
;
13
(
4
):
365
377
.

408.

Simón
C
,
Moreno
C
,
Remohí
J
,
Pellicer
A
.
Cytokines and embryo implantation
.
J Reprod Immunol
.
1998
;
39
(
1-2
):
117
131
.

409.

Simón
C
,
Martín
JC
,
Pellicer
A
.
Paracrine regulators of implantation
.
Best Pract Res Clin Obstet Gynaecol
.
2000
;
14
(
5
):
815
826
.

410.

Achache
H
,
Revel
A
.
Endometrial receptivity markers, the journey to successful embryo implantation
.
Hum Reprod Update
.
2006
;
12
(
6
):
731
746
.

411.

Michael
AE
,
Papageorghiou
AT
.
Potential significance of physiological and pharmacological glucocorticoids in early pregnancy
.
Hum Reprod Update
.
2008
;
14
(
5
):
497
517
.

412.

Lindsay
JR
,
Nieman
LK
.
The hypothalamic-pituitary-adrenal axis in pregnancy: challenges in disease detection and treatment
.
Endocr Rev
.
2005
;
26
(
6
):
775
799
.

413.

Cottrell
EC
,
Seckl
JR
.
Prenatal stress, glucocorticoids and the programming of adult disease
.
Front Behav Neurosci
.
2009
;
3
:
19
.

414.

Tanaka
K
,
Minoura
H
,
Isobe
T
,
Yonaha
H
,
Kawato
H
,
Wang
DF
,
Yoshida
T
,
Kojima
M
,
Kangawa
K
,
Toyoda
N
.
Ghrelin is involved in the decidualization of human endometrial stromal cells
.
J Clin Endocrinol Metab
.
2003
;
88
(
5
):
2335
2340
.

415.

Kawamura
K
,
Sato
N
,
Fukuda
J
,
Kodama
H
,
Kumagai
J
,
Tanikawa
H
,
Nakamura
A
,
Honda
Y
,
Sato
T
,
Tanaka
T
.
Ghrelin inhibits the development of mouse preimplantation embryos in vitro
.
Endocrinology
.
2003
;
144
(
6
):
2623
2633
.

416.

Gualillo
O
,
Caminos
J
,
Blanco
M
,
Garcìa-Caballero
T
,
Kojima
M
,
Kangawa
K
,
Dieguez
C
,
Casanueva
F
.
Ghrelin, a novel placental-derived hormone
.
Endocrinology
.
2001
;
142
(
2
):
788
794
.

417.

Papotti
M
,
Ghè
C
,
Cassoni
P
,
Catapano
F
,
Deghenghi
R
,
Ghigo
E
,
Muccioli
G
.
Growth hormone secretagogue binding sites in peripheral human tissues
.
J Clin Endocrinol Metab
.
2000
;
85
(
10
):
3803
3807
.

418.

Du
C
,
Li
H
,
Cao
G
,
Xilingaowa
,
Wang
C
,
Li
C
.
Expression of the orexigenic peptide ghrelin and the type 1a growth hormone secretagogue receptor in sheep oocytes and pre-implantation embryos produced in vitro
.
Reprod Domest Anim
.
2010
;
45
(
1
):
92
98
.

419.

Speakman
JR
.
The physiological costs of reproduction in small mammals
.
Philos Trans R Soc Lond B Biol Sci
.
2008
;
363
(
1490
):
375
398
.

420.

Tawadros
N
,
Salamonsen
LA
,
Dimitriadis
E
,
Chen
C
.
Facilitation of decidualization by locally produced ghrelin in the human endometrium
.
Mol Hum Reprod
.
2007
;
13
(
7
):
483
489
.

421.

Rak-Mardyła
A
,
Gregoraszczuk
E
.
Effect of ghrelin on proliferation, apoptosis and secretion of progesterone and hCG in the placental JEG-3 cell line
.
Reprod Biol
.
2010
;
10
(
2
):
159
165
.

422.

Zhang
K
,
Wei
HX
,
Zhang
YH
,
Wang
SH
,
Li
Y
,
Dai
YP
,
Li
N
.
Effects of ghrelin on in vitro development of porcine in vitro fertilized and parthenogenetic embryos
.
J Reprod Dev
.
2007
;
53
(
3
):
647
653
.

423.

Wang
Z
,
Lin
P
,
Yu
S
.
Effects of ghrelin on developmental competence and gene expression of in vitro fertilized ovine embryos
.
Theriogenology
.
2013
;
79
(
4
):
695
701
.

424.

Suzuki
H
,
Sasaki
Y
,
Shimizu
M
,
Matsuzaki
M
,
Hashizume
T
,
Kuwayama
H
.
Ghrelin and leptin did not improve meiotic maturation of porcine oocytes cultured in vitro
.
Reprod Domest Anim
.
2010
;
45
(
5
):
927
930
.

425.

Dovolou
E
,
Periquesta
E
,
Messinis
IE
,
Tsiligianni
T
,
Dafopoulos
K
,
Gutierrez-Adan
A
,
Amiridis
GS
.
Daily supplementation with ghrelin improves in vitro bovine blastocysts formation rate and alters gene expression related to embryo quality
.
Theriogenology
.
2014
;
81
(
4
):
565
571
.

426.

Sominsky
L
,
Spencer
SJ
.
Eating behavior and stress: a pathway to obesity
.
Front Psychol
.
2014
;
5
:
434
.

427.

Feuer
S
,
Rinaudo
P
.
Preimplantation stress and development
.
Birth Defects Res C Embryo Today
.
2012
;
96
(
4
):
299
314
.

428.

Fuglsang
J
,
Skjaerbaek
C
,
Espelund
U
,
Frystyk
J
,
Fisker
S
,
Flyvbjerg
A
,
Ovesen
P
.
Ghrelin and its relationship to growth hormones during normal pregnancy
.
Clin Endocrinol (Oxf)
.
2005
;
62
(
5
):
554
559
.

429.

Palik
E
,
Baranyi
E
,
Melczer
Z
,
Audikovszky
M
,
Szöcs
A
,
Winkler
G
,
Cseh
K
.
Elevated serum acylated (biologically active) ghrelin and resistin levels associate with pregnancy-induced weight gain and insulin resistance
.
Diabetes Res Clin Pract
.
2007
;
76
(
3
):
351
357
.

430.

Shibata
K
,
Hosoda
H
,
Kojima
M
,
Kangawa
K
,
Makino
Y
,
Makino
I
,
Kawarabayashi
T
,
Futagami
K
,
Gomita
Y
.
Regulation of ghrelin secretion during pregnancy and lactation in the rat: possible involvement of hypothalamus
.
Peptides
.
2004
;
25
(
2
):
279
287
.

431.

Tham
E
,
Liu
J
,
Innis
S
,
Thompson
D
,
Gaylinn
BD
,
Bogarin
R
,
Haim
A
,
Thorner
MO
,
Chanoine
JP
.
Acylated ghrelin concentrations are markedly decreased during pregnancy in mothers with and without gestational diabetes: relationship with cholinesterase
.
Am J Physiol Endocrinol Metab
.
2009
;
296
(
5
):
E1093
E1100
.

432.

De Vriese
C
,
Gregoire
F
,
Lema-Kisoka
R
,
Waelbroeck
M
,
Robberecht
P
,
Delporte
C
.
Ghrelin degradation by serum and tissue homogenates: identification of the cleavage sites
.
Endocrinology
.
2004
;
145
(
11
):
4997
5005
.

433.

Slattery
DA
,
Neumann
ID
.
No stress please! Mechanisms of stress hyporesponsiveness of the maternal brain
.
J Physiol
.
2008
;
586
(
2
):
377
385
.

434.

Douglas
AJ
,
Brunton
PJ
,
Bosch
OJ
,
Russell
JA
,
Neumann
ID
.
Neuroendocrine responses to stress in mice: hyporesponsiveness in pregnancy and parturition
.
Endocrinology
.
2003
;
144
(
12
):
5268
5276
.

435.

Kammerer
M
,
Adams
D
,
Castelberg Bv
B
,
Glover
V
.
Pregnant women become insensitive to cold stress
.
BMC Pregnancy Childbirth
.
2002
;
2
(
1
):
8
.

436.

Altemus
M
,
Deuster
PA
,
Galliven
E
,
Carter
CS
,
Gold
PW
.
Suppression of hypothalmic-pituitary-adrenal axis responses to stress in lactating women
.
J Clin Endocrinol Metab
.
1995
;
80
(
10
):
2954
2959
.

437.

Schulte
HM
,
Weisner
D
,
Allolio
B
.
The corticotrophin releasing hormone test in late pregnancy: lack of adrenocorticotrophin and cortisol response
.
Clin Endocrinol (Oxf)
.
1990
;
33
(
1
):
99
106
.

438.

Magiakou
MA
,
Mastorakos
G
,
Rabin
D
,
Dubbert
B
,
Gold
PW
,
Chrousos
GP
.
Hypothalamic corticotropin-releasing hormone suppression during the postpartum period: implications for the increase in psychiatric manifestations at this time
.
J Clin Endocrinol Metab
.
1996
;
81
(
5
):
1912
1917
.

439.

Magiakou
MA
,
Mastorakos
G
,
Rabin
D
,
Margioris
AN
,
Dubbert
B
,
Calogero
AE
,
Tsigos
C
,
Munson
PJ
,
Chrousos
GP
.
The maternal hypothalamic-pituitary-adrenal axis in the third trimester of human pregnancy
.
Clin Endocrinol (Oxf)
.
1996
;
44
(
4
):
419
428
.

440.

Li
L
,
Ferin
M
,
Sauer
MV
,
Lobo
RA
.
Serum and follicular fluid ghrelin levels negatively reflect human oocyte quality and in vitro embryo development
.
Fertil Steril
.
2011
;
96
(
5
):
1116
1120
.

441.

Inal
HA
,
Yilmaz
N
,
Gorkem
U
,
Oruc
AS
,
Timur
H
.
The impact of follicular fluid adiponectin and ghrelin levels based on BMI on IVF outcomes in PCOS
.
J Endocrinol Invest
.
2016
;
39
(
4
):
431
437
.

442.

Várnagy
A
,
Bódis
J
,
Kovács
GL
,
Sulyok
E
,
Rauh
M
,
Rascher
W
.
Metabolic hormones in follicular fluid in women undergoing in vitro fertilization
.
J Reprod Med
.
2013
;
58
(
7-8
):
305
311
.

443.

Vidal
C
,
Roa
J
,
Pinilla
L
,
Pellicer
A
,
Tena-Sempere
M
.
Maternal serum ghrelin levels in early IVF pregnancies: lack of prognostic value for viable pregnancy and altered post-prandial responses
.
Hum Reprod
.
2008
;
23
(
4
):
958
963
.

444.

Arck
PC
,
Rücke
M
,
Rose
M
,
Szekeres-Bartho
J
,
Douglas
AJ
,
Pritsch
M
,
Blois
SM
,
Pincus
MK
,
Bärenstrauch
N
,
Dudenhausen
JW
,
Nakamura
K
,
Sheps
S
,
Klapp
BF
.
Early risk factors for miscarriage: a prospective cohort study in pregnant women
.
Reprod Biomed Online
.
2008
;
17
(
1
):
101
113
.

445.

Joachim
R
,
Zenclussen
AC
,
Polgar
B
,
Douglas
AJ
,
Fest
S
,
Knackstedt
M
,
Klapp
BF
,
Arck
PC
.
The progesterone derivative dydrogesterone abrogates murine stress-triggered abortion by inducing a Th2 biased local immune response
.
Steroids
.
2003
;
68
(
10-13
):
931
940
.

446.

Parker
VJ
,
Douglas
AJ
.
Stress in early pregnancy: maternal neuro-endocrine-immune responses and effects
.
J Reprod Immunol
.
2010
;
85
(
1
):
86
92
.

447.

Martin
JR
,
Lieber
SB
,
McGrath
J
,
Shanabrough
M
,
Horvath
TL
,
Taylor
HS
.
Maternal ghrelin deficiency compromises reproduction in female progeny through altered uterine developmental programming
.
Endocrinology
.
2011
;
152
(
5
):
2060
2066
.

448.

Ng
PC
,
Lee
CH
,
Lam
CW
,
Chan
IH
,
Wong
E
,
Fok
TF
.
Ghrelin in preterm and term newborns: relation to anthropometry, leptin and insulin
.
Clin Endocrinol (Oxf)
.
2005
;
63
(
2
):
217
222
.

449.

Ohkawa
N
,
Shoji
H
,
Kitamura
T
,
Suganuma
H
,
Yoshikawa
N
,
Suzuki
M
,
Lee
T
,
Hisata
K
,
Shimizu
T
.
IGF-I, leptin and active ghrelin levels in very low birth weight infants during the first 8 weeks of life
.
Acta Paediatr
.
2010
;
99
(
1
):
37
41
.

450.

Cortelazzi
D
,
Cappiello
V
,
Morpurgo
PS
,
Ronzoni
S
,
Nobile De Santis
MS
,
Cetin
I
,
Beck-Peccoz
P
,
Spada
A
.
Circulating levels of ghrelin in human fetuses
.
Eur J Endocrinol
.
2003
;
149
(
2
):
111
116
.

451.

Zhu
X
,
Cao
Y
,
Voogd
K
,
Steiner
DF
.
On the processing of proghrelin to ghrelin
.
J Biol Chem
.
2006
;
281
(
50
):
38867
38870
.

452.

O’Brien
M
,
Earley
P
,
Morrison
JJ
,
Smith
TJ
.
Ghrelin in the human myometrium
.
Reprod Biol Endocrinol
.
2010
;
8
:
55
.

453.

Hehir
MP
,
Glavey
SV
,
Morrison
JJ
.
Uterorelaxant effect of ghrelin on human myometrial contractility
.
Am J Obstet Gynecol
.
2008
;
198
(
3
):
323
325
.

454.

Johnstone
HA
,
Wigger
A
,
Douglas
AJ
,
Neumann
ID
,
Landgraf
R
,
Seckl
JR
,
Russell
JA
.
Attenuation of hypothalamic-pituitary-adrenal axis stress responses in late pregnancy: changes in feedforward and feedback mechanisms
.
J Neuroendocrinol
.
2000
;
12
(
8
):
811
822
.

455.

Wigger
A
,
Lörscher
P
,
Oehler
I
,
Keck
ME
,
Naruo
T
,
Neumann
ID
.
Nonresponsiveness of the rat hypothalamo-pituitary-adrenocortical axis to parturition-related events: inhibitory action of endogenous opioids
.
Endocrinology
.
1999
;
140
(
6
):
2843
2849
.

456.

Larson-Meyer
DE
,
Ravussin
E
,
Heilbronn
L
,
DeJonge
L
.
Ghrelin and peptide YY in postpartum lactating and nonlactating women
.
Am J Clin Nutr
.
2010
;
91
(
2
):
366
372
.

457.

Ilcol
YO
,
Hizli
B
.
Active and total ghrelin concentrations increase in breast milk during lactation
.
Acta Paediatr
.
2007
;
96
(
11
):
1632
1639
.

458.

Dündar
NO
,
Dündar
B
,
Cesur
G
,
Yilmaz
N
,
Sütçu
R
,
Ozgüner
F
.
Ghrelin and adiponectin levels in colostrum, cord blood and maternal serum
.
Pediatr Int
.
2010
;
52
(
4
):
622
625
.

459.

Kierson
JA
,
Dimatteo
DM
,
Locke
RG
,
Mackley
AB
,
Spear
ML
.
Ghrelin and cholecystokinin in term and preterm human breast milk
.
Acta Paediatr
.
2006
;
95
(
8
):
991
995
.

460.

Cesur
G
,
Ozguner
F
,
Yilmaz
N
,
Dundar
B
.
The relationship between ghrelin and adiponectin levels in breast milk and infant serum and growth of infants during early postnatal life
.
J Physiol Sci
.
2012
;
62
(
3
):
185
190
.

461.

Aydin
S
,
Aydin
S
,
Ozkan
Y
,
Kumru
S
.
Ghrelin is present in human colostrum, transitional and mature milk
.
Peptides
.
2006
;
27
(
4
):
878
882
.

462.

Sullivan
EC
,
Hinde
K
,
Mendoza
SP
,
Capitanio
JP
.
Cortisol concentrations in the milk of rhesus monkey mothers are associated with confident temperament in sons, but not daughters
.
Dev Psychobiol
.
2011
;
53
(
1
):
96
104
.

463.

Grey
KR
,
Davis
EP
,
Sandman
CA
,
Glynn
LM
.
Human milk cortisol is associated with infant temperament
.
Psychoneuroendocrinology
.
2013
;
38
(
7
):
1178
1185
.

464.

Glynn
LM
,
Davis
EP
,
Schetter
CD
,
Chicz-Demet
A
,
Hobel
CJ
,
Sandman
CA
.
Postnatal maternal cortisol levels predict temperament in healthy breastfed infants
.
Early Hum Dev
.
2007
;
83
(
10
):
675
681
.

465.

Steculorum
SM
,
Collden
G
,
Coupe
B
,
Croizier
S
,
Lockie
S
,
Andrews
ZB
,
Jarosch
F
,
Klussmann
S
,
Bouret
SG
.
Neonatal ghrelin programs development of hypothalamic feeding circuits
.
J Clin Invest
.
2015
;
125
(
2
):
846
858
.

466.

Collden
G
,
Balland
E
,
Parkash
J
,
Caron
E
,
Langlet
F
,
Prevot
V
,
Bouret
SG
.
Neonatal overnutrition causes early alterations in the central response to peripheral ghrelin
.
Mol Metab
.
2014
;
4
(
1
):
15
24
.

467.

Sominsky
L
,
Ziko
I
,
Nguyen
TX
,
Andrews
ZB
,
Spencer
SJ
.
Early life disruption to the ghrelin system with over-eating is resolved in adulthood in male rats
.
Neuropharmacology
.
2016
;
113
(
Pt A
):
21
30
.

468.

Kodomari
I
,
Maruoka
T
,
Yamauchi
R
,
Wada
E
,
Wada
K
.
Ghrelin alters postnatal endocrine secretion and behavior in mouse offspring
.
Neurochem Int
.
2009
;
54
(
3-4
):
222
228
.

469.

Angelidis
G
,
Dafopoulos
K
,
Messini
CI
,
Valotassiou
V
,
Georgoulias
P
,
Messinis
IE
.
Ghrelin: new insights into female reproductive system-associated disorders and pregnancy
.
Reprod Sci
.
2012
;
19
(
9
):
903
910
.

470.

Repaci
A
,
Gambineri
A
,
Pagotto
U
,
Pasquali
R
.
Ghrelin and reproductive disorders
.
Mol Cell Endocrinol
.
2011
;
340
(
1
):
70
79
.

471.

Pacifico
L
,
Anania
C
,
Poggiogalle
E
,
Osborn
JF
,
Prossomariti
G
,
Martino
F
,
Chiesa
C
.
Relationships of acylated and des-acyl ghrelin levels to bone mineralization in obese children and adolescents
.
Bone
.
2009
;
45
(
2
):
274
279
.

472.

Briggs
DI
,
Enriori
PJ
,
Lemus
MB
,
Cowley
MA
,
Andrews
ZB
.
Diet-induced obesity causes ghrelin resistance in arcuate NPY/AgRP neurons
.
Endocrinology
.
2010
;
151
(
10
):
4745
4755
.

473.

Briggs
DI
,
Lockie
SH
,
Benzler
J
,
Wu
Q
,
Stark
R
,
Reichenbach
A
,
Hoy
AJ
,
Lemus
MB
,
Coleman
HA
,
Parkington
HC
,
Tups
A
,
Andrews
ZB
.
Evidence that diet-induced hyperleptinemia, but not hypothalamic gliosis, causes ghrelin resistance in NPY/AgRP neurons of male mice
.
Endocrinology
.
2014
;
155
(
7
):
2411
2422
.

474.

Briggs
DI
,
Lockie
SH
,
Wu
Q
,
Lemus
MB
,
Stark
R
,
Andrews
ZB
.
Calorie-restricted weight loss reverses high-fat diet-induced ghrelin resistance, which contributes to rebound weight gain in a ghrelin-dependent manner
.
Endocrinology
.
2013
;
154
(
2
):
709
717
.

475.

Elsenbruch
S
,
Benson
S
,
Rücke
M
,
Rose
M
,
Dudenhausen
J
,
Pincus-Knackstedt
MK
,
Klapp
BF
,
Arck
PC
.
Social support during pregnancy: effects on maternal depressive symptoms, smoking and pregnancy outcome
.
Hum Reprod
.
2007
;
22
(
3
):
869
877
.

476.

Latendresse
G
.
The interaction between chronic stress and pregnancy: preterm birth from a biobehavioral perspective
.
J Midwifery Womens Health
.
2009
;
54
(
1
):
8
17
.

477.

Hoffman
S
,
Hatch
MC
.
Depressive symptomatology during pregnancy: evidence for an association with decreased fetal growth in pregnancies of lower social class women
.
Health Psychol
.
2000
;
19
(
6
):
535
543
.

478.

Orr
ST
,
James
SA
,
Blackmore Prince
C
.
Maternal prenatal depressive symptoms and spontaneous preterm births among African-American women in Baltimore, Maryland
.
Am J Epidemiol
.
2002
;
156
(
9
):
797
802
.

479.

Orr
ST
,
Reiter
JP
,
Blazer
DG
,
James
SA
.
Maternal prenatal pregnancy-related anxiety and spontaneous preterm birth in Baltimore, Maryland
.
Psychosom Med
.
2007
;
69
(
6
):
566
570
.

480.

Vianna
P
,
Bauer
ME
,
Dornfeld
D
,
Chies
JA
.
Distress conditions during pregnancy may lead to pre-eclampsia by increasing cortisol levels and altering lymphocyte sensitivity to glucocorticoids
.
Med Hypotheses
.
2011
;
77
(
2
):
188
191
.

481.

Bock
J
,
Wainstock
T
,
Braun
K
,
Segal
M
.
Stress in utero: prenatal programming of brain plasticity and cognition
.
Biol Psychiatry
.
2015
;
78
(
5
):
315
326
.

482.

Bale
TL
,
Baram
TZ
,
Brown
AS
,
Goldstein
JM
,
Insel
TR
,
McCarthy
MM
,
Nemeroff
CB
,
Reyes
TM
,
Simerly
RB
,
Susser
ES
,
Nestler
EJ
.
Early life programming and neurodevelopmental disorders
.
Biol Psychiatry
.
2010
;
68
(
4
):
314
319
.

483.

Entringer
S
,
Wadhwa
PD
.
Developmental programming of obesity and metabolic dysfunction: role of prenatal stress and stress biology
.
Nestle Nutr Inst Workshop Ser
.
2013
;
74
:
107
120
.

484.

De Vriendt
T
,
Moreno
LA
,
De Henauw
S
.
Chronic stress and obesity in adolescents: scientific evidence and methodological issues for epidemiological research
.
Nutr Metab Cardiovasc Dis
.
2009
;
19
(
7
):
511
519
.

485.

Ananth
CV
,
Keyes
KM
,
Wapner
RJ
.
Pre-eclampsia rates in the United States, 1980-2010: age-period-cohort analysis
.
BMJ
.
2013
;
347
:
f6564
.

486.

Hernández-Díaz
S
,
Toh
S
,
Cnattingius
S
.
Risk of pre-eclampsia in first and subsequent pregnancies: prospective cohort study
.
BMJ
.
2009
;
338
:
b2255
.

487.

Sibai
BM
.
Preeclampsia as a cause of preterm and late preterm (near-term) births
.
Semin Perinatol
.
2006
;
30
(
1
):
16
19
.

488.

Ananth
CV
,
Basso
O
.
Impact of pregnancy-induced hypertension on stillbirth and neonatal mortality
.
Epidemiology
.
2010
;
21
(
1
):
118
123
.

489.

Ness
RB
,
Sibai
BM
.
Shared and disparate components of the pathophysiologies of fetal growth restriction and preeclampsia
.
Am J Obstet Gynecol
.
2006
;
195
(
1
):
40
49
.

490.

Bodnar
LM
,
Ness
RB
,
Markovic
N
,
Roberts
JM
.
The risk of preeclampsia rises with increasing prepregnancy body mass index
.
Ann Epidemiol
.
2005
;
15
(
7
):
475
482
.

491.

Taylor
BD
,
Ness
RB
,
Olsen
J
,
Hougaard
DM
,
Skogstrand
K
,
Roberts
JM
,
Haggerty
CL
.
Serum leptin measured in early pregnancy is higher in women with preeclampsia compared with normotensive pregnant women
.
Hypertension
.
2015
;
65
(
3
):
594
599
.

492.

Acromite
M
,
Ziotopoulou
M
,
Orlova
C
,
Mantzoros
C
.
Increased leptin levels in preeclampsia: associations with BMI, estrogen and SHBG levels
.
Hormones (Athens)
.
2004
;
3
(
1
):
46
52
.

493.

Aydin
S
,
Guzel
SP
,
Kumru
S
,
Aydin
S
,
Akin
O
,
Kavak
E
,
Sahin
I
,
Bozkurt
M
,
Halifeoglu
I
.
Serum leptin and ghrelin concentrations of maternal serum, arterial and venous cord blood in healthy and preeclamptic pregnant women
.
J Physiol Biochem
.
2008
;
64
(
1
):
51
59
.

494.

Wu
W
,
Fan
X
,
Yu
Y
,
Wang
Y
.
Maternal serum ratio of ghrelin to obestatin decreased in preeclampsia
.
Pregnancy Hypertens
.
2015
;
5
(
4
):
263
266
.

495.

Turgut
A
,
Ozler
A
,
Goruk
NY
,
Tunç
SY
,
Sak
ME
,
Evsen
MS
,
Evliyaoglu
O
,
Gul
T
.
Serum levels of the adipokines, free fatty acids, and oxidative stress markers in obese and non-obese preeclamptic patients
.
Clin Exp Obstet Gynecol
.
2015
;
42
(
4
):
473
479
.

496.

Kishimoto
I
,
Tokudome
T
,
Hosoda
H
,
Miyazato
M
,
Kangawa
K
.
Ghrelin and cardiovascular diseases
.
J Cardiol
.
2012
;
59
(
1
):
8
13
.

497.

Mao
Y
,
Tokudome
T
,
Kishimoto
I
.
Ghrelin and blood pressure regulation
.
Curr Hypertens Rep
.
2016
;
18
(
2
):
15
.

498.

Makino
Y
,
Hosoda
H
,
Shibata
K
,
Makino
I
,
Kojima
M
,
Kangawa
K
,
Kawarabayashi
T
.
Alteration of plasma ghrelin levels associated with the blood pressure in pregnancy
.
Hypertension
.
2002
;
39
(
3
):
781
784
.

499.

McGovern-Gooch
KR
,
Rodrigues
T
,
Darling
JE
,
Sieburg
MA
,
Abizaid
A
,
Hougland
JL
.
Ghrelin octanoylation is completely stabilized in biological samples by alkyl fluorophosphonates
.
Endocrinology
.
2016
;
157
(
11
):
4330
4338
.

500.

Zhang
S
,
Ding
Z
,
Liu
H
,
Chen
Z
,
Wu
J
,
Zhang
Y
,
Yu
Y
.
Association between mental stress and gestational hypertension/preeclampsia: a meta-analysis
.
Obstet Gynecol Surv
.
2013
;
68
(
12
):
825
834
.

501.

László
KD
,
Liu
XQ
,
Svensson
T
,
Wikström
AK
,
Li
J
,
Olsen
J
,
Obel
C
,
Vestergaard
M
,
Cnattingius
S
.
Psychosocial stress related to the loss of a close relative the year before or during pregnancy and risk of preeclampsia
.
Hypertension
.
2013
;
62
(
1
):
183
189
.

502.

Kurki
T
,
Hiilesmaa
V
,
Raitasalo
R
,
Mattila
H
,
Ylikorkala
O
.
Depression and anxiety in early pregnancy and risk for preeclampsia
.
Obstet Gynecol
.
2000
;
95
(
4
):
487
490
.

503.

Qiu
C
,
Williams
MA
,
Calderon-Margalit
R
,
Cripe
SM
,
Sorensen
TK
.
Preeclampsia risk in relation to maternal mood and anxiety disorders diagnosed before or during early pregnancy
.
Am J Hypertens
.
2009
;
22
(
4
):
397
402
.

504.

Yu
Y
,
Zhang
S
,
Wang
G
,
Hong
X
,
Mallow
EB
,
Walker
SO
,
Pearson
C
,
Heffner
L
,
Zuckerman
B
,
Wang
X
.
The combined association of psychosocial stress and chronic hypertension with preeclampsia
.
Am J Obstet Gynecol
.
2013
;
209
(
5
):
438.e1
438.e12
.

505.

Khatun
S
,
Kanayama
N
,
Belayet
HM
,
Masui
M
,
Sugimura
M
,
Kobayashi
T
,
Terao
T
.
Induction of preeclampsia like phenomena by stimulation of sympathetic nerve with cold and fasting stress
.
Eur J Obstet Gynecol Reprod Biol
.
1999
;
86
(
1
):
89
97
.

506.

Kanayama
N
,
Tsujimura
R
,
She
L
,
Maehara
K
,
Terao
T
.
Cold-induced stress stimulates the sympathetic nervous system, causing hypertension and proteinuria in rats
.
J Hypertens
.
1997
;
15
(
4
):
383
389
.

507.

Takiuti
NH
,
Kahhale
S
,
Zugaib
M
.
Stress in pregnancy: a new Wistar rat model for human preeclampsia
.
Am J Obstet Gynecol
.
2002
;
186
(
3
):
544
550
.

508.

Raso
GM
,
Bianco
G
,
Iacono
A
,
Esposito
E
,
Autore
G
,
Ferrante
MC
,
Calignano
A
,
Meli
R
.
Maternal adaptations to pregnancy in spontaneously hypertensive rats: leptin and ghrelin evaluation
.
J Endocrinol
.
2007
;
194
(
3
):
611
619
.

509.

Nonoshita
A
,
Nishi
Y
,
Takushima
S
,
Oshima
M
,
Hosoda
H
,
Kangawa
K
,
Kojima
M
,
Mifune
H
,
Tanaka
E
,
Hori
D
,
Kamura
T
.
Dynamics of placental ghrelin production and its receptor expression in a Dahl salt-sensitive rat model of intrauterine growth restriction
.
Placenta
.
2010
;
31
(
5
):
358
364
.

510.

Hunt
KJ
,
Schuller
KL
.
The increasing prevalence of diabetes in pregnancy
.
Obstet Gynecol Clin North Am
.
2007
;
34
(
2
):
173
199, vii
(vii.)
.

511.

Ferrara
A
.
Increasing prevalence of gestational diabetes mellitus: a public health perspective
.
Diabetes Care
.
2007
;
30
(
Suppl 2
):
S141
S146
.

512.

Kim
C
,
Newton
KM
,
Knopp
RH
.
Gestational diabetes and the incidence of type 2 diabetes: a systematic review
.
Diabetes Care
.
2002
;
25
(
10
):
1862
1868
.

513.

Zhang
S
,
Folsom
AR
,
Flack
JM
,
Liu
K
.
Body fat distribution before pregnancy and gestational diabetes: findings from coronary artery risk development in young adults (CARDIA) study
.
BMJ
.
1995
;
311
(
7013
):
1139
1140
.

514.

Chu
SY
,
Callaghan
WM
,
Kim
SY
,
Schmid
CH
,
Lau
J
,
England
LJ
,
Dietz
PM
.
Maternal obesity and risk of gestational diabetes mellitus
.
Diabetes Care
.
2007
;
30
(
8
):
2070
2076
.

515.

Schwartz
R
,
Gruppuso
PA
,
Petzold
K
,
Brambilla
D
,
Hiilesmaa
V
,
Teramo
KA
.
Hyperinsulinemia and macrosomia in the fetus of the diabetic mother
.
Diabetes Care
.
1994
;
17
(
7
):
640
648
.

516.

Silverman
BL
,
Metzger
BE
,
Cho
NH
,
Loeb
CA
.
Impaired glucose tolerance in adolescent offspring of diabetic mothers. Relationship to fetal hyperinsulinism
.
Diabetes Care
.
1995
;
18
(
5
):
611
617
.

517.

Daniells
S
,
Grenyer
BF
,
Davis
WS
,
Coleman
KJ
,
Burgess
JA
,
Moses
RG
.
Gestational diabetes mellitus: is a diagnosis associated with an increase in maternal anxiety and stress in the short and intermediate term?
Diabetes Care
.
2003
;
26
(
2
):
385
389
.

518.

Lydon
K
,
Dunne
FP
,
Owens
L
,
Avalos
G
,
Sarma
KM
,
O’Connor
C
,
Nestor
L
,
McGuire
BE
.
Psychological stress associated with diabetes during pregnancy: a pilot study
.
Ir Med J
.
2012
;
105
(
5
,
Suppl
)
26
28
.

519.

Pearson
S
,
Schmidt
M
,
Patton
G
,
Dwyer
T
,
Blizzard
L
,
Otahal
P
,
Venn
A
.
Depression and insulin resistance: cross-sectional associations in young adults
.
Diabetes Care
.
2010
;
33
(
5
):
1128
1133
.

520.

Kan
C
,
Silva
N
,
Golden
SH
,
Rajala
U
,
Timonen
M
,
Stahl
D
,
Ismail
K
.
A systematic review and meta-analysis of the association between depression and insulin resistance [published correction appears in Diabetes Care. 2013;36(5):1429]
.
Diabetes Care
.
2013
;
36
(
2
):
480
489
.

521.

Sepa
A
,
Wahlberg
J
,
Vaarala
O
,
Frodi
A
,
Ludvigsson
J
.
Psychological stress may induce diabetes-related autoimmunity in infancy
.
Diabetes Care
.
2005
;
28
(
2
):
290
295
.

522.

Entringer
S
,
Wust
S
,
Kumsta
R
,
Layes
IM
,
Nelson
EL
,
Hellhammer
DH
,
Wadhwa
PD
.
Prenatal psychosocial stress exposure is associated with insulin resistance in young adults
.
Am J Obstet Gynecol
.
2008
;
199
(
5
):
498.e1
498.e7
.

523.

Coussons-Read
ME
,
Okun
ML
,
Nettles
CD
.
Psychosocial stress increases inflammatory markers and alters cytokine production across pregnancy
.
Brain Behav Immun
.
2007
;
21
(
3
):
343
350
.

524.

Pantham
P
,
Aye
IL
,
Powell
TL
.
Inflammation in maternal obesity and gestational diabetes mellitus
.
Placenta
.
2015
;
36
(
7
):
709
715
.

525.

Richardson
AC
,
Carpenter
MW
.
Inflammatory mediators in gestational diabetes mellitus
.
Obstet Gynecol Clin North Am
.
2007
;
34
(
2
):
213
224, viii
(viii.)
.

526.

Gibson
W
,
Liu
J
,
Gaylinn
B
,
Thorner
MO
,
Meneilly
GS
,
Babich
SL
,
Thompson
D
,
Chanoine
JP
.
Effects of glucose and insulin on acyl ghrelin and desacyl ghrelin, leptin, and adiponectin in pregnant women with diabetes
.
Metabolism
.
2010
;
59
(
6
):
841
847
.

527.

Riedl
M
,
Maier
C
,
Handisurya
A
,
Luger
A
,
Kautzky-Willer
A
.
Insulin resistance has no impact on ghrelin suppression in pregnancy
.
J Intern Med
.
2007
;
262
(
4
):
458
465
.

528.

Telejko
B
,
Kuzmicki
M
,
Zonenberg
A
,
Modzelewska
A
,
Niedziolko-Bagniuk
K
,
Ponurkiewicz
A
,
Wawrusiewicz-Kurylonek
N
,
Nikolajuk
A
,
Szamatowicz
J
,
Laudanski
P
,
Kretowski
A
,
Gorska
M
.
Ghrelin in gestational diabetes: serum level and mRNA expression in fat and placental tissue
.
Exp Clin Endocrinol Diabetes
.
2010
;
118
(
2
):
87
92
.

529.

Lappas
M
,
Jinks
D
,
Ugoni
A
,
Louizos
CC
,
Permezel
M
,
Georgiou
HM
.
Post-partum plasma C-peptide and ghrelin concentrations are predictive of type 2 diabetes in women with previous gestational diabetes mellitus
.
J Diabetes
.
2015
;
7
(
4
):
506
511
.

530.

Ehrmann
DA
.
Polycystic ovary syndrome
.
N Engl J Med
.
2005
;
352
(
12
):
1223
1236
.

531.

Dunaif
A
.
Insulin resistance and the polycystic ovary syndrome: mechanism and implications for pathogenesis
.
Endocr Rev
.
1997
;
18
(
6
):
774
800
.

532.

Garcia-Rudaz
C
,
Armando
I
,
Levin
G
,
Escobar
ME
,
Barontini
M
.
Peripheral catecholamine alterations in adolescents with polycystic ovary syndrome
.
Clin Endocrinol (Oxf)
.
1998
;
49
(
2
):
221
228
.

533.

Teede
H
,
Deeks
A
,
Moran
L
.
Polycystic ovary syndrome: a complex condition with psychological, reproductive and metabolic manifestations that impacts on health across the lifespan
.
BMC Med
.
2010
;
8
:
41
.

534.

Tsilchorozidou
T
,
Honour
JW
,
Conway
GS
.
Altered cortisol metabolism in polycystic ovary syndrome: insulin enhances 5alpha-reduction but not the elevated adrenal steroid production rates
.
J Clin Endocrinol Metab
.
2003
;
88
(
12
):
5907
5913
.

535.

Fassnacht
M
,
Schlenz
N
,
Schneider
SB
,
Wudy
SA
,
Allolio
B
,
Arlt
W
.
Beyond adrenal and ovarian androgen generation: increased peripheral 5 alpha-reductase activity in women with polycystic ovary syndrome
.
J Clin Endocrinol Metab
.
2003
;
88
(
6
):
2760
2766
.

536.

Vassiliadi
DA
,
Barber
TM
,
Hughes
BA
,
McCarthy
MI
,
Wass
JA
,
Franks
S
,
Nightingale
P
,
Tomlinson
JW
,
Arlt
W
,
Stewart
PM
.
Increased 5 alpha-reductase activity and adrenocortical drive in women with polycystic ovary syndrome
.
J Clin Endocrinol Metab
.
2009
;
94
(
9
):
3558
3566
.

537.

Ikezaki
A
,
Hosoda
H
,
Ito
K
,
Iwama
S
,
Miura
N
,
Matsuoka
H
,
Kondo
C
,
Kojima
M
,
Kangawa
K
,
Sugihara
S
.
Fasting plasma ghrelin levels are negatively correlated with insulin resistance and PAI-1, but not with leptin, in obese children and adolescents
.
Diabetes
.
2002
;
51
(
12
):
3408
3411
.

538.

Moran
LJ
,
Noakes
M
,
Clifton
PM
,
Wittert
GA
,
Tomlinson
L
,
Galletly
C
,
Luscombe
ND
,
Norman
RJ
.
Ghrelin and measures of satiety are altered in polycystic ovary syndrome but not differentially affected by diet composition
.
J Clin Endocrinol Metab
.
2004
;
89
(
7
):
3337
3344
.

539.

Barber
TM
,
Casanueva
FF
,
Karpe
F
,
Lage
M
,
Franks
S
,
McCarthy
MI
,
Wass
JA
.
Ghrelin levels are suppressed and show a blunted response to oral glucose in women with polycystic ovary syndrome
.
Eur J Endocrinol
.
2008
;
158
(
4
):
511
516
.

540.

Micic
D
,
Sumarac-Dumanovic
M
,
Kendereski
A
,
Cvijovic
G
,
Zoric
S
,
Pejkovic
D
,
Micic
J
,
Milic
N
,
Dieguez
C
,
Casanueva
FF
.
Total ghrelin levels during acute insulin infusion in patients with polycystic ovary syndrome
.
J Endocrinol Invest
.
2007
;
30
(
10
):
820
827
.

541.

Panidis
D
,
Farmakiotis
D
,
Koliakos
G
,
Rousso
D
,
Kourtis
A
,
Katsikis
I
,
Asteriadis
C
,
Karayannis
V
,
Diamanti-Kandarakis
E
.
Comparative study of plasma ghrelin levels in women with polycystic ovary syndrome, in hyperandrogenic women and in normal controls
.
Hum Reprod
.
2005
;
20
(
8
):
2127
2132
.

542.

Altuğ Şen
T
,
Köken
R
,
Narcı
A
,
Yılmazer
M
.
Homocysteine and ghrelin link with polcystic ovary syndrome in relation to obesity
.
J Pediatr Adolesc Gynecol
.
2011
;
24
(
4
):
211
217
.

543.

Bideci
A
,
Camurdan
MO
,
Yeşilkaya
E
,
Demirel
F
,
Cinaz
P
.
Serum ghrelin, leptin and resistin levels in adolescent girls with polycystic ovary syndrome
.
J Obstet Gynaecol Res
.
2008
;
34
(
4
):
578
584
.

544.

Glintborg
D
,
Andersen
M
,
Hagen
C
,
Frystyk
J
,
Hulstrøm
V
,
Flyvbjerg
A
,
Hermann
AP
.
Evaluation of metabolic risk markers in polycystic ovary syndrome (PCOS). Adiponectin, ghrelin, leptin and body composition in hirsute PCOS patients and controls
.
Eur J Endocrinol
.
2006
;
155
(
2
):
337
345
.

545.

Pagotto
U
,
Gambineri
A
,
Vicennati
V
,
Heiman
ML
,
Tschöp
M
,
Pasquali
R
.
Plasma ghrelin, obesity, and the polycystic ovary syndrome: correlation with insulin resistance and androgen levels
.
J Clin Endocrinol Metab
.
2002
;
87
(
12
):
5625
5629
.

546.

Orio
F
, Jr ,
Lucidi
P
,
Palomba
S
,
Tauchmanova
L
,
Cascella
T
,
Russo
T
,
Zullo
F
,
Colao
A
,
Lombardi
G
,
De Feo
P
.
Circulating ghrelin concentrations in the polycystic ovary syndrome
.
J Clin Endocrinol Metab
.
2003
;
88
(
2
):
942
945
.

547.

Houjeghani
S
,
Pourghassem Gargari
B
,
Farzadi
L
.
Serum leptin and ghrelin levels in women with polycystic ovary syndrome: correlation with anthropometric, metabolic, and endocrine parameters
.
Int J Fertil Steril
.
2012
;
6
(
2
):
117
126
.

548.

Arusoglu
G
,
Koksal
G
,
Cinar
N
,
Tapan
S
,
Aksoy
DY
,
Yildiz
BO
.
Basal and meal-stimulated ghrelin, PYY, CCK levels and satiety in lean women with polycystic ovary syndrome: effect of low-dose oral contraceptive
.
J Clin Endocrinol Metab
.
2013
;
98
(
11
):
4475
4482
.

549.

Schöfl
C
,
Horn
R
,
Schill
T
,
Schlösser
HW
,
Müller
MJ
,
Brabant
G
.
Circulating ghrelin levels in patients with polycystic ovary syndrome
.
J Clin Endocrinol Metab
.
2002
;
87
(
10
):
4607
4610
.

550.

McLaughlin
T
,
Abbasi
F
,
Lamendola
C
,
Frayo
RS
,
Cummings
DE
.
Plasma ghrelin concentrations are decreased in insulin-resistant obese adults relative to equally obese insulin-sensitive controls
.
J Clin Endocrinol Metab
.
2004
;
89
(
4
):
1630
1635
.

551.

Mitkov
M
,
Pehlivanov
B
,
Orbetzova
M
.
Serum ghrelin level in women with polycystic ovary syndrome and its relationship with endocrine and metabolic parameters
.
Gynecol Endocrinol
.
2008
;
24
(
11
):
625
630
.

552.

Gambineri
A
,
Pagotto
U
,
Tschöp
M
,
Vicennati
V
,
Manicardi
E
,
Carcello
A
,
Cacciari
M
,
De Iasio
R
,
Pasquali
R
.
Anti-androgen treatment increases circulating ghrelin levels in obese women with polycystic ovary syndrome
.
J Endocrinol Invest
.
2003
;
26
(
7
):
629
634
.

553.

Sağsöz
N
,
Orbak
Z
,
Noyan
V
,
Yücel
A
,
Uçar
B
,
Yildiz
L
.
The effects of oral contraceptives including low-dose estrogen and drospirenone on the concentration of leptin and ghrelin in polycystic ovary syndrome
.
Fertil Steril
.
2009
;
92
(
2
):
660
666
.

554.

Yen
SS
,
Vela
P
,
Rankin
J
.
Inappropriate secretion of follicle-stimulating hormone and luteinizing hormone in polycystic ovarian disease
.
J Clin Endocrinol Metab
.
1970
;
30
(
4
):
435
442
.

555.

Komarowska
H
,
Stangierski
A
,
Warmuz-Stangierska
I
,
Lodyga
M
,
Ochmanska
K
,
Wasko
R
,
Wanic-Kossowska
M
,
Ruchala
M
.
Differences in the psychological and hormonal presentation of lean and obese patients with polycystic ovary syndrome
.
Neuroendocrinol Lett
.
2013
;
34
(
7
):
669
674
.

556.

Waśko
R
,
Komarowska
H
,
Warenik-Szymankiewicz
A
,
Sowiński
J
.
Elevated ghrelin plasma levels in patients with polycystic ovary syndrome
.
Horm Metab Res
.
2004
;
36
(
3
):
170
173
.

557.

Mircea
CN
,
Lujan
ME
,
Pierson
RA
.
Metabolic fuel and clinical implications for female reproduction
.
J Obstet Gynaecol Can
.
2007
;
29
(
11
):
887
902
.

558.

Soriano-Guillén
L
,
Barrios
V
,
Campos-Barros
A
,
Argente
J
.
Ghrelin levels in obesity and anorexia nervosa: effect of weight reduction or recuperation
.
J Pediatr
.
2004
;
144
(
1
):
36
42
.

559.

Korek
E
,
Krauss
H
,
Gibas-Dorna
M
,
Kupsz
J
,
Piątek
M
,
Piątek
J
.
Fasting and postprandial levels of ghrelin, leptin and insulin in lean, obese and anorexic subjects
.
Prz Gastroenterol
.
2013
;
8
(
6
):
383
389
.

560.

Poyastro Pinheiro
A
,
Thornton
LM
,
Plotonicov
KH
,
Tozzi
F
,
Klump
KL
,
Berrettini
WH
,
Brandt
H
,
Crawford
S
,
Crow
S
,
Fichter
MM
,
Goldman
D
,
Halmi
KA
,
Johnson
C
,
Kaplan
AS
,
Keel
P
,
LaVia
M
,
Mitchell
J
,
Rotondo
A
,
Strober
M
,
Treasure
J
,
Woodside
DB
,
Von Holle
A
,
Hamer
R
,
Kaye
WH
,
Bulik
CM
.
Patterns of menstrual disturbance in eating disorders
.
Int J Eat Disord
.
2007
;
40
(
5
):
424
434
.

561.

Hoffman
ER
,
Zerwas
SC
,
Bulik
CM
.
Reproductive issues in anorexia nervosa
.
Expert Rev Obstet Gynecol
.
2011
;
6
(
4
):
403
414
.

562.

Castellano
JM
,
Navarro
VM
,
Fernández-Fernández
R
,
Nogueiras
R
,
Tovar
S
,
Roa
J
,
Vazquez
MJ
,
Vigo
E
,
Casanueva
FF
,
Aguilar
E
,
Pinilla
L
,
Dieguez
C
,
Tena-Sempere
M
.
Changes in hypothalamic KiSS-1 system and restoration of pubertal activation of the reproductive axis by kisspeptin in undernutrition
.
Endocrinology
.
2005
;
146
(
9
):
3917
3925
.

563.

Aiken
CE
,
Tarry-Adkins
JL
,
Ozanne
SE
.
Transgenerational developmental programming of ovarian reserve
.
Sci Rep
.
2015
;
5
:
16175
.

564.

Chan
KA
,
Bernal
AB
,
Vickers
MH
,
Gohir
W
,
Petrik
JJ
,
Sloboda
DM
.
Early life exposure to undernutrition induces ER stress, apoptosis, and reduced vascularization in ovaries of adult rat offspring
.
Biol Reprod
.
2015
;
92
(
4
):
110
.

565.

Tolle
V
,
Kadem
M
,
Bluet-Pajot
MT
,
Frere
D
,
Foulon
C
,
Bossu
C
,
Dardennes
R
,
Mounier
C
,
Zizzari
P
,
Lang
F
,
Epelbaum
J
,
Estour
B
.
Balance in ghrelin and leptin plasma levels in anorexia nervosa patients and constitutionally thin women
.
J Clin Endocrinol Metab
.
2003
;
88
(
1
):
109
116
.

566.

Otto
B
,
Cuntz
U
,
Fruehauf
E
,
Wawarta
R
,
Folwaczny
C
,
Riepl
RL
,
Heiman
ML
,
Lehnert
P
,
Fichter
M
,
Tschöp
M
.
Weight gain decreases elevated plasma ghrelin concentrations of patients with anorexia nervosa
.
Eur J Endocrinol
.
2001
;
145
(
5
):
669
673
.

567.

Tanaka
M
,
Naruo
T
,
Yasuhara
D
,
Tatebe
Y
,
Nagai
N
,
Shiiya
T
,
Nakazato
M
,
Matsukura
S
,
Nozoe
S
.
Fasting plasma ghrelin levels in subtypes of anorexia nervosa
.
Psychoneuroendocrinology
.
2003
;
28
(
7
):
829
835
.

568.

Kawakami
A
,
Okada
N
,
Rokkaku
K
,
Honda
K
,
Ishibashi
S
,
Onaka
T
.
Leptin inhibits and ghrelin augments hypothalamic noradrenaline release after stress
.
Stress
.
2008
;
11
(
5
):
363
369
.

569.

Chuang
JC
,
Zigman
JM
.
Ghrelin’s roles in stress, mood, and anxiety regulation
.
Int J Pept
.
2010
;
2010
.

570.

Currie
PJ
,
John
CS
,
Nicholson
ML
,
Chapman
CD
,
Loera
KE
.
Hypothalamic paraventricular 5-hydroxytryptamine inhibits the effects of ghrelin on eating and energy substrate utilization
.
Pharmacol Biochem Behav
.
2010
;
97
(
1
):
152
155
.

571.

Currie
PJ
,
Khelemsky
R
,
Rigsbee
EM
,
Dono
LM
,
Coiro
CD
,
Chapman
CD
,
Hinchcliff
K
.
Ghrelin is an orexigenic peptide and elicits anxiety-like behaviors following administration into discrete regions of the hypothalamus
.
Behav Brain Res
.
2012
;
226
(
1
):
96
105
.

572.

Chen
HY
,
Trumbauer
ME
,
Chen
AS
,
Weingarth
DT
,
Adams
JR
,
Frazier
EG
,
Shen
Z
,
Marsh
DJ
,
Feighner
SD
,
Guan
XM
,
Ye
Z
,
Nargund
RP
,
Smith
RG
,
Van der Ploeg
LH
,
Howard
AD
,
MacNeil
DJ
,
Qian
S
.
Orexigenic action of peripheral ghrelin is mediated by neuropeptide Y and agouti-related protein
.
Endocrinology
.
2004
;
145
(
6
):
2607
2612
.

573.

Bertoldi
ML
,
Luque
EM
,
Carlini
VP
,
Vincenti
LM
,
Stutz
G
,
Santillán
ME
,
Ruiz
RD
,
Fiol de Cuneo
M
,
Martini
AC
.
Inhibitory effects of ghrelin on sexual behavior: role of the peptide in the receptivity reduction induced by food restriction in mice
.
Horm Metab Res
.
2011
;
43
(
7
):
494
499
.

574.

Delhanty
PJ
,
Huisman
M
,
Baldeon-Rojas
LY
,
van den Berge
I
,
Grefhorst
A
,
Abribat
T
,
Leenen
PJ
,
Themmen
AP
,
van der Lely
AJ
.
Des-acyl ghrelin analogs prevent high-fat-diet-induced dysregulation of glucose homeostasis
.
FASEB J
.
2013
;
27
(
4
):
1690
1700
.

575.

Delhanty
PJ
,
Neggers
SJ
,
van der Lely
AJ
.
Mechanisms in endocrinology: ghrelin: the differences between acyl- and des-acyl ghrelin
.
Eur J Endocrinol
.
2012
;
167
(
5
):
601
608
.

576.

Delhanty
PJ
,
Sun
Y
,
Visser
JA
,
van Kerkwijk
A
,
Huisman
M
,
van Ijcken
WF
,
Swagemakers
S
,
Smith
RG
,
Themmen
AP
,
van der Lely
AJ
.
Unacylated ghrelin rapidly modulates lipogenic and insulin signaling pathway gene expression in metabolically active tissues of GHSR deleted mice
.
PLoS One
.
2010
;
5
(
7
):
e11749
.

577.

Özcan
B
,
Neggers
SJ
,
Miller
AR
,
Yang
HC
,
Lucaites
V
,
Abribat
T
,
Allas
S
,
Huisman
M
,
Visser
JA
,
Themmen
AP
,
Sijbrands
EJ
,
Delhanty
PJ
,
van der Lely
AJ
.
Does des-acyl ghrelin improve glycemic control in obese diabetic subjects by decreasing acylated ghrelin levels?
Eur J Endocrinol
.
2014
;
170
(
6
):
799
807
.

578.

Kenny
R
,
Cai
G
,
Bayliss
JA
,
Clarke
M
,
Choo
YL
,
Miller
AA
,
Andrews
ZB
,
Spencer
SJ
.
Endogenous ghrelin’s role in hippocampal neuroprotection after global cerebral ischemia: does endogenous ghrelin protect against global stroke?
Am J Physiol Regul Integr Comp Physiol
.
2013
;
304
(
11
):
R980
R990
.

579.

Bayliss
JA
,
Lemus
M
,
Santos
VV
,
Deo
M
,
Elsworth
JD
,
Andrews
ZB
.
Acylated but not des-acyl ghrelin is neuroprotective in an MPTP mouse model of Parkinson’s disease
.
J Neurochem
.
2016
;
137
(
3
):
460
471
.

580.

Julien
M
,
Kay
RG
,
Delhanty
PJ
,
Allas
S
,
Granata
R
,
Barton
C
,
Constable
S
,
Ghigo
E
,
van der Lely
AJ
,
Abribat
T
.
In vitro and in vivo stability and pharmacokinetic profile of unacylated ghrelin (UAG) analogues
.
Eur J Pharm Sci
.
2012
;
47
(
4
):
625
635
.

581.

Allas
S
,
Delale
T
,
Ngo
N
,
Julien
M
,
Sahakian
P
,
Ritter
J
,
Abribat
T
,
van der Lely
AJ
.
Safety, tolerability, pharmacokinetics and pharmacodynamics of AZP-531, a first-in-class analogue of unacylated ghrelin, in healthy and overweight/obese subjects and subjects with type 2 diabetes
.
Diabetes Obes Metab
.
2016
;
18
(
9
):
868
874
.