The average age for pubertal onset in girls has declined over recent decades. Epidemiological studies in humans and experimental studies in animals suggest a causal role for endocrine disrupting chemicals (EDCs) that are present in our environment. Of concern, current testing and screening regimens are inadequate in identifying EDCs that may affect pubertal maturation, not least because they do not consider early-life exposure. Also, the causal relationship between EDC exposure and pubertal timing is still a matter of debate. To address this issue, we have used current knowledge to elaborate a network of putative adverse outcome pathways (pAOPs) to identify how chemicals can affect pubertal onset. By using the AOP framework, we highlight current gaps in mechanistic understanding that need to be addressed and simultaneously point towards events causative of pubertal disturbance that could be exploited for alternative test methods. We propose 6 pAOPs that could explain the disruption of pubertal timing by interfering with the central hypothalamic trigger of puberty, GnRH neurons, and by so doing highlight specific modes of action that could be targeted for alternative test method development.

Puberty is a multifaceted process whereby a child develops into a reproductive mature individual. Central to this process is the reactivation and maturation of the hypothalamic-pituitary-gonadal (HPG) axis that leads to gonadarche. The average time of pubertal onset marked by breast development in girls is 10–11 years of age, whereas the average age of menarche is around 12–13 years. There has been a steady decline in age at pubertal onset and maturation over the last decades, attributed to both improved lifestyle [1, 2] and potential exposure to endocrine disrupting chemicals (EDCs) in the environment [2, 3].

Late-onset puberty in girls appears to be associated with a slight decrease in bone density and increase in metabolic disorders [4], whereas early puberty is associated with increased risk of breast cancer [5], metabolic disorders such as insulin resistance and obesity [6], and depression and antisocial behavior [7] later in life. Prenatal and early postnatal disturbance of sexual development may also herald late-life fertility issues or reproductive disorders [8]. This association between EDC exposure, disturbed puberty, and a range of other reproductive disorders in women has prompted increased research focused on the issue, with emerging evidence for there being clear links between early-life exposure and late-life reproductive effects. However, our mechanistic knowledge about causal relationships between chemical exposure and disease outcomes remains inadequate with regard to devising robust chemical test strategies capable of detecting compounds that may be harmful to developing girls. To better address this shortcoming, we present a collection of putative adverse outcome pathways (pAOPs) that we believe will serve as signposts for future research and development on the area. We have chosen to focus the review on female puberty.

Sexual maturation, or puberty, is initiated by a subset of hypothalamic neurons secreting gonadotropin-releasing hormone (GnRH). These neurons release GnRH in a pulsatile manner into the hypophyseal portal blood system. In the anterior pituitary, GnRH binds to its receptor expressed by gonadotropic cells and induces the release of 2 gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). In turn, LH and FSH stimulate gametogenesis and steroidogenesis. The release of sex steroids is therefore modulated by pituitary gonadotropic cells, which are themselves influenced by the frequency with which GnRH is released. Sex steroids modulate the activity of the HPG axis through positive and negative feedback loops (shown in Fig. 1) [9]. Estradiol and testosterone exert a negative feedback, which is active throughout development. Starting at puberty, estrogens also produce a positive feedback to induce ovulation in females [9].

Fig. 1.

HPG axis. Secretion of GnRH governs the HPG axis by stimulating the release of LH/FSH, which in turn will induce the release of sex steroids by the gonads. Sex steroids exert negative feedback when their serum levels increase in order to stabilize their concentrations. At puberty, they can also exert positive feedback leading to the preovulatory LH surge in females. These feedbacks act through kisspeptin neurons. The release of GnRH is under the control of a complex network including neurotransmitters (GABA and Glu), neuropeptides (kisspeptin), glial factors and peripheral hormones (leptin). EDCs can disturb this regulating system at many levels. HPG, hypothalamic-pituitary-gonadal; GnRH, gonadotropin-releasing hormone; LH, luteinizing hormone; FSH, follicle-stimulating hormone; Glu, glutamate; EDCs, endocrine disrupting chemicals; RP3V, rostral periventricular area of the third ventricle; ARC, arcuate nucleus; E2, estradiol; Kiss, kisspeptin; Dyn, dynorphin; NKB, neurokinin B; KNDy: KNDy neurons

Fig. 1.

HPG axis. Secretion of GnRH governs the HPG axis by stimulating the release of LH/FSH, which in turn will induce the release of sex steroids by the gonads. Sex steroids exert negative feedback when their serum levels increase in order to stabilize their concentrations. At puberty, they can also exert positive feedback leading to the preovulatory LH surge in females. These feedbacks act through kisspeptin neurons. The release of GnRH is under the control of a complex network including neurotransmitters (GABA and Glu), neuropeptides (kisspeptin), glial factors and peripheral hormones (leptin). EDCs can disturb this regulating system at many levels. HPG, hypothalamic-pituitary-gonadal; GnRH, gonadotropin-releasing hormone; LH, luteinizing hormone; FSH, follicle-stimulating hormone; Glu, glutamate; EDCs, endocrine disrupting chemicals; RP3V, rostral periventricular area of the third ventricle; ARC, arcuate nucleus; E2, estradiol; Kiss, kisspeptin; Dyn, dynorphin; NKB, neurokinin B; KNDy: KNDy neurons

Close modal

Puberty results from an increase in GnRH pulsatile secretion. In female rats, hypothalamic GnRH content gradually increases during the first days of life. From day 12, GnRH secretion becomes pulsatile and increases in frequency and amplitude until puberty [10-12]. This developmental increase was reproduced ex vivo using incubation of hypothalamic explants [13]. As illustrated in Figure 2, a gradual increase in GnRH secretion frequency occurs in both sexes between postnatal days 5 and 25 [13]. This activation of pulsatile GnRH secretion causes an increase in LH and FSH secretion that will stimulate gonad maturation and thereby sex steroid production and release. Classically, the increase in GnRH secretion around puberty is considered to be under the control of a variety of hypothalamic genes organized in coordinated networks. The current hypothesis postulates that a loss of trans-synaptic inhibition, together with a rise in excitatory inputs, is responsible for the activation of GnRH release [14]. This rise in excitatory inputs is associated with the arrival of axonal projections from neurons in the arcuate nucleus (ARC), such as kisspeptin neurons, in the neighborhood of GnRH cell bodies in the preoptic area [15]. This results from a shift in expression of puberty activating and inhibiting genes during pubertal transition and a marked switch in the regulation of the GnRH promoter [16].

Fig. 2.

Postnatal evolution of pulsatile GnRH secretion measured in vitro in the female rat. A gradual increase in GnRH secretion frequency occurs between postnatal days 5 and 25. Adapted from Bourguignon and Franchimont 1984 [12]. GnRH, gonadotropin-releasing hormone.

Fig. 2.

Postnatal evolution of pulsatile GnRH secretion measured in vitro in the female rat. A gradual increase in GnRH secretion frequency occurs between postnatal days 5 and 25. Adapted from Bourguignon and Franchimont 1984 [12]. GnRH, gonadotropin-releasing hormone.

Close modal

Secular decrease in age at menarche took place between 1890 and 1960 and was probably caused by improvement in nutritional and socioeconomic status [1, 17]. While age at menarche appears to have stabilized, several American and European studies have indicated an advancement of breast development in girls over the last 30 years [18-20]. In addition, more recent data suggest that age distribution of pubertal signs is skewed towards earliness for initial pubertal stages and toward lateness for final pubertal stages [3, 21]. Besides this worldwide secular trend, some specific populations appear to show a high prevalence of precocious puberty (breast development before the age of 8 in girls). In Belgium and other developed countries, migrating children have a markedly increased risk of sexual precocity [17]. Such rapid evolution of developmental landmarks led to the hypothesis that puberty timing could be affected by exposure to environmental factors.

Human populations are exposed to an increasing number of synthetic chemicals [22]. Among 85,000 chemicals in use, ∼1,000 have been identified as having the ability to disrupt normal endocrine function [23]. These chemicals, often referred to as EDCs, are defined as any exogenous chemical, or mixture of chemicals, that interferes with any aspect of hormone action and leads to adverse effects in an intact organism or its progeny [24]. EDCs are found in plastics (bisphenol A [BPA]) and plasticizers (phthalates), solvents and lubricants (polychlorinated biphenyls, polybrominated biphenyls, and dioxins), pesticides (methoxychlor, chlorpyrifos, and dichlorodiphenyltrichloroethane), and pharmaceutical agents (diethylstilbestrol [DES]). EDCs have a wide impact on populations given their ubiquitous presence in our environment. Reports about the advancing onset of puberty in several countries have led to the hypothesis that the increasing burden of EDCs could be a contributing factor.

Demonstrating the effects of EDCs on human puberty is complex due to the concomitant exposure to a large number of EDCs, as well as the long and diverse period of exposure from conception onwards. However, some studies have identified that early-life exposure to EDCs could affect pubertal timing. Daughters of women working in greenhouses exposed to pesticides showed earlier breast development than daughters of unexposed women [25]. Recently, age at puberty was shown to be lowered by in utero exposure to phytoestrogens in a British cohort [26] while the consumption of soy products during infancy is linked to early menarche in girls in the same cohort [26] and to altered menstrual patterns in young adults in an American cohort [27, 28]. Exposure to flame retardants such as PBDEs also appears to affect puberty timing in girls with effects depending on timing of exposure [29-31]. Finally, several animal models of EDC exposure have confirmed the impact of EDCs on pubertal timing, as reviewed by Parent et al. [3]. However, mechanisms of action remain incompletely understood. This hampers the development and use of test methods for chemical risk assessment, including nonanimal screening methods. Addressing this shortcoming is central to our current Horizon 2020 project FREIA, where we use the AOP framework specifically to guide future efforts by us and others to elaborate new and improved test strategies [32, 33]. In the following, we propose to clarify additional mechanisms of interest using the AOP approach.

The AOP framework, or Knowledge Base (KB), is sponsored by the Organization for Economic Co-operation and Development (OECD) and serves as a central hub for AOP resources (https://aopkb.oecd.org/index.html). Within the AOP-KB, AOP Wiki (https://aopwiki.org/) serves as the primary repository for AOPs developed as part of the OECD AOP Development Effort by the Extended Advisory Group on Molecular Screening and Toxicogenomics. These online sites provide extensive resources for those wishing to learn more or themselves contributing towards advancing toxicological risk assessment and regulation efforts. Below, we will only describe the basic components of an AOP.

An AOP is a pragmatic description of the mechanistic principles underpinning cause-effect relationships leading up to an adverse effect in an intact organism. An AOP is assembled from a set of building blocks [34] starting with a molecular initiating event (MIE; a direct interaction between a chemical and a molecular target). This initial interaction triggers subsequent key events (KEs; a measurable change in biological or physiological state necessary to progress through the AOP), finally culminating in an adverse outcome (AO; an observable change in biological or physiological state of an intact organism typically of regulatory significance). Finally, the various KEs are linked by key event relationships (KERs), which are biologically plausible and scientifically based relationships describing causal and predictive connections between events. KERs are thus essential components of any encyclopedic AOP, as they allow us to predict that an AO is likely to manifest based only on data from upstream KEs or MIEs. This is the essential principle behind the AOP concept, to facilitate the use of nonanimal test methods to predict adversity in intact organisms based on mechanistic understanding of toxic responses.

Although AOPs are similar to classical molecular pathways describing normal development or disease etiologies [35], there are clear distinctions. Firstly, an AOP should be of relevance to chemical risk assessment or regulatory decision-making. The AO should thus be of regulatory significance and describe an observable pathological or clinical effect in an intact organism that is indicative of a disease state resulting from exposure to a toxicant [36]. Notably, however, AOPs are strictly biological descriptions not defined by any specific chemical [37]. Secondly, the various KEs should be limited only to those steps that are “both measurable and essential to the progression towards a specific AO” [38]. In other words, an AOP is not an exhaustive description of all the steps taking place between an MIE and an AO, but a short-hand version focusing on data that are usable for chemical risk assessment purposes. Thirdly, the KE string should be linked by biologically plausible and predictive relationships, or KERs, to facilitate the use of alternative test method data (e.g., in silico or in vitro) when determining potential chemical toxicity to intact organisms.

It is generally recognized that a single AOP is not necessarily robust enough to capture all toxic events that could result in a relevant toxic effect, or AO. But rather than constructing highly complex AOPs in an effort to address this shortcoming, the AOP thinking is to elaborate several related AOPs that can be linked through individual KEs to form the so-called AOP networks [39]. We recently proposed 10 pAOPs for female reproductive disorders arising from early-life exposure to chemicals, where most were grouped in smaller networks, some of which themselves are connectable by additional shared KEs [33]. In this review, we expand on this AOP network by including pAOPs pertaining to disrupted pubertal timing in girls in response to EDC exposure.

In this review, we have developed pAOPs that could help explain the disruption of pubertal onset by EDC exposure and, by so doing, facilitate the development of additional test methods for chemical risk assessment purposes. All of these new pAOPs include a shared central element: modification of GnRH and LH/FSH release. As outlined in the Introduction section, pulsatile GnRH release increases to its maximum frequency at puberty when it induces a rise in LH and FSH, ultimately leading to gonadal maturation. The pubertal increase in GnRH release results from a concurrent decrease in inhibitory inputs and increase in stimulatory signals on GnRH secretion. The exact nature of this upstream regulation is still incompletely understood. However, some major hypothalamic regulators such as GABA, glutamate, and kisspeptin neurons are known to act directly on GnRH neurons and to be controlled by transcriptional and peripheral factors.

pAOPs Involving Kisspeptin Neurons

pAOP-1: “Increased Activation of ER in RP3V Leads to Early Puberty”

This pAOP proposes to link an increase in estrogen receptor (ER) activation in the rostral periventricular area of the third ventricle (RP3V), leading to an early puberty via an elevated hypothalamic kisspeptin level (shown in Fig. 3).

Fig. 3.

pAOP network for disrupted puberty onset involving kisspeptin neurons. Pulsatile GnRH release increases to its maximum frequency at puberty when it induces a rise in LH and FSH, ultimately leading to gonadal maturation. Kisspeptin neurons, located in the RP3V and ARC, are a major activator of GnRH release. Disruption of pubertal timing could occur after a modification of kisspeptin level due to pAOP-1: an increased activation of ER in RP3V; pAOP-2: a decreased TAC2 expression in ARC; pAOP-3: an inhibition of the methylation of Kiss1 TFs; or pAOP-4: a decreased sensitivity to leptin. pAOP, putative adverse outcome pathway; GnRH, gonadotropin-releasing hormone; LH, luteinizing hormone; FSH, follicle-stimulating hormone; RP3V, rostral periventricular area of the third ventricle; ARC, arcuate nucleus; ER, estrogen receptor; TF, transcription factors; TAC2, tachykinin 2.

Fig. 3.

pAOP network for disrupted puberty onset involving kisspeptin neurons. Pulsatile GnRH release increases to its maximum frequency at puberty when it induces a rise in LH and FSH, ultimately leading to gonadal maturation. Kisspeptin neurons, located in the RP3V and ARC, are a major activator of GnRH release. Disruption of pubertal timing could occur after a modification of kisspeptin level due to pAOP-1: an increased activation of ER in RP3V; pAOP-2: a decreased TAC2 expression in ARC; pAOP-3: an inhibition of the methylation of Kiss1 TFs; or pAOP-4: a decreased sensitivity to leptin. pAOP, putative adverse outcome pathway; GnRH, gonadotropin-releasing hormone; LH, luteinizing hormone; FSH, follicle-stimulating hormone; RP3V, rostral periventricular area of the third ventricle; ARC, arcuate nucleus; ER, estrogen receptor; TF, transcription factors; TAC2, tachykinin 2.

Close modal

Supporting Evidence

Kisspeptin is an essential activator of GnRH release during puberty, as evidenced by hypogonadotropic hypogonadic patients with mutations of the kisspeptin receptor [40, 41]. There are 2 distinct hypothalamic populations of kisspeptin neurons in rodents, one in the RP3V which is sensitive to estrogens and mediates the estradiol positive feedback and one in the ARC, negatively regulated by estradiol [42]. The expression of Kiss1 mRNA, the number of kisspeptin neurons in RP3V, and their synaptic contacts with GnRH neurons increase just before the onset of puberty [43-45]. Studies in transgenic mice lacking ERα in kisspeptin neurons suggest that estrogens are necessary for increased expression of kisspeptin in RP3V during puberty [46, 47]. The same observation has been made with ERβ, with receptor invalidation inducing delayed puberty and a decrease in the number of kisspeptin-positive cells in RP3V at postnatal day 25 [48]. This population of kisspeptin neurons appears to be involved in the last step of puberty onset, to stimulate the first LH surge to induce the first ovulation and complete puberty [49].

Evidence for Chemical Triggers

Many chemicals are able to bind to and activate ER [50], and among them several are suspected to induce early puberty [3]. Initial publications measuring Kiss1 expression in the whole hypothalamus without discriminating the 2 key populations observed either an increase [51] or a decrease [52, 53] after early exposure to ER modulators BPA or DES. More recent studies have observed an increase in RP3V kisspeptin expression after perinatal BPA exposure [54-56]; two of them linked this increase with a disruption of pubertal timing [54, 55]. Despite a similar effect on kisspeptin expression, the effects on vaginal opening were opposite. Perinatal exposure to BPA 5 μg/kg/day was associated with early puberty [55] while maternal exposure to a higher dose (BPA 50 μg/kg/day) was associated with a delay [54]. It is also notable that Patisaul et al. [57] did not see any modification in kisspeptin immunoreactivity in the anterior ventral periventricular nucleus after neonatal exposure to the same high BPA dose (50 μg/kg/day). The same group reported also an advance of vaginal opening after neonatal exposure to another EDC, the phytoestrogen genistein, but this early puberty was associated with a decrease in kisspeptin fiber density in the anterior ventral periventricular nucleus [58]. More studies reporting the impact of EDC exposure on kisspeptin expression in RP3V are reviewed by Lopez-Rodriguez et al. [59]. As reported by Parent et al. [3], several publications show that pubertal effects vary with timing or doses of exposure. However, all indicate that modifications of ER activation or kisspeptin level in RP3V are associated with changes in puberty onset in both directions.

pAOP-2: “Decrease of TAC2 in ARC Leads to a Delay of Puberty”

This pAOP proposes to link a decrease in tachykinin 2 (TAC2) gene expression in the ARC to delayed puberty via a reduction of kisspeptin stimulation (shown in Fig. 3).

Supporting Evidence

In the ARC, kisspeptins are mainly expressed by KNDy neurons. These neurons coexpress kisspeptin, neurokinin B (NKB), and dynorphin (DYN). This population is critical for puberty onset. As for Kiss1 and Kiss1R mutations, the loss-of-function mutations in TAC3 (coding for NKB in human) or TACR3 (coding for its receptor) cause pubertal failure and hypogonadotropic hypogonadism in humans [60, 61]. In female rats, the inhibition of DYN signaling by the administration of its receptor antagonist leads to early puberty [62]. Thus, NKB and DYN appear to exert a reciprocal modulation of kisspeptin release, which is critical for GnRH secretion modulation. Using GCaMP6 fiber photometry technology, the Herbison lab recently demonstrated the crucial role of ARC kisspeptin neurons by showing that those neurons show a rhythmic increase in intracellular calcium corresponding to LH release [63].

Evidence for Chemical Triggers

The expression of Kiss1 and Tac2 in the ARC is sensitive to early stress such as undernutrition or high-fat diet [64-66]. Early exposure to EDCs also leads to modifications of Kiss1 expression in the ARC, although the resulting effects depend on the nature, dose, and period of exposure. Neonatal exposure to dibutyl phthalate (0.5–50 mg/kg/day) increases Kiss1 mRNA and kisspeptin immunoreactivity in the ARC and leads to advanced puberty in female rats [67] while exposure to genistein (10 mg/kg/day) during the first 3 days of life leads to a decrease in kisspeptin immunoreactive fibers in the ARC and to advanced vaginal opening [68]. Recently, Ruiz-Pino et al. [55] have shown by in situ hybridization that the expression of both Kiss1 and Tac2 in the ARC decreases after perinatal exposure to a low dose of BPA (5 μg/kg/day) in mice. This decrease was, however, associated with early onset of puberty but a decrease in LH level [55]. Those examples illustrate the sensitivity of the KNDy system to EDC exposure even if the consequences can vary depending on the dose or period of exposure.

pAOP-3: “Inhibition of the Methylation of Kiss1 Transcriptional Regulators Leads to a Delay of Puberty”

This pAOP proposes to link an inhibition of the methylation of Kiss1 transcriptional regulators to delayed puberty via a reduction in kisspeptin expression (shown in Fig. 3).

Supporting Evidence

Transgenic mouse harboring mutations in KISS1 or in KISS1R genes [69] or prepubertal rats treated with an effective kisspeptin antagonist [70] fail to undergo pubertal maturation on time, indicating the importance of kisspeptin in pubertal timing. Our understanding of the molecular mechanisms responsible for the precise timing of puberty has expanded recently, due in part to the recognition of the role of epigenetic mechanisms in the control of normal pubertal maturation. It is now clearly established that a switch from epigenetic repression to activation within kisspeptin neurons in the ARC is a core mechanism underlying the initiation of female puberty [71, 72]. The transcriptional activity of activating genes such as Kiss1 is repressed by silencing molecules, epitomized by the polycomb (PcG) complex. PcG proteins catalyze the deposition of histone PTMs associated with gene silencing at key regulatory regions. As puberty approaches, these PcG proteins are evicted from promoter regions controlling puberty-activating genes in the ARC, and as a result of this loss, the content of histone repressive marks is reduced [73, 74]. This hypothesis has been confirmed by the administration of an inhibitor of DNA methylation which delays puberty in female rats and is associated with the absence of decreased expression of the transcriptional repressors Eed and Cbx7 [74].

Evidence for Chemical Triggers

Several studies have shown that exposure to EDCs can modify DNA methyl-transferase expression in the brain [75-78]. In addition, hypothalamic methylation of Esr1, coding for ERalpha, is increased at puberty after gestational exposure to BPA [76]. Recently, other early stresses such as subnutrition have been shown to delay puberty through an increase in Sirt1 and decrease in Kiss1 expression in the ARC. Sirt1 is an epigenetic enzyme which induces histone deacetylation and heterochromatin formation to downregulate gene expression [79]. No study has so far documented direct effect of EDCs on the epigenetic control of Kiss1 expression and puberty onset, but the data described above suggest that this is a likely mechanism.

pAOP-4: “Decreased Leptin Sensitivity Leads to Late Puberty in Females”

This pAOP proposes to link reduced hypothalamic sensitivity to leptin with delayed puberty via an indirect reduction of kisspeptin stimulation in the ARC (shown in Fig. 3).

Supporting Evidence

Leptin is produced by adipose tissue in direct proportion with body fat content. It informs the central nervous system about the status of fat stores and exerts anorectic effects [80]. Leptin-deficient mice develop obesity, but also an absence of puberty and infertility [81, 82]. If food availability is limited, these mice have normal weight but infertility remains and can only be corrected by administration of leptin [83, 84]. Leptin, therefore, appears to be a necessary factor for the initiation of puberty, evidenced by its ability to induce GnRH secretion ex vivo from the prepubertal rat hypothalamus [85]. Indeed, intraperitoneal injection of leptin in 15-day-old female rats results in an acceleration of GnRH pulsatile release [85], but leptin does not act directly on GnRH neurons [86]. Kiss-peptin level is particularly sensitive to changes in metabolic and nutritional cues, and its expression follows leptin levels. A reduction in leptin level is associated with an inhibition of Kiss1 expression while leptin administration increases Kiss1 gene transcription [87]. Recent data indicate that interaction with PACAP neurons, expressing pituitary adenylate cyclase activating polypeptide, in the ventral premammillary nucleus (PMV) and making direct contact with kisspeptin neurons, could be the pathway explaining leptin action on the hypothalamus for puberty control [88]. The deletion of this population in PMV causes delayed puberty, and PMV is a relevant site of leptin action as activating leptin signalization in PMV restores puberty in leptin-deficient mice [89]. Other intermediary neurons have been proposed to convey leptin signals such as GABAergic neurons [90], nitric oxide neurons [91], or POMC neurons [92].

Evidence for Chemical Triggers

Decreased hypothalamic sensitivity to leptin is observed in food restricted [53, 93], but also in obese rats [94]. Early-life exposure to DES or BPA increases leptin levels in rodents [53, 95, 96]. In addition, the stimulatory effect of leptin on GnRH secretion is diminished both by early postnatal exposure to DES and by prenatal nutritional restriction. In a rat model using combined prenatal dietary restriction and postnatal DES exposure, the stimulating effect of leptin on GnRH secretion is completely obliterated [53]. Thus, developmental exposure to EDCs appears to affect GnRH response to the hypothalamic action of leptin. However, intermediary neurons mediating this action remain to be identified.

pAOPs Involving Neurotransmitters

pAOP-5: “Elevated GABA Leads to Late Puberty in Females”

This AOP proposes to link increased hypothalamic GABAergic neurotransmission to delayed puberty in girls (shown in Fig. 4).

Fig. 4.

pAOP network for disrupted onset involving neurotransmitters. The pubertal increase in GnRH release inducing the rise of LH/FSH and gonadal stimulation is under the control of excitatory and inhibitory neurotransmitters. In the hypothalamus, an increase in GABAergic neurotransmission can lead to a delayed puberty (pAOP-5) while an increase of glutamatergic neurotransmission can induce an early puberty (pAOP-6). pAOP, putative adverse outcome pathway; GnRH, gonadotropin-releasing hormone; LH, luteinizing hormone; FSH, follicle-stimulating hormone.

Fig. 4.

pAOP network for disrupted onset involving neurotransmitters. The pubertal increase in GnRH release inducing the rise of LH/FSH and gonadal stimulation is under the control of excitatory and inhibitory neurotransmitters. In the hypothalamus, an increase in GABAergic neurotransmission can lead to a delayed puberty (pAOP-5) while an increase of glutamatergic neurotransmission can induce an early puberty (pAOP-6). pAOP, putative adverse outcome pathway; GnRH, gonadotropin-releasing hormone; LH, luteinizing hormone; FSH, follicle-stimulating hormone.

Close modal

Supporting Evidence

Even though GABA (γ-aminobutyric acid) direct action on GnRH neurons seems excitatory, the global effect of GABA on the GnRH network is inhibitory [97]. GABA levels in the preoptic area of the hypothalamus decrease just before puberty [98], and the inhibitory effect of GABA on pulsatile GnRH secretion is relatively higher between days 5 and 15 postnatally but decreases at puberty in the rodent [99-101]. This decrease in GABAergic input is crucial for the reactivation of GnRH release at puberty.

Evidence for Chemical Triggers

GABA action on GnRH neurons can be modulated by environmental/peripheral inputs, such as metabolic or steroidal status or EDC exposure. For instance, the frequency of spontaneous GABAergic postsynaptic currents in GnRH neurons and GnRH neuronal activity is decreased by acute fasting [102, 103]. GABAergic transmission is also regulated by estradiol and progesterone. ERα agonists reduce GABA transmission frequency to GnRH neurons [104, 105] while progesterone is an allosteric agonist of the GABA A receptor [103]. Early exposure to EDCs such as BPA can lead to increased [106-108] or decreased [77, 106, 109, 110] GABAergic neurotransmission depending on the dose and the timing of exposure. Gestational exposure to BPA leads to increased GABA release associated with decreased GnRH and testosterone release in male rats [107]. Postnatal exposure to a very low dose of BPA led to delayed puberty associated with higher hypothalamic GABAergic tone and slower GnRH pulsatile release [111]. Recently, children (from 7 to 12 years old) living near a ferromanganese alloy plant in Brazil have been reported to develop early onset of puberty [112]. A translational study in rats showed that exposure to manganese stimulates GnRH release and advances puberty onset decreasing GABA A receptor signaling in the preoptic area [113]. All these pieces of evidence show that the GABAergic pathway is influenced by environmental inputs, including chemical compounds that can disturb GnRH release and pubertal timing.

pAOP-6: “Elevated Glutamate Leads to Early Puberty in Females”

This AOP proposes to link increased hypothalamic glutamatergic neurotransmission to advanced puberty in girls (shown in Fig. 4).

Supporting Evidence

Glutamate concentration increases in the hypothalamus throughout postnatal development until puberty [98]. Its NMDA receptors appear to play an important role in the regulation of GnRH secretion and the initiation of puberty. Intermittent administration of NMDA, in a pulsatile mode, can induce early puberty in monkeys [114] and in rats [115, 116]. Similarly, administration of specific NMDA receptor antagonists delays the onset of puberty [117, 118] and inhibits LH secretion in vivo [119, 120] and GnRH secretion in vitro [121].

Evidence for Chemical Triggers

Several studies indicate an impact of EDCs on glutamatergic signalization in the brain [122-124] but very few have focused on the hypothalamic GnRH network. In adult females, Aroclor 1221, a PCB mix, decreases NMDA-NR2b subunit mRNA expression in the preoptic area [125]. In female rats, neonatal exposure to dichlorodiphenyltrichloroethane, an estrogenic insecticide, induces early puberty and increases glutamate-evoked GnRH release. This last effect involves AMPA, estrogen, and androgen receptors [126, 127]. A decrease in glutamate and in GnRH release was observed in prepubertal male rats after perinatal exposure to BPA [128]. While more investigations in this field are required, these first pieces of evidence suggest an impact of EDCs on the glutamatergic pathway regulating GnRH release.

The AOP concept implies that improved speed and accuracy of chemical testing can be achieved by relying more on alternative test methods than on animal testing or on improving current in vivo test methods by including new and very sensitive endpoints. Currently, in vivo toxicity test guidelines established by OECD include examination of puberty timing by assessing age at vaginal opening in females and preputial separation in males, as well as the weight of the gonads [38]. More sensitive targets could be examined. Puberty timing serves as an AO in putative AOPs identified here. The identification of KEs that are measurable in vivo or in vitro would open up for opportunities to predict adverse effects on puberty timing. Puberty is induced by an increase in GnRH pulsatile release. Hypothalamic explants obtained from female rats have the particularity to release GnRH in the incubation medium in a pulsatile pattern similar to what is reported in vivo [13]. Changes in pulsatile GnRH secretion ex vivo appear to be particularly sensitive to EDC exposure [53, 111, 126, 129] and represent a KE to detect or predict disruption of puberty. This technique is nevertheless challenging to perform in large scale. A new tool, the development of GnRH-secreting neurons from human pluripotent stem cells [130, 131], could be relevant in the future to perform this kind of analysis, but these new cells require further characterization of their pulsatile secretory properties. However, the changes in GnRH release are often the consequences of the disruption of GnRH regulators.

As described in this review, we see a potential for test assay development in relation to KEs associated with kisspeptins and GABA/glutamate. The level of expression of those key hypothalamic factors using transcriptomic (RNAsequencing and RT-qPCR) or proteomic (Western blot and immunohistochemistry) analysis after early in vivo exposure could help identify chemicals that disrupt puberty. Specific in vitro assays need also to be developed to reduce or avoid animal use and to supplement OECD in vitro test guidelines. Currently, only the identified KE “increased activity of ER” is included in these guidelines. The activity of kisspeptin or GABA/glutamate promotors can be studied by gene reporter assay. Using this technique, Heger et al. [53, 132] have previously shown that the kisspeptin promotor is sensitive to BPA and DES. In addition, rodent hypothalamic primary cell culture [132], specific immortalized hypothalamic cell lines [133], hypothalamic neurons generated from human pluripotent stem cells [134], or new models of complex neuronal cocultures [135] could be developed as a tool to determine the impact of EDC on the expression of a specific target such as kisspeptin or GABA/glutamate.

Puberty is a critical step towards reproductive maturity and health, and altered pubertal timing is associated with an increased risk of pathologies later in life [136-139]. Herein, we have proposed 6 AOPs that can help explain, at the mechanistic level, how EDCs disturb puberty onset. The difficulty of building AOPs relevant to disturbed puberty lies in the fact that the hypothalamic control of puberty is still incompletely understood. Many knowledge gaps persist regarding the specific role of the 2 populations of kisspeptin neurons, the leptin targets in the hypothalamus, and the epigenetic regulation of GnRH neuron activity. Our 6 pAOPs focus only on identified EDC targets. For instance, kisspeptin neurons are also regulated by metabolic sensors (such as mTor and AMPK) [140], other hypothalamic neurons (as POMC-AgRP neurons) [66], or specific protein such as Makorin ring finger protein 3 [141, 142], but which have yet to be described as EDC targets. Along the same line, other epigenetic mechanisms regulating puberty have recently been identified (miRNA and tritorax group) [143, 144], but their sensitivity to EDCs remains to be studied. GnRH neuron migration has been suggested to be affected by EDCs [145, 146], but more investigations are needed to confirm this theory and to build an AOP. In addition, the HPG axis that regulates puberty onset is influenced by gonadal feedback while gonads are themselves targets of EDCs. This latter aspect was not discussed here as a large review of putative ovarian AOPs has been published recently [147]. Nevertheless, the development of our 6 pAOPs has underlined KEs that can detect or predict disruption of puberty: pulsatile GnRH release and hypothalamic expression of kisspeptin and of GABA/glutamate. Those factors can be considered as good biomarkers for in vivo or in vitro test methods. Based on these data, it is now time to develop robust alternative test assays to help safeguard female reproductive health.

The authors have no conflicts of interest to declare.

This project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 825100

D. Franssen, A.-S. Parent, and T. Svingen wrote the manuscript; J. Boberg, D. Lopez Rodriguez, and Majorie Van Duursen critically reviewed the manuscript. All authors have read and agreed to the submitted version of the manuscript.

1.
Tanner
J
.
Growth at adolescence
.
Blakwell S. Oxford
;
1962
.
2.
Parent
A-SS
,
Teilmann
G
,
Juul
A
,
Skakkebaek
NE
,
Toppari
J
,
Bourguignon
J-PJP
.
The timing of normal puberty and the age limits of sexual precocity: variations around the world, secular trends, and changes after migration
.
Endocr Rev
.
2003 Oct
;
24
(
5
):
668
93
.
3.
Parent
AS
,
Franssen
D
,
Fudvoye
J
,
Gérard
A
,
Bourguignon
JP
.
Developmental variations in environmental influences including endocrine disruptors on pubertal timing and neuroendocrine control: revision of human observations and mechanistic insight from rodents
.
Front Neuroendocrinol
.
2015
;
38
:
12
36
. .
4.
Zhu
J
,
Chan
YM
.
Adult consequences of self-limited delayed puberty
.
Pediatrics
.
2017
;
139
(
6
):
e20163177
.
5.
Day
FR
,
Thompson
DJ
,
Helgason
H
,
Chasman
DI
,
Finucane
H
,
Sulem
P
,
Genomic analyses identify hundreds of variants associated with age at menarche and support a role for puberty timing in cancer risk
.
Nat Genet
.
2017
;
49
(
6
):
834
41
. .
6.
Cheng
TS
,
Day
FR
,
Lakshman
R
,
Ong
KK
.
Association of puberty timing with type 2 diabetes: a systematic review and meta analysis
.
PLoS Med
.
2020 Jan 6
;
17
(
1
):
e1003017
.
7.
Mendle
J
,
Ryan
RM
,
McKone
KMP
.
Age at menarche, depression, and antisocial behavior in adulthood
.
Pediatrics
.
2018 Jan
;
141
(
1
):
e20171703
.
8.
Johansson
HKL
,
Svingen
T
,
Fowler
PA
,
Vinggaard
AM
,
Boberg
J
.
Environmental influences on ovarian dysgenesis: developmental windows sensitive to chemical exposures
.
Nat Rev Endocrinol
.
2017
;
13
(
7
):
400
14
. .
9.
Plant
TM
,
Zeleznik
AJ
, editors.
Knobil and Neill’s physiology of reproduction
. 4th ed.
2015
.
10.
Becú-Villalobos
D
,
González Iglesias
A
,
Díaz-Torga
G
,
Hockl
P
,
Libertun
C
.
Brain sexual differentiation and gonadotropins secretion in the rat
.
Cell Mol Neurobiol
.
1997
;
17
(
6
):
699
715
. .
11.
Harris
GC
,
Levine
JE
.
Pubertal acceleration of pulsatile gonadotropin-releasing hormone release in male rats as revealed by microdialysis
.
Endocrinology
.
2003
;
144
(
1
):
163
71
. .
12.
Sisk
CL
,
Richardson
HN
,
Chappell
PE
,
Levine
JE
,
Program
N
.
In vivo gonadotropin-releasing hormone secretion in female rats during peripubertal development and on proestrus
.
Endocrinology
.
2001
;
142
(
7
):
2929
36
. .
13.
Bourguignon
JP
,
Franchimont
P
.
Puberty-related increase in episodic LHRH release from rat hypothalamus in vitro
.
Endocrinology
.
1984
;
114
(
5
):
1941
3
. .
14.
Ojeda
,
Skinner
MK
.
Chapter 38. Puberty in the rat
. In:
Knobil
E
,
Neill
JD
, editors.
Knobil and Neill’s physiology of reproduction
.
Gulf Professional Publishing
;
2006
.
Vol. 2
; p.
2061
103
.
15.
Caron
E
,
Ciofi
P
,
Prevot
V
,
Bouret
SG
.
Alteration in neonatal nutrition causes perturbations in hypothalamic neural circuits controlling reproductive function
.
J Neurosci
.
2012
;
32
(
33
):
11486
94
. .
16.
Messina
A
,
Langlet
F
,
Chachlaki
K
,
Roa
J
,
Rasika
S
,
Jouy
N
,
A microRNA switch regulates the rise in hypothalamic GnRH production before puberty
.
Nat Neurosci
.
2016
;
19
(
6
):
835
44
. .
17.
Parent
AS
,
Teilmann
G
,
Juul
A
,
Skakkebaek
NE
,
Toppari
J
,
Bourguignon
JP
.
The timing of normal puberty and the age limits of sexual precocity: variations around the world, secular trends, and changes after migration
.
Endocr Rev
.
2003 Oct
;
24
(
5
):
668
93
. .
18.
Aksglaede
L
,
Sørensen
K
,
Petersen
JH
,
Skakkebaek
NE
,
Juul
A
.
Recent decline in age at breast development: the Copenhagen puberty study
.
Pediatrics
.
2009
;
123
(
5
):
e932
. .
19.
Euling
SY
,
Herman-Giddens
ME
,
Lee
PA
,
Selevan
SG
,
Juul
A
,
Sørensen
TI
,
Examination of US puberty-timing data from 1940 to 1994 for secular trends: panel findings
.
Pediatrics
.
2008
;
121
(
Suppl 3
):
S172
. .
20.
Biro
FM
,
Greenspan
LC
,
Galvez
MP
,
Pinney
SM
,
Teitelbaum
S
,
Windham
GC
,
Onset of breast development in a longitudinal cohort
.
Pediatrics
.
2013
;
132
(
6
):
1019
27
. .
21.
Fudvoye
J
,
Lopez-Rodriguez
D
,
Franssen
D
,
Parent
AS
.
Endocrine disrupters and possible contribution to pubertal changes
.
Best Pract Res Clin Endocrinol Metab
.
2019
;
33
(
3
):
101300
.
22.
Lamb
JC
,
Boffetta
P
,
Foster
WG
,
Goodman
JE
,
Hentz
KL
,
Rhomberg
LR
,
Critical comments on the WHO-UNEP State of the Science of Endocrine Disrupting Chemicals: 2012
.
Regul Toxicol Pharmacol
.
2014
;
69
(
1
):
22
40
.
23.
Bergman
A
,
Heindel
JJ
,
Kasten
T
,
Kidd
KA
,
Jobling
S
,
Neira
M
,
The impact of endocrine disruption: a consensus statement on the state of the science
.
Environ Health Perspect
.
2013
;
121
(
4
):
A104
6
. .
24.
WHO/UNEP
.
Global assessment of the state-of-the-science of endocrine disruptors
. In:
Damstra
T
,
Barlow
S
,
Bergman
A
,
Kavlock
R
,
Van Der Kraakeds
G
, editors.
2013
.
25.
Wohlfahrt-Veje
C
,
Andersen
HR
,
Schmidt
IM
,
Aksglaede
L
,
Sørensen
K
,
Juul
A
,
Early breast development in girls after prenatal exposure to non-persistent pesticides
.
Int J Androl
.
2012
;
35
(
3
):
273
82
. .
26.
Marks
KJ
,
Hartman
TJ
,
Taylor
EV
,
Rybak
ME
,
Northstone
K
,
Marcus
M
.
Exposure to phytoestrogens in utero and age at menarche in a contemporary British cohort
.
Environ Res
.
2017
;
155
:
287
93
. .
27.
Adgent
MA
,
Daniels
JL
,
Rogan
WJ
,
Adair
L
,
Edwards
LJ
,
Westreich
D
,
Early-life soy exposure and age at menarche
.
Paediatr Perinat Epidemiol
.
2012 Mar
;
26
(
2
):
163
75
. .
28.
Strom
BL
,
Schinnar
R
,
Ziegler
EE
,
Barnhart
KT
,
Sammel
MD
,
Macones
GA
,
Exposure to soy-based formula in infancy and endocrinological and reproductive outcomes in young adulthood
.
JAMA
.
2001
;
286
(
7
):
807
14
. .
29.
Harley
KG
,
Rauch
SA
,
Chevrier
J
,
Kogut
K
,
Parra
KL
,
Trujillo
C
,
Association of prenatal and childhood PBDE exposure with timing of puberty in boys and girls
.
Environ Int
.
2017
;
100
:
132
8
. .
30.
Windham
GC
,
Pinney
SM
,
Voss
RW
,
Sjödin
A
,
Biro
FM
,
Greenspan
LC
,
Brominated flame retardants and other persistent organohalogenated compounds in relation to timing of puberty in a longitudinal study of girls
.
Environ Health Perspect
.
2015 Oct
;
123
(
10
):
1046
52
. .
31.
Chen
A
,
Chung
E
,
DeFranco
EA
,
Pinney
SM
,
Dietrich
KN
.
Serum PBDEs and age at menarche in adolescent girls: analysis of the National Health and Nutrition Examination Survey 2003–2004
.
Environ Res
.
2011
;
111
(
6
):
831
7
. .
32.
van Duursen
MBM
,
Boberg
J
,
Christiansen
S
,
Connolly
L
,
Damdimopoulou
P
,
Filis
P
,
Safeguarding female reproductive health against endocrine disrupting chemicals: the FREIA project
.
Int J Mol Sci
.
2020
;
21
(
9
):
3215
.
33.
Johansson
HKL
,
Damdimopoulou
P
,
van Duursen
MBM
,
Boberg
J
,
Franssen
D
,
de Cock
M
,
Putative adverse outcome pathways for female reproductive disorders to improve testing and regulation of chemicals
.
Arch Toxicol
.
2020 Oct
;
94
(
10
):
3359
79
.
34.
Villeneuve
DL
,
Crump
D
,
Garcia-Reyero
N
,
Hecker
M
,
Hutchinson
TH
,
LaLone
CA
,
Adverse outcome pathway development II: best practices
.
Toxicol Sci
.
2014
;
142
(
2
):
321
30
. .
35.
Draskau
MK
,
Spiller
CM
,
Boberg
J
,
Bowles
J
,
Svingen
T
.
Developmental biology meets toxicology: contributing reproductive mechanisms to build adverse outcome pathways
.
Mol Hum Reprod
.
2020 Jan
;
26
(
2
):
111
6
.
36.
Krewski
D
,
Acosta
D
 Jr
,
Andersen
M
,
Anderson
H
,
Bailar
JC
 3rd
,
Boekelheide
K
,
Toxicity testing in the 21st century: a vision and a strategy
.
J Toxicol Environ Health B Crit Rev
.
2010
;
13
(
2–4
):
51
138
.
37.
Ankley
GT
,
Edwards
SW
.
The adverse outcome pathway: a multifaceted framework supporting 21st century toxicology
.
Curr Opin Toxicol
.
2018
;
9
:
1
7
.
38.
OECD
.
Users’ handbook supplement to the guidance document for developing and assessing adverse outcome pathways
.
2016
.
39.
Knapen
D
,
Vergauwen
L
,
Villeneuve
DL
,
Ankley
GT
.
The potential of AOP networks for reproductive and developmental toxicity assay development
.
Reprod Toxicol
.
2015
;
56
:
52
5
. .
40.
Seminara
SB
,
Messager
S
,
Chatzidaki
EE
,
Thresher
RR
,
Acierno
JS
,
Shagoury
JK
,
The GPR54 gene as a regulator of puberty
.
N Engl J Med
.
2003 Oct
;
349
(
17
):
1614
27
. .
41.
de Roux
N
,
Genin
E
,
Carel
JC
,
Matsuda
F
,
Chaussain
JL
,
Milgrom
E
.
Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54
.
Proc Natl Acad Sci U S A
.
2003 Sep
;
100
(
19
):
10972
6
. .
42.
García-Galiano
D
,
Pinilla
L
,
Tena-Sempere
M
.
Sex steroids and the control of the Kiss1 system: developmental roles and major regulatory actions
.
J Neuroendocrinol
.
2012 Jan
;
24
(
1
):
22
33
.
43.
Clarkson
J
,
Herbison
AE
.
Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons
.
Endocrinology
.
2006
;
147
(
12
):
5817
25
. .
44.
Navarro
VM
,
Castellano
JM
,
Fernández-Fernández
R
,
Barreiro
ML
,
Roa
J
,
Sanchez-Criado
JE
,
Developmental and hormonally regulated messenger ribonucleic acid expression of KiSS-1 and its putative receptor, GPR54, in rat hypothalamus and potent luteinizing hormone-releasing activity of KiSS-1 peptide
.
Endocrinology
.
2004 Oct
;
145
(
10
):
4565
74
. .
45.
Semaan
SJ
,
Kauffman
AS
.
Daily successive changes in reproductive gene expression and neuronal activation in the brains of pubertal female mice
.
Mol Cell Endocrinol
.
2015
;
401
:
84
97
. .
46.
Clarkson
J
,
Boon
WC
,
Simpson
ER
,
Herbison
AE
.
Postnatal development of an estradiol-kisspeptin positive feedback mechanism implicated in puberty onset
.
Endocrinology
.
2009
;
150
(
7
):
3214
20
. .
47.
Mayer
C
,
Acosta-Martinez
M
,
Dubois
SL
,
Wolfe
A
,
Radovick
S
,
Boehm
U
,
Timing and completion of puberty in female mice depend on estrogen receptor alpha-signaling in kisspeptin neurons
.
Proc Natl Acad Sci U S A
.
2010
;
107
(
52
):
22693
8
. .
48.
Naulé
L
,
Robert
V
,
Parmentier
C
,
Martini
M
,
Keller
M
,
Cohen-Solal
M
,
Delayed pubertal onset and prepubertal Kiss1 expression in female mice lacking central oestrogen receptor beta
.
Hum Mol Genet
.
2015
;
24
(
25
):
7326
38
. .
49.
Herbison
AE
.
Control of puberty onset and fertility by gonadotropin-releasing hormone neurons
.
Nat Rev Endocrinol
.
2016
;
12
(
8
):
452
66
. .
50.
Diamanti-Kandarakis
E
,
Bourguignon
JP
,
Giudice
LC
,
Hauser
R
,
Prins
GS
,
Soto
AM
,
Endocrine-disrupting chemicals: an endocrine society scientific statement
.
Endocr Rev
.
2009
;
30
(
4
):
293
342
. .
51.
Xi
W
,
Lee
CK
,
Yeung
WS
,
Giesy
JP
,
Wong
MH
,
Zhang
X
,
Effect of perinatal and postnatal bisphenol A exposure to the regulatory circuits at the hypothalamus-pituitary-gonadal axis of CD-1 mice
.
Reprod Toxicol
.
2011
;
31
(
4
):
409
17
. .
52.
Navarro
VM
,
Sánchez-Garrido
MA
,
Castellano
JM
,
Roa
J
,
García-Galiano
D
,
Pineda
R
,
Persistent impairment of hypothalamic KiSS-1 system after exposures to estrogenic compounds at critical periods of brain sex differentiation
.
Endocrinology
.
2009
;
150
(
5
):
2359
67
. .
53.
Franssen
D
,
Ioannou
YS
,
Alvarez-real
A
,
Gerard
A
,
Mueller
JK
,
Heger
S
,
Pubertal timing after neonatal diethylstilbestrol exposure in female rats: neuroendocrine vs peripheral effects and additive role of prenatal food restriction
.
Reprod Toxicol
.
2014
;
44
:
63
72
. .
54.
Naulé
L
,
Picot
M
,
Martini
M
,
Parmentier
C
,
Hardin-Pouzet
H
,
Keller
M
,
Neuroendocrine and behavioral effects of maternal exposure to oral bisphenol A in female mice
.
J Endocrinol
.
2014
;
220
(
3
):
375
88
. .
55.
Ruiz-Pino
F
,
Miceli
D
,
Franssen
D
,
Vazquez
MJ
,
Farinetti
A
,
Castellano
JM
,
Environmentally relevant perinatal exposures to bisphenol A disrupt postnatal Kiss1/NKB neuronal maturation and puberty onset in female mice
.
Environ Health Perspect
.
2019 Oct
;
127
(
10
):
107011
. .
56.
Wang
X
,
Chang
F
,
Bai
Y
,
Chen
F
,
Zhang
J
,
Chen
L
.
Bisphenol A enhances kisspeptin neurons in anteroventral periventricular nucleus of female mice
.
J Endocrinol
.
2014
;
221
(
2
):
201
13
. .
57.
Patisaul
HB
,
Todd
KL
,
Mickens
JA
,
Adewale
HB
.
Impact of neonatal exposure to the ERalpha agonist PPT, bisphenol-A or phytoestrogens on hypothalamic kisspeptin fiber density in male and female rats
.
Neurotoxicology
.
2009
;
30
(
3
):
350
7
. .
58.
Bateman
HL
,
Patisaul
HB
.
Disrupted female reproductive physiology following neonatal exposure to phytoestrogens or estrogen specific ligands is associated with decreased GnRH activation and kisspeptin fiber density in the hypothalamus
.
Neurotoxicology
.
2008 Nov
;
29
(
6
):
988
97
. .
59.
Lopez-Rodriguez
D
,
Franssen
D
,
Bakker
J
,
Lomniczi
A
,
Parent
AS
.
Cellular and molecular features of EDC exposure: consequences for the GnRH network
.
Nat Rev Endocrinol
.
2021 Feb
;
17
(
3
):
83
96
.
60.
Topaloglu
AK
,
Reimann
F
,
Guclu
M
,
Yalin
AS
,
Kotan
LD
,
Porter
KM
,
TAC3 and TACR3 mutations in familial hypogonadotropic hypogonadism reveal a key role for Neurokinin B in the central control of reproduction
.
Nat Genet
.
2009 Mar
;
41
(
3
):
354
8
. .
61.
Guran
T
,
Tolhurst
G
,
Bereket
A
,
Rocha
N
,
Porter
K
,
Turan
S
,
Hypogonadotropic hypogonadism due to a novel missense mutation in the first extracellular loop of the neurokinin B receptor
.
J Clin Endocrinol Metab
.
2009
;
94
(
10
):
3633
9
. .
62.
Nakahara
T
,
Uenoyama
Y
,
Iwase
A
,
Oishi
S
,
Nakamura
S
,
Minabe
S
,
Chronic peripheral administration of kappa-opioid receptor antagonist advances puberty onset associated with acceleration of pulsatile luteinizing hormone secretion in female rats
.
J Reprod Dev
.
2013
;
59
(
5
):
479
84
. .
63.
Clarkson
J
,
Han
SY
,
Piet
R
,
McLennan
T
,
Kane
GM
,
Ng
J
,
Definition of the hypothalamic GnRH pulse generator in mice
.
Proc Natl Acad Sci U S A
.
2017 Nov
;
114
(
47
):
E10216
23
. .
64.
Castellano
JM
,
Bentsen
AH
,
Sánchez-Garrido
MA
,
Ruiz-Pino
F
,
Romero
M
,
Garcia-Galiano
D
,
Early metabolic programming of puberty onset: impact of changes in postnatal feeding and rearing conditions on the timing of puberty and development of the hypothalamic kisspeptin system
.
Endocrinology
.
2011 Sep
;
152
(
9
):
3396
408
. .
65.
Navarro
VM
,
Tena-Sempere
M
.
Neuroendocrine control by kisspeptins: role in metabolic regulation of fertility
.
Nat Rev Endocrinol
.
2012 Jan
;
8
(
1
):
40
53
. .
66.
Navarro
VM
.
Metabolic regulation of kisspeptin: the link between energy balance and reproduction
.
Nat Rev Endocrinol
.
2020
;
16
(
8
):
407
20
.
67.
Hu
J
,
Du
G
,
Zhang
W
,
Huang
H
,
Chen
D
,
Wu
D
,
Short-term neonatal/prepubertal exposure of dibutyl phthalate (DBP) advanced pubertal timing and affected hypothalamic kisspeptin/GPR54 expression differently in female rats
.
Toxicology
.
2013
;
314
(
1
):
65
75
. .
68.
Losa
SM
,
Todd
KL
,
Sullivan
AW
,
Cao
J
,
Mickens
JA
,
Patisaul
HB
.
Neonatal exposure to genistein adversely impacts the ontogeny of hypothalamic kisspeptin signaling pathways and ovarian development in the peripubertal female rat
.
Reprod Toxicol
.
2011
;
31
(
3
):
280
9
. .
69.
Colledge
WH
.
Transgenic mouse models to study Gpr54/kisspeptin physiology
.
Peptides
.
2009 Jan
;
30
(
1
):
34
41
. .
70.
Pineda
R
,
Garcia-Galiano
D
,
Roseweir
A
,
Romero
M
,
Sanchez-Garrido
MA
,
Ruiz-Pino
F
,
Critical roles of kisspeptins in female puberty and preovulatory gonadotropin surges as revealed by a novel antagonist
.
Endocrinology
.
2010
;
151
(
2
):
722
30
. .
71.
Lomniczi
A
,
Loche
A
,
Castellano
JM
,
Ronnekleiv
OK
,
Bosch
M
,
Kaidar
G
,
Epigenetic control of female puberty
.
Nat Neurosci
.
2013 Mar
;
16
(
3
):
281
9
. .
72.
Aylwin
CF
,
Vigh-Conrad
K
,
Lomniczi
A
,
Aylwin
CF
,
Vigh-Conrad
K
.
The emerging role of chromatin remodeling factors in female pubertal development
.
Neuroendocrinology
.
2019 Feb
;
109
(
3
):
208
17
. .
73.
Ojeda
SR
,
Lomniczi
A
,
Sandau
U
,
Matagne
V
.
New concepts on the control of the onset of puberty
.
Endocr Dev
.
2010
;
17
:
44
51
. .
74.
Lomniczi
A
,
Wright
H
,
Castellano
JM
,
Matagne
V
,
Toro
CA
,
Ramaswamy
S
,
Epigenetic regulation of puberty via Zinc finger protein-mediated transcriptional repression
.
Nat Commun
.
2015
;
6
(
1
):
10195
. .
75.
Kumar
D
,
Thakur
MK
.
Effect of perinatal exposure to Bisphenol-A on DNA methylation and histone acetylation in cerebral cortex and hippocampus of postnatal male mice
.
J Toxicol Sci
.
2017
;
42
(
3
):
281
9
. .
76.
Kundakovic
M
,
Gudsnuk
K
,
Franks
B
,
Madrid
J
,
Miller
RL
,
Perera
FP
,
Sex-specific epigenetic disruption and behavioral changes following low-dose in utero bisphenol A exposure
.
Proc Natl Acad Sci U S A
.
2013
;
110
(
24
):
9956
61
. .
77.
Zhou
R
,
Chen
F
,
Chang
F
,
Bai
Y
,
Chen
L
.
Persistent overexpression of DNA methyltransferase 1 attenuating GABAergic inhibition in basolateral amygdala accounts for anxiety in rat offspring exposed perinatally to low-dose bisphenol A
.
J Psychiatr Res
.
2013 Oct
;
47
(
10
):
1535
44
. .
78.
Walker
DM
,
Goetz
BM
,
Gore
AC
.
Dynamic postnatal developmental and sex-specific neuroendocrine effects of prenatal polychlorinated biphenyls in rats
.
Mol Endocrinol
.
2014 Jan
;
28
(
1
):
99
115
. .
79.
Vazquez
MJ
,
Toro
CA
,
Castellano
JM
,
Ruiz-Pino
F
,
Roa
J
,
Beiroa
D
,
SIRT1 mediates obesity- and nutrient-dependent perturbation of pubertal timing by epigenetically controlling Kiss1 expression
.
Nat Commun
.
2018
;
9
(
1
):
4194
. .
80.
Meister
B
.
Control of food intake via leptin receptors in the hypothalamus
.
Vitam Horm
.
2000
;
59
:
265
304
. [Online]. Available from: http://www.ncbi.nlm.nih.gov/pubmed/10714243. .
81.
Farooqi
IS
.
Leptin and the onset of puberty: insights from rodent and human genetics
.
Semin Reprod Med
.
2002
;
20
(
2
):
139
44
. .
82.
Halaas
JL
,
Gajiwala
KS
,
Maffei
M
,
Cohen
SL
,
Chait
BT
,
Rabinowitz
D
,
Weight-reducing effects of the plasma protein encoded by the obese gene
.
Science
.
1995
;
269
(
5223
):
543
6
. .
83.
Barash
IA
,
Cheung
CC
,
Weigle
DS
,
Ren
H
,
Kabigting
EB
,
Kuijper
JL
,
Leptin is a metabolic signal to the reproductive system
.
Endocrinology
.
1996
;
137
(
7
):
3144
7
. .
84.
Chehab
FF
,
Lim
ME
,
Lu
R
.
Correction of the sterility defect in homozygous obese female mice by treatment with the human recombinant leptin
.
Nat Genet
.
1996 Mar
;
12
(
3
):
318
20
. .
85.
Lebrethon
MC
,
Aganina
A
,
Fournier
M
,
Gérard
A
,
Parent
AS
,
Bourguignon
JP
.
Effects of in vivo and in vitro administration of ghrelin, leptin and neuropeptide mediators on pulsatile gonadotrophin-releasing hormone secretion from male rat hypothalamus before and after puberty
.
J Neuroendocrinol
.
2007 Mar
;
19
(
3
):
181
8
. .
86.
Quennell
JH
,
Mulligan
AC
,
Tups
A
,
Liu
X
,
Phipps
SJ
,
Kemp
CJ
,
Leptin indirectly regulates gonadotropin-releasing hormone neuronal function
.
Endocrinology
.
2009
;
150
(
6
):
2805
12
. .
87.
Manfredi-Lozano
M
,
Roa
J
,
Tena-Sempere
M
.
Connecting metabolism and gonadal function: novel central neuropeptide pathways involved in the metabolic control of puberty and fertility
.
Front Neuroendocrinol
.
2018
;
48
:
37
49
. .
88.
Ross
RA
,
Leon
S
,
Madara
JC
,
Schafer
D
,
Fergani
C
,
Maguire
CA
,
PACAP neurons in the ventral premammillary nucleus regulate reproductive function in the female mouse
.
Elife
.
2018
;
7
:
e35960
. .
89.
Donato
J
,
Cravo
RM
,
Frazão
R
,
Gautron
L
,
Scott
MM
,
Lachey
J
,
Leptin’s effect on puberty in mice is relayed by the ventral premammillary nucleus and does not require signaling in Kiss1 neurons
.
J Clin Invest
.
2011
;
121
(
1
):
355
68
. .
90.
Martin
C
,
Navarro
VM
,
Simavli
S
,
Vong
L
,
Carroll
RS
,
Lowell
BB
,
Leptin-responsive GABAergic neurons regulate fertility through pathways that result in reduced kisspeptinergic tone
.
J Neurosci
.
2014
;
34
(
17
):
6047
56
. .
91.
Bellefontaine
N
,
Chachlaki
K
,
Parkash
J
,
Vanacker
C
,
Colledge
W
,
d'Anglemont de Tassigny
X
,
Leptin-dependent neuronal NO signaling in the preoptic hypothalamus facilitates reproduction
.
J Clin Invest
.
2014
;
124
(
6
):
2550
9
. .
92.
Manfredi-Lozano
M
,
Roa
J
,
Ruiz-Pino
F
,
Piet
R
,
Garcia-Galiano
D
,
Pineda
R
,
Defining a novel leptin-melanocortin-kisspeptin pathway involved in the metabolic control of puberty
.
Mol Metab
.
2016 Oct
;
5
(
10
):
844
57
. .
93.
Bautista
CJ
,
Boeck
L
,
Larrea
F
,
Nathanielsz
PW
,
Zambrano
E
.
Effects of a maternal low protein isocaloric diet on milk leptin and progeny serum leptin concentration and appetitive behavior in the first 21 days of neonatal life in the rat
.
Pediatr Res
.
2008
;
63
(
4
):
358
63
. .
94.
Ramírez
S
,
Claret
M
.
Hypothalamic ER stress: a bridge between leptin resistance and obesity
.
FEBS Lett
.
2015
;
589
(
14
):
1678
87
. .
95.
Angle
BM
,
Do
RP
,
Ponzi
D
,
Stahlhut
RW
,
Drury
BE
,
Nagel
SC
,
Metabolic disruption in male mice due to fetal exposure to low but not high doses of bisphenol A (BPA): evidence for effects on body weight, food intake, adipocytes, leptin, adiponectin, insulin and glucose regulation
.
Reprod Toxicol
.
2013
;
42
:
256
68
. .
96.
Newbold
RR
,
Padilla-Banks
E
,
Snyder
RJ
,
Phillips
TM
,
Jefferson
WN
.
Developmental exposure to endocrine disruptors and the obesity epidemic
.
Reprod Toxicol
.
2007 Apr
;
23
(
3
):
290
6
. .
97.
Ojeda
S
,
Skinner
M
.
Puberty in the rat
. 3rd ed.
San Diego
:
Academic Press/Elsevier
;
2006
. p.
2061
126
. .
98.
Goroll
D
,
Arias
P
,
Wuttke
W
.
Preoptic release of amino acid neurotransmitters evaluated in peripubertal and young adult female rats by push-pull perfusion
.
Neuroendocrinology
.
1993
;
58
(
1
):
11
5
. .
99.
Bourguignon
J-P
,
Gerard
A
,
Purnelle
G
,
Czajkowski
V
,
Yamanaka
C
,
Lemaı
M
.
Duality of glutamatergic and gabaergic control of pulsatile GnRH secretion by rat hypothalamic explants: II. Reduced NR2C- and GABA A -receptor-mediated inhibition at initiation of sexual maturation
.
J Neuroendocrinol
.
1997
;
9
:
193
9
.
100.
Bourguignon
J-P
,
Gérard
A
,
Purnelle
G
,
Czajkowski
V
,
Yamanaka
C
,
Lemaı
M
,
Duality of glutamatergic and gabaergic control of pulsatile GnRH secretion by rat hypothalamic explants: II. ReducedNR2C- and GABAA-receptor-mediated inhibition atinitiation of sexual maturation
.
J Neuroendocrinol
.
1997
;
9
(
3
):
193
9
.
101.
Parent
AS
,
Matagne
V
,
Bourguignon
JP
.
Control of puberty by excitatory amino acid neurotransmitters and its clinical implications
.
Endocrine
.
2005
;
28
(
3
):
281
6
. .
102.
Sullivan
SD
,
Moenter
SM
.
Gamma-aminobutyric acid neurons integrate and rapidly transmit permissive and inhibitory metabolic cues to gonadotropin-releasing hormone neurons
.
Endocrinology
.
2004 Mar
;
145
(
3
):
1194
202
. .
103.
Sullivan
SD
,
DeFazio
RA
,
Moenter
SM
.
Metabolic regulation of fertility through presynaptic and postsynaptic signaling to gonadotropin-releasing hormone neurons
.
J Neurosci
.
2003 Sep
;
23
(
24
):
8578
85
. .
104.
Chu
Z
,
Andrade
J
,
Shupnik
MA
,
Moenter
SM
.
Differential regulation of gonadotropin-releasing hormone neuron activity and membrane properties by acutely applied estradiol: dependence on dose and estrogen receptor subtype
.
J Neurosci
.
2009
;
29
(
17
):
5616
27
. .
105.
Kwakowsky
A
,
Cheong
RY
,
Herbison
AE
,
Ábrahám
IM
.
Non-classical effects of estradiol on cAMP responsive element binding protein phosphorylation in gonadotropin-releasing hormone neurons: mechanisms and role
.
Front Neuroendocrinol
.
2014 Jan
;
35
(
1
):
31
41
. .
106.
Cabaton
NJ
,
Canlet
C
,
Wadia
PR
,
Tremblay-Franco
M
,
Gautier
R
,
Molina
J
,
Effects of low doses of bisphenol A on the metabolome of perinatally exposed CD-1 mice
.
Environ Health Perspect
.
2013
;
121
(
5
):
586
93
. .
107.
Cardoso
N
,
Pandolfi
M
,
Lavalle
J
,
Carbone
S
,
Ponzo
O
,
Scacchi
P
,
Probable gamma-aminobutyric acid involvement in bisphenol A effect at the hypothalamic level in adult male rats
.
J Physiol Biochem
.
2011
;
67
(
4
):
559
67
. .
108.
Ogi
H
,
Itoh
K
,
Ikegaya
H
,
Fushiki
S
.
Alterations of neurotransmitter norepinephrine and gamma-aminobutyric acid correlate with murine behavioral perturbations related to bisphenol A exposure
.
Brain Dev
.
2015 Sep
;
37
(
8
):
739
46
. .
109.
Parent
AS
,
Franssen
D
,
Fudvoye
J
,
Pinson
A
,
Bourguignon
JP
.
Current changes in pubertal timing: revised vision in relation with environmental factors including endocrine disruptors
.
Endocr Dev
.
2016
;
29
:
174
84
. .
110.
Zhou
R
,
Bai
Y
,
Yang
R
,
Zhu
Y
,
Chi
X
,
Li
L
,
Abnormal synaptic plasticity in basolateral amygdala may account for hyperactivity and attention-deficit in male rat exposed perinatally to low-dose bisphenol-A
.
Neuropharmacology
.
2011
;
60
(
5
):
789
98
. .
111.
Franssen
D
,
Gérard
A
,
Hennuy
B
,
Donneau
AF
,
Bourguignon
JP
,
Parent
AS
.
Delayed neuroendocrine sexual maturation in female rats after a very low dose of bisphenol A through altered gabaergic neurotransmission and opposing effects of a high dose
.
Endocrinology
.
2016
;
157
(
5
):
1740
50
. .
112.
dos Santos
NR
,
Rodrigues
JLG
,
Bandeira
MJ
,
Anjos
ALDS
,
Araújo
CFS
,
Adan
LFF
,
Manganese exposure and association with hormone imbalance in children living near a ferro-manganese alloy plant
.
Environ Res
.
2019
;
172
:
166
74
. .
113.
Yang
X
,
Tan
J
,
Xu
X
,
Yang
H
,
Wu
F
,
Xu
B
,
Prepubertal overexposure to manganese induce precocious puberty through GABAA receptor/nitric oxide pathway in immature female rats
.
Ecotoxicol Environ Saf
.
2020 Jan
;
188
:
109898
.
114.
Gay
VL
,
Plant
TM
.
N-methyl-d, l-aspartate elicits hypothalamic gonadotropin-releasing hormone release in prepubertal male rhesus monkeys (macaca mulatto)
.
Endocrinology
.
1987
;
120
(
6
):
2289
96
.
115.
Smyth
C
,
Wilkinson
M
.
A critical period for glutamate receptor-mediated induction of precocious puberty in female rats
.
J Neuroendocrinol
.
1994
;
6
(
3
):
275
84
. [Online]. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7920593. .
116.
Urbanski
HF
,
Ojeda
SR
.
Activation of luteinizing hormone-releasing hormone release advances the onset of female puberty
.
Neuroendocrinology
.
1987
;
46
(
3
):
273
6
. .
117.
Meijs-Roelofs
HM
,
Kramer
P
,
van Leeuwen
EC
.
The N-methyl-d-aspartate receptor antagonist MK-801 delays the onset of puberty and may acutely block the first spontaneous LH surge and ovulation in the rat
.
J Endocrinol
.
1991
;
131
(
3
):
435
41
. .
118.
Urbanski
HF
,
Ojeda
SR
.
A role for n-methyl-d-aspartate (NMDA) receptors in the control of lh secretion and initiation of female puberty
.
Endocrinology
.
1990
;
126
(
3
):
1774
6
. .
119.
Urbanski
HF
,
Ojeda
SR
.
A role for N-methyl-D-aspartate (NMDA) receptors in the control of LH secretion and initiation of female puberty
.
Endocrinology
.
1990 Mar
;
126
(
3
):
1774
6
. .
120.
Bourguignon
JP
,
Gérard
A
,
Franchimont
P
.
Direct activation of gonadotropin-releasing hormone secretion through different receptors to neuroexcitatory amino acids
.
Neuroendocrinology
.
1989
;
49
(
4
):
402
8
. .
121.
Bourguignon
JP
,
Gerard
A
,
Mathieu
J
,
Mathieu
A
,
Franchimont
P
.
Maturation of the hypothalamic control of pulsatile gonadotropin-releasing hormone secretion at onset of puberty. I. Increased activation of N-methyl-D-aspartate receptors
.
Endocrinology
.
1990
;
127
(
2
):
873
81
. .
122.
Zhang
H
,
Kuang
H
,
Luo
Y
,
Liu
S
,
Meng
L
,
Pang
Q
,
Low-dose bisphenol A exposure impairs learning and memory ability with alterations of neuromorphology and neurotransmitters in rats
.
Sci Total Environ
.
2019
;
697
:
134036
.
123.
Chen
Z
,
Li
T
,
Zhang
L
,
Wang
H
,
Hu
F
.
Bisphenol A exposure remodels cognition of male rats attributable to excitatory alterations in the hippocampus and visual cortex
.
Toxicology
.
2018
;
410
:
132
41
. .
124.
Alavian-Ghavanini
A
,
Lin
PI
,
Lind
PM
,
Risén Rimfors
S
,
Halin Lejonklou
M
,
Dunder
L
,
Prenatal bisphenol A exposure is linked to epigenetic changes in glutamate receptor subunit gene grin2b in female rats and humans
.
Sci Rep
.
2018
;
8
(
1
):
11315
. .
125.
Dickerson
SM
,
Cunningham
SL
,
Gore
AC
.
Prenatal PCBs disrupt early neuroendocrine development of the rat hypothalamus
.
Toxicol Appl Pharmacol
.
2011
;
252
(
1
):
36
46
. .
126.
Rasier
G
,
Parent
AS
,
Gérard
A
,
Lebrethon
MC
,
Bourguignon
JP
.
Early maturation of gonadotropin-releasing hormone secretion and sexual precocity after exposure of infant female rats to estradiol or dichlorodiphenyltrichloroethane
.
Biol Reprod
.
2007 Oct
;
77
(
4
):
734
42
. .
127.
Rasier
G
,
Parent
A-S
,
Gérard
A
,
Denooz
R
,
Lebrethon
M-C
,
Charlier
C
,
Mechanisms of interaction of endocrine-disrupting chemicals with glutamate-evoked secretion of gonadotropin-releasing hormone
.
Toxicol Sci
.
2008 Mar
;
102
(
1
):
33
41
. .
128.
Cardoso
N
,
Pandolfi
M
,
Ponzo
O
,
Carbone
S
,
Szwarcfarb
B
,
Scacchi
P
,
Evidence to suggest glutamic acid involvement in Bisphenol A effect at the hypothalamic level in prepubertal male rats
.
Neuro Endocrinol Lett
.
2010
;
31
(
4
):
512
6
.
129.
Lopez-Rodriguez
D
,
Franssen
D
,
Sevrin
E
,
Gérard
A
,
Balsat
C
,
Blacher
S
,
Persistent vs transient alteration of folliculogenesis and estrous cycle after neonatal vs adult exposure to bisphenol A
.
Endocrinology
.
2019 Nov
;
160
(
11
):
2558
72
.
130.
Lund
C
,
Yellapragada
V
,
Vuoristo
S
,
Balboa
D
,
Trova
S
,
Allet
C
,
Characterization of the human GnRH neuron developmental transcriptome using a GNRH1-TdTomato reporter line in human pluripotent stem cells
.
Dis Model Mech
.
2020
;
13
(
3
):
dmm040105
.
131.
Lund
C
,
Pulli
K
,
Yellapragada
V
,
Giacobini
P
,
Lundin
K
,
Vuoristo
S
,
Development of gonadotropin-releasing hormone-secreting neurons from human pluripotent stem cells
.
Stem Cell Rep
.
2016
;
7
(
2
):
149
57
. .
132.
Mueller
JK
,
Heger
S
.
Endocrine disrupting chemicals affect the gonadotropin releasing hormone neuronal network
.
Reprod Toxicol
.
2014
;
44
:
73
84
. .
133.
McIlwraith
EK
,
Loganathan
N
,
Belsham
DD
.
Phoenixin expression is regulated by the fatty acids palmitate, docosahexaenoic acid and oleate, and the endocrine disrupting chemical bisphenol a in immortalized hypothalamic neurons
.
Front Neurosci
.
2018 Nov
;
12
:
838
. .
134.
Merkle
FT
,
Maroof
A
,
Wataya
T
,
Sasai
Y
,
Studer
L
,
Eggan
K
,
Generation of neuropeptidergic hypothalamic neurons from human pluripotent stem cells
.
Development
.
2015
;
142
(
4
):
633
43
. .
135.
Enright
HA
,
Lam
D
,
Sebastian
A
,
Sales
AP
,
Cadena
J
,
Hum
NR
,
Functional and transcriptional characterization of complex neuronal co-cultures
.
Sci Rep
.
2020 Dec
;
10
(
1
):
11007
. .
136.
Ibáñez
L
,
Díaz
R
,
López-Bermejo
A
,
Marcos
MV
.
Clinical spectrum of premature pubarche: links to metabolic syndrome and ovarian hyperandrogenism
.
Rev Endocr Metab Disord
.
2009 Mar
;
10
(
1
):
63
76
. .
137.
Carter
R
,
Jaccard
J
,
Silverman
WK
,
Pina
AA
.
Pubertal timing and its link to behavioral and emotional problems among ‘at-risk’ African American adolescent girls
.
J Adolesc
.
2009
;
32
(
3
):
467
81
. .
138.
Lakshman
R
,
Forouhi
NG
,
Sharp
SJ
,
Luben
R
,
Bingham
SA
,
Khaw
KT
,
Early age at menarche associated with cardiovascular disease and mortality
.
J Clin Endocrinol Metab
.
2009
;
94
(
12
):
4953
60
. .
139.
Lakshman
R
,
Forouhi
N
,
Luben
R
,
Bingham
S
,
Khaw
K
,
Wareham
N
,
Association between age at menarche and risk of diabetes in adults: results from the EPIC-Norfolk cohort study
.
Diabetologia
.
2008
;
51
(
5
):
781
6
. .
140.
Vazquez
MJ
,
Velasco
I
,
Tena-Sempere
M
.
Novel mechanisms for the metabolic control of puberty: implications for pubertal alterations in early-onset obesity and malnutrition
.
J Endocrinol
.
2019 Aug
;
242
(
2
):
R51
65
.
141.
Abreu
AP
,
Toro
CA
,
Song
YB
,
Navarro
VM
,
Bosch
MA
,
Eren
A
,
MKRN3 inhibits the reproductive axis through actions in kisspeptin-expressing neurons
.
J Clin Invest
.
2020 Aug
;
140
(
8
):
4486
500
.
142.
Heras
V
,
Sangiao-Alvarellos
S
,
Manfredi-Lozano
M
,
Sanchez-Tapia
MJ
,
Ruiz-Pino
F
,
Roa
J
,
Hypothalamic miR-30 regulates puberty onset via repression of the puberty-suppressing factor, Mkrn3
.
PLoS Biol
.
2019 Nov
;
17
(
11
):
e3000532
. .
143.
Lomniczi
A
,
Aylwin
C
,
Vigh-Conrad
K
.
The emerging role of chromatin remodeling factors in female pubertal development
.
Neuroendocrinology
.
2019
;
109
(
3
):
208
17
.
144.
Sangiao-Alvarellos
S
,
Manfredi-Lozano
M
,
Ruiz-Pino
F
,
Navarro
VM
,
Sánchez-Garrido
MA
,
Leon
S
,
Changes in hypothalamic expression of the Lin28/let-7 system and related microRNAs during postnatal maturation and after experimental manipulations of puberty
.
Endocrinology
.
2013
;
154
(
2
):
942
55
. .
145.
Pillon
D
,
Cadiou
V
,
Angulo
L
,
Duittoz
AH
.
Maternal exposure to 17-alpha-ethinylestradiol alters embryonic development of GnRH-1 neurons in mouse
.
Brain Res
.
2012 Jan
;
1433
:
29
37
. .
146.
Vosges
M
,
Le Page
Y
,
Chung
BC
,
Combarnous
Y
,
Porcher
JM
,
Kah
O
,
17alpha-ethinylestradiol disrupts the ontogeny of the forebrain GnRH system and the expression of brain aromatase during early development of zebrafish
.
Aquat Toxicol
.
2010 Sep
;
99
(
4
):
479
91
. .
147.
Johansson
HKL
,
Damdimopoulou
P
,
van Duursen
MBM
,
Boberg
J
,
Franssen
D
,
de Cock
M
,
Putative adverse outcome pathways for female reproductive disorders to improve testing and regulation of chemicals
.
Arch Toxicol
.
2020 Oct 1
;
94
(
10
):
3359
79
.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.