Sei sulla pagina 1di 8

Reprod Dom Anim 45, e495–e502 (2010); doi: 10.1111/j.1439-0531.2009.01355.

x
ISSN 0936-6768

Review Article

Neuroendocrine, Metabolic and Genomic Cues Signalling the Onset of Puberty in


Females
CA Meza-Herrera1, A Gonzalez-Bulnes2, RT Kridli3, M Mellado4, CF Arechiga-Flores5, H Salinas6 and JM Luginbuhl7
1
Universidad Autónoma Chapingo, Unidad Regional Universitaria de Zonas Áridas, Bermejillo, Durango, Me´xico; 2INIA, Departamento de
Reproducción, Madrid, Spain; 3Jordan University of Science and Technology, Irbid, Jordan; 4Universidad Autonoma Agraria Antonio Narro,
Mexico; 5Universidad Autonoma de Zacatecas, Mexico; 6Instituto Nacional de Investigaciones Forestales Agricolas y Pecuarias, Mexico; 7North
Carolina State University. Raleigh, NC, USA

Contents concepts, which are involved in the establishment of


Puberty is the result of a dynamic interaction between genetic the hypothalamic-hypophyseal-gonadal axis function
factors and environmental cues, all of which lead to the that promotes the onset of the reproductive function
attainment of reproductive capacity. Thus, significant changes during puberty. To achieve that purpose, some basic
in hormone secretion occur from the pre-pubertal to the aspects of the function of the hypothalamic-hypophy-
pubertal stage. The objective of this review is to provide an seal-gonadal axis, the control of the axis by neurotrans-
update of some endocrine, physiological, metabolic and mitters and the interaction between reproductive
genetic concepts involved in the establishment of the hypo-
function and metabolic status will be considered.
thalamic-hypophyseal-gonadal axis function promoting the
onset of the reproductive function during puberty. To achieve Finally, the role of the novel kisspeptin system and the
this purpose, basic aspects of the function of the hypotha- GPR54 receptor as modulators of puberty will be
lamic-hypophyseal-gonadal axis, the control of the axis by considered, aside from the hierarchical expression of
neurotransmitters and the interaction between reproductive the main genes acting as regulators of the onset of
function and metabolic status will be considered. Finally, the puberty: OCT-2, TTF-1 and EAP-1.
role of the novel kisspeptin system and the GPR54 receptor as
modulators of puberty will be considered, in addition to the
hierarchical expression of the main genes acting as regulators Basic Functional Aspects of the Hypothalamic-
of the onset of puberty. Hypophyseal-Gonadal Axis
The onset of puberty is a centrally-regulated process,
Introduction and the detailed mechanisms of its function are still
unknown. It is recognized, yet, that increases in the
The transit from pre-puberty to the pubertal state activity of the GnRH pulse generator and then in the
includes significant changes in the function of the pulsatile secretions of gonadotrophins FSH and LH
hypothalamic-hypophyseal-gonadal axis. Prior to pub- take place. During puberty, the pulsatile secretions of
erty, secretion of the gonadotrophin releasing hormone gonadotrophins are increased during the day, promoting
(GnRH) is low. As puberty approaches, the hypotha- a normal gonadal development and function because of
lamic gonadostat is still depressed, whilst the amplitude the activation of the GnRH pulse generator (Teresawa
of GnRH pulses increases. The concentrations of 2005; Clarkson and Herbison 2006; Messinis 2006;
gonadotrophic hormones, follicle stimulating hormone Ojeda et al. 2006a).
(FSH) and luteinizing hormone (LH) gradually increase The classical gonadostat theory states that a decreased
during puberty, thus stimulating growth and maturation sensitivity to the oestradiol negative feedback must occur
of follicles as well as oestrogen production by the in the hypothalamic-hypohpyseal centres controlling
ovaries (Huffman et al. 1987; Apter 1997; Apter and gonadotropin secretion in order for pubertal onset
Hermanson 2002; Teresawa 2005; Messinis 2006; Mahdi (Ramirez and McCann 1963). In this way, a decreased
and Khallili 2008). sensitivity to esteroid feedback allows increases in
Immediately prior to puberty, the highly efficient gonadotropin secretion, which promotes ovarian follicle
negative feedback system from the ovarian steroids is maturation, and ovulation. Yet, certain concepts of the
particularly active. However, as puberty approaches, the gonadostat theory may not be suitable for all species. In
positive feedback signal from the ovarian steroids is sheep, GnRH secretion is markedly suppressed prior
activated and becomes highly efficient, usually observing puberty, whereas at the beginning of puberty, the
the establishment of normal cyclicity at the end of that hypothalamic gonadostat is also depressed and the
stage. For this reason, the complete transition from amplitude of the GnRH pulses increases (Foster and
prepubertal anoestrus into puberty is an indispensable Ryan 1979). Thereafter, concentrations of LH and FSH
prerequisite for the establishment of reproductive func- are gradually increased during puberty, stimulating both
tion (Teresawa 2005; Clarkson and Herbison 2006; follicular maturation and ovarian oestrogen production.
Messinis 2006; Ojeda et al. 2006a). The decapeptide GnRH is synthesized in the pre-optic
The objective of this review is to actualize some area of the hypothalamus and, in adult animals it is
endocrine, physiological, metabolic and genetic

 2009 Blackwell Verlag GmbH


e496 CA Meza-Herrera, A Gonzalez-Bulnes, RT Kridli, M Mellado, CF Arechiga-Flores, H Salinas and JM Luginbuhl

released in a pulsatile fashion, every 60 min, inside the 2000; Mahesh and Brann 2005; Parent et al. 2005;
portal system which connects to the hypophysis. Then, Clarkson and Herbison 2006; Ojeda et al. 2006a,b).
GnRH binds to its high affinity receptor (GnRH-R) Not only serotonin (5-HT) and the gamma amino
located not only on the cellular membrane of the butyric acid (GABA), but also the catecholamines (CA)
gonadothrops, but also on the ovary and uterus. In display qualitative differences upon GnRH and LH
young animals, the pulse interval is much longer, 90– secretion, mainly at the beginning of the prepubertal
120 min. It is the acceleration of the frequency of the stage than in late puberty and the adult stage (Moguil-
pulse and the increase in the amplitude of the pulse, as evsky and Wuttke 2001). Similarly, it has been estab-
well as the total GnRH concentration that activate lished that the main trans-synaptic excitatory event
transduction signals to promote both the synthesis and stimulating the onset of puberty is an increase in the
the intermittent release of LH and FSH, thus generating glutamatergic neurotransmission because it is the main
the onset of the pubertal process. Once bound to its way of trans-synaptic communication in the hypothal-
receptor, GnRH works through protein kinase-C to amus. This is a complex process controlled by a plethora
induce synthesis and the secretion of LH and FSH. The of genes required for the synthesis, transport and release
GnRH-R expression is regulated by GnRH itself as well of glutamate, as well as the expression of several
as by gonadal steroids (Huffman et al. 1987; Apter 1997; receptors, both ionotropics and metabotropics which
Roth et al. 2001; Apter and Hermanson 2002). mediate the actions of glutamate (Ebling et al. 1989;
Those neurons releasing GnRH represent the critical Dhandapani and Brann 2000; Mahesh and Brann 2005;
cellular type, which, once activated, induce puberty Parent et al. 2005; Clarkson and Herbison 2006; Ojeda
(Clarkson and Herbison 2006). For that reason, an et al. 2006a,b).
appropriate expression of the GnRH-R in the gonado- In primates, the LHRH neurons of the female
trophs is critical for the signalling and secretion of hypothalamus are active during the neonatal period,
gonadotrophins mediating the ulterior sexual develop- but they subsequently enter into an arrested stage in the
ment (Zapatero-Caballero et al. 2004). The GnRH juvenile and prepubertal periods, because of the high
neurons are important not only because of their GABA levels in the median eminence (ME). The
involvement in the onset of reproductive function, but observed reduction in GABA levels, as the animal
also in the development of neuromodulatory functions grows results in an increase in LHRH release in the ME,
in the adult (Whitlock et al. 2006). generating the onset of the pubertal process. This
The discovery of the ligand activator of the upstream reduction in GABA levels seems to allow glutamate
signal route from GnRH linked to the G-protein levels in the ME to increase and seems to contribute to
receptor gives greater significance to the central role the generation of the pubertal increase of LHRH
the hypothalamus plays in the regulation of puberty (Teresawa 2005).
(Clarkson and Herbison 2006; Hughes and Kumanan Both, the administration of 5-hydroxytryptophan, a
2006). The onset of puberty is associated with an serotonin precursor, and the activation of the GABAergic
increase in the amplitude and frequency of LH. Later system, increase the release of GnRH and LH during
on, a progressive increase in the LH pulsatility occurs prepubertal stages, although such positive effect disap-
during the day, while a progressive reduction of the pears during late puberty and promotes an inhibitory
amplification, previously occurring during the night, is action upon the GnRH neurons in adult animals. Like
observed. Prepubertal FSH concentrations are relatively glutamate, GABA production requires the participation
high, and a continuous process of follicular development of different proteins involved in the synthesis, metabo-
and atresia occurs, while relatively high concentrations lism, transport and release of this neurotransmitter. As
of oestradiol are observed. Only after this initial increase no changes in the mRNA expression of the enzymes
in LH is the ovarian steroidogenesis activated, leading responsible for GABA, GAD65 and GAD67 have been
to increases in oestrogen secretion. Thereafter, both observed during the sexual development in primates, it
follicular development and maturation occur under seems that the expression of such genes is not an event
FSH stimulation (Suttie et al. 1991; Apter 1997). related to the onset of puberty. Yet, by analogy with the
glutamate system, it can be inferred that other possible
checkpoints in GABA metabolism include the vesicular
The Control of Puberty by Modulation of transport and ⁄ or the expression of GABA receptors.
Different Neurotransmitter Systems Concerning the latter, four GABA transporters have
As previously mentioned, GnRH is the primary mes- been described: GAT-1, GAT-2, GAT-3 and BGT-1;
senger involved in both sexual maturation and onset of GAT-1 is the predominant transporter in the mamma-
puberty. The activity of this neuronal system is, in turn, lian brain and contains a responsive element to oestro-
controlled by different neurotransmitter systems. The gen (Teresawa 2005; Ojeda et al. 2006b). Conversely,
onset of puberty includes changes in a prepubertal inhibition of CA synthesis (adrenaline and noradrena-
secretion type of gonadotrophins characterized by a low line) by alpha-methyl.p-tyrosine promotes an increase in
activity of the GnRH neurons, to later promote an LH secretion during the early prepubertal phase and an
increase in the amplitude and frequency of GnRH inhibitory effect during both the late pubertal phase and
pulses. At the beginning of puberty, several neurotrans- the adult stage. The latter highlights the inhibitory effect
mitter systems are involved in the change of GnRH displayed by CA upon the hypothalamus-hypophyseal
secretion pattern, which act through quantitative and axis function to change to a stimulatory effect during
qualitative modifications upon the GnRH secretion late puberty and adult stage (Moguilevsky and Wuttke
pattern (Ebling et al. 1989; Dhandapani and Brann 2001).

 2009 Blackwell Verlag GmbH


Signalling Mechanisms Controlling Puberty e497

One of the most important orexigenic factors is the Puberty is characterized by an increase in the sensi-
neuropeptide-Y (NPY), a 36-amino acid peptide. It is a bility of GnRH to glutamate and an increase in the
member of the pancreatic polypeptides family and is activity of the enzyme glutaminase in the hypothalamus.
very abundant in the CNS, including the hypothalamus, The secretion of GnRH by glutamatergic and GAB-
as well as the peripheral nervous system (Wojcik- Aergic neurons seems to occur in an independent way in
Gladysz and Polkowska 2006). Wankowska et al. the peripubertal stage. In addition, the activation in the
(2002), evaluating the intracerebroventricular infusion transynaptic glutamatergic regulation of GnRH is
of NPY upon the secretory activity of the hypophyseal required, as well as a coordinated activity of glutama-
gonadotrophs, reported that NPY can be an important tergic neurons and astroglial cells, which also display an
component in the mechanisms promoting synthesis and important active role during puberty (Parent et al.
storage of LH in the gonadotrophs, without observable 2005).
changes on the LH serum concentrations. The same Glutamate generates different actions in the hypotha-
authors concluded that NPY does not affect LH release lamic mechanism of puberty as has been demonstrated
in prepubertal ewe lambs. Similarly, such effect was LH- in different animal models (Parent et al. 2005). The pre-
specific without apparent changes in FSH. optic area has been implicated as the primary site of
On the other hand, opiodergic and neuroexcitatory action of NMDA, whereas NMDA agonists seem to act
amino acid systems display quantitative differences in the mainly at the level of the ME and the arquate nuclei
secretion of GnRH-LH during the prepubertal, peri- (Mahesh and Brann 2005). Participation of the kainate
pubertal and adult stages. Opioids have a significant receptor in the induction of an oestradiol-dependent
inhibitory effect during early pre-puberty that decreases precocious puberty suggests that this receptor can be
during sexual maturation until puberty is reached. In involved in the activation of GnRH secretion mediated
fact, endogenous opioids are important inhibitory neu- by oestradiol, which has been observed during puberty.
rotransmitters and control the pulsatile LH release in In addition, and in parallel to the increase in glutamate
peripubertal stages (Ebling et al. 1989). Peptide opioids release in the hypothalamus during the puberty process,
as a group do not seem to inhibit the onset of puberty, but DL-amino methyl propionic acid receptors are also
it is possible that their inhibitory tone can be exerted by a increased and play an important role as mediators of
neuronal subset, which selectively utilize for neurotrans- the regulatory effects of glutamate during the establish-
mission to the B-endorphin, dinorphin or Met ⁄ Leu- ment of puberty (Zamorano et al. 1998).
enkephalin (Teresawa 2005; Ojeda et al. 2006b). As previously mentioned, glutamate has been
On the contrary, excitatory amino acids (EAA) involved in critical processes of the establishment of
increase their stimulatory effect upon GnRH-LH secre- puberty, hormonal pulsatility and sexual behaviour.
tion during sexual maturation through increases in the Glutamate can promote many of these effects through
release of aspartate and glutamate, as well as by activation of the release of the gaseous neurotransmitter
increasing the sensibility of the N-methyl-D-aspartate nitric oxide. This neurotransmitter stimulates the GnRH
(NMDA) receptors (Moguilevsky and Wuttke 2001). release through activation of the enzyme guanylate
According to Mahesh and Brann (2005) and Parent cyclase, which in turn promotes an increase in the
et al. (2005), EAA in general and glutamate in particular production of cGMP and the posterior release of GnRH
exert a marked stimulatory effect upon the reproductive (Dhandapani and Brann 2000).
axis, particularly at the time of puberty. In fact, Little is known about the transcriptional control of
administration of glutamate and its agonists promote the genes which codify the enzymes involved in the
the pulsatile release of LH in animals previously exposed synthesis, metabolism and transport of glutamate
to steroids. Conversely, antagonists of the ionotropic (Ojeda et al. 2006b). Yet, the importance of these
receptors of glutamate inhibit the LH release, and homeostatic systems has been recently evaluated
abolish both the secretion of LH induced by E2 and the through the use of proteomic quantitative techniques.
pre-ovulatory LH surge. According to Teresawa (2005), In fact Roth et al. (2006), analysing some of the proteins
the pubertal reduction of the GABAergic inhibition is involved in the synthesis and degradation of glutamate
followed by an increase in the glutamatergic tone. in the astroglial cells, reported that the enzyme gluta-
Ojeda et al. (2006a) reported that the onset of puberty mate dehydrogenase catalyses the synthesis of gluta-
requires a reciprocal communication neuron-glia which mate. They indicated that the highest content of
involves EAA, mainly glutamate and aspartate. In glutamate in the hypothalamus was observed during
addition, some growth factors and the coordinated proestrus, while the concentration of the enzyme gluta-
action of a group of neuromodulators, all of which mine synthase that catalyses the metabolism from
representing a higher level of control governing the glutamate to glutamine decreased during proestrus. An
pubertal process, are also necessary. Dhandapani and increased production of glutamate and LHRH was
Brann (2000) as well as Mahesh and Brann (2005) observed in the hypothalamus prior to the pre-ovulatory
mentioned that both the neuromodulator glutamate as LH surge. The latter suggests that an increase in the
well as nitric oxide play an essential role in the excitatory effects generated by the glutamatergic signals
establishment of the pre-ovulatory surge of GnRH in of glial origin upon the neuronal LHRH networks is an
mammals. Respectively, it has also been reported that event, which contributes to the activation in the pulsatile
glutamate, NMDA and kainate stimulate GnRH secre- pubertal secretion of LHRH. The transcriptional regu-
tion in immature mammals, whereas NMDA-R stimu- lation of the expression of the vesicular transporters of
lation generates a precocious puberty in rats and glutamate can represent a very important checkpoint of
monkeys. glutamate homeostasis, because the vesicular transporters

 2009 Blackwell Verlag GmbH


e498 CA Meza-Herrera, A Gonzalez-Bulnes, RT Kridli, M Mellado, CF Arechiga-Flores, H Salinas and JM Luginbuhl

of glutamate are extremely important in the minute-to- to play a central role as a regulatory signal in the
minute control of the release of such neurotransmitter integration of the energetic homeostasis and reproduc-
(Ojeda et al. 2006b). tive function (Fernandez-Fernandez et al. 2006).
Leptin, the product of the OB gene, has been related
to the onset of puberty in several animal models. The
Modulation of Neuroendocrine Systems and onset of puberty differs among genus, and such differ-
Metabolic Status ence can be due to the sexual differences observed in the
Metabolic status and nutrient availability are some of mechanisms of sexual organization regarding the feed-
the main environmental factors required for the estab- back systems of steroids (Foster et al. 2006). In prepu-
lishment of reproductive function. The deficit of nutri- bertal females, leptin concentrations increase slowly
ents in several animal species promotes disturbances in with age and body fat reserves. Besides the increase of
gonadotrophic hormone secretion both in immature and fat tissue reserves during puberty, there are also
adult mammals. It has been proposed that decrease in increases in leptin. In general, plasmatic concentrations
the pulsatile secretion of GnRH generates a reduction in of leptin are significantly correlated with the adipose
the synthesis and release of LH. In addition, such a mass level both in males and females. Leptin is bound to
scenario has been further proposed to be one of the a high affinity binding protein similar to the soluble
main etiologic factors to cause nutritionally induced receptor of leptin (Apter and Hermanson 2002; Apter
suppression of the hypophyseal-gonadal function 2003).
(Wojcik-Gladysz and Polkowska 2006). According to Carbone et al. (2005), exogenous
As previously mentioned, GnRH neurons are reason- administration of leptin generated a significant increase
ably mature at birth; yet, as the growth process starts, of LH levels in adult animals as well as of the glutamate
pulsatile release of GnRH is suppressed. In fact, in levels in the hypothalamus. This hypothalamic excit-
ruminants, there is an increase in gonadotrophic activity atory amino acid has been involved in the neurotrans-
in the early stages after birth, which generates an mitter activation of the NMDA-R. These results
increase in the development of antral follicles on the demonstrate that leptin stimulates the reproductive axis
ovary. Endocrine regulation of this stimulation of the during a determined period of sexual maturation and
ovarian follicular system is not fully understood that the NMDA-R is involved in the facilitatory
(Rawlings et al. 2003). action of leptin upon the gonadal axis during sexual
This initial endocrine activity seems to be arrested by maturation.
the suppression of negative feedback mechanisms until Administration of exogenous leptin increases the
females reach a critical live weight (lipostatic theory), release of GnRH in the hypothalamus during prepu-
which can allow the onset of puberty, initiate oestrous bertal and peripubertal stages. In peripubertal females,
cycles and later activate the reproductive function. such increase is accompanied by a significant decrease in
Respectively, there is evidence that antral follicles and the hypothalamic release of GABA and an increase in
genital tubular development occur in a parallel fashion. the release of aspartate. The latter suggests that in this
Similarly, the early increase in the number and size of phase of sexual maturation, leptin exerts a stimulatory
antral follicles can partially be due to changes in both effect upon GnRH, inducing the release of EAA, and
the secretion pattern and the potency of FSH, whereas reducing the release of inhibitory amino acids such as
the increased development in the follicular population is GABA, all of them involved in the control of GnRH
probably caused by increases in the frequency of the release. In prepubertal stages, yet, the stimulatory effect
GnRH pulse generator, and in turn by LH and FSH. of leptin upon GnRH seems to be related to the
The latter occurrs once a critical live weight or certain stimulatory action of GABA, which in this stage of
percentage of body fat is acquired (Rawlings et al. development increases the release of GnRH (Reynoso
2003). et al. 2003).
Low protein levels in the diet promote not only a According to Ponzo et al. (2005), exogenous admin-
reduction in live weight but also a decrease in average istration of leptin to prepubertal and peripubertal
daily gain, exerting an inhibitory effect upon the animals increased the release of hypothalamic GnRH,
synthesis and release of LH. Yet, this effect was not plasma LH concentrations as well as the release of
observed when considering FSH. For this reason, glutamate. An increase in aspartate levels was also
protein concentration in the diet can be an important observed, but only in peripubertal animals treated with
modulator of those neuronal processes promoting the leptin, without changes in GABA levels between treat-
increase in LH levels during puberty (Polkowska et al. ments. The latter demonstrates that leptin promotes
2003). Similarly, severe undernutrition can prevent increases in the hypothalamic-hypophyseal function,
ovulation in ewe lambs by abolishing the function of leading to serum increases of GnRH and LH, and that
the GnRH system, which governs the production and the excitatory amino acid neurotransmitter system
secretion of LH with a high frequency pattern (Foster seems to be involved in these changes.
and Olster 1985). Both leptin and ghrelin exert important roles in the
Even though the close relationship between live regulation of intake behaviour, metabolic status and
weight and fertility has been recognized, the peripheral reproduction by acting upon the CNS and other
signals and the neuroendocrine networks responsible for reproductive organs. As markers of the nutritional
such phenomenon have only been recently identified. status, these hormones can act upon the CNS to
One key event in this field was the cloning of the initiate the complex process of puberty and to maintain
adipocyte derivative hormone, leptin, which has shown a normal reproductive function in the adult stage.

 2009 Blackwell Verlag GmbH


Signalling Mechanisms Controlling Puberty e499

By acting through the brain, these hormones can act as their function as suppressors’ molecules of tumoral
a connection between the level of metabolic reserves metastasis in the brain, also act as integrator elements in
and the reproductive system to regulate energy require- the communication neuron-to-neuron and glia-to-neu-
ments for a normal reproductive function (Budak et al. ron, building a functional unit able to initiate the
2006). puberty process (Ojeda et al. 2006b).
Leptin, which signals the level of energy storage of the Mutations in the GPR54 receptor expression pro-
body, displays an essential role in the metabolic control duce hypogonadotrophic hypogonadism, an indication
of the KiSS1 system, as kisspeptin-neurons co-express that signalling through this receptor is a prerequisite
leptin receptors. Leptin is capable of normalizing the for sexual maturation. Central administration of kiss-
defective expression of the KiSS1 gene in biological peptin stimulates secretion of both GnRH and
models characterized by an impaired secretion of gonadotrophins in prepubertal and adult animals.
gonadotrophins linked to hypoleptinemia stages (Tena- Similarly, increases in the expression of the KiSS1
Sempere 2006a, 2006b). Other factors could play an and GPR54 genes have been observed during the
important role in the attainment of puberty in mam- pubertal development, whereas activation of GPR54 by
mals. These include environmental as well as genetic kisspeptin administration is enough to induce the
factors and the interaction amongst these (Land 1978). activation of the gonadotrophic axis in immature
Environmental factors involve season of birth, nutri- animals (Gottsch et al. 2006; Tena-Sempere 2006a,
tional status including body weight, total body fat, 2006b).
metabolites such as glucose, cholesterol and triglyce- Neurons expressing kisspeptin are direct targets of
rides, as well as metabolic hormones (Cheung et al. the steroid feedback action, both positive and negative,
1997). In goats, it was recently demonstrated that which differently regulate the mRNA KiSS1 expression
puberty was depended upon season of birth, and that in several brain areas and have been defined with a
heavier animals born in November also depicted a relevant role in the establishment of puberty. A double
higher ovulation rate. Therefore, some benefit could be site of kisspeptin action in the brain has been
expected in breeding goat kids at a higher body weight suggested, either in the hypothalamic-hypophyseal
to obtain a higher prolificacy at the first kidding region or the ME, an area located outside the blood
(Zarazaga et al. 2009). brain barrier. Neurons containing kisspeptin are
located in discreet subsets of the pre-optic area and
the arquate nuclei of the brain, whereas those neurons
The Kisspeptin-GPR54 System in the containing the GPR54 receptors are more diffusely
Modulation of GnRH and Puberty distributed and include GnRH neurons as well as the
The hypothalamic gene KiSS1 has been considered as adenohypophysis. In primates, the KiSS1 and GPR54
an essential integrator of peripheral cues including the mRNA levels significantly increase in the hypothala-
gonadal steroids as well as the nutritional status mus at the time of puberty suggesting that an increase
(Tena-Sempere 2006a). In fact, recent data suggest a in the GPR54 mediated signal contributes to the
prominent role of KiSS1 in the metabolic control of pubertal activation of GnRH secretion (Ojeda et al.
fertility when considering that expression of the 2006b).
hypothalamic gene KiSS1 is negatively regulated under The pulsatile increase in GnRH secretion observed in
conditions of negative energy balance, whereas admin- the prepubertal stages occurs due to the coordinated
istration of kisspeptin is capable of reversing a changes in the trans-synaptic communication and to the
hypogonadotrophic stage observed under scenarios of interaction between neuron-to-glial cells. As the excit-
undernutrition and metabolic disturbance (Tena- atory signals from neurons and glial cells increase, a
Sempere 2006b). reduction in inhibition in the trans-synaptic tone is
Kisspeptin, a peptide made up of 53 amino acids, a observed, generating the pubertal activation of GnRH
product of the KiSS1 gene and its receptor GPR54 secretion. The more prevalent neuronal systems invoked
which is linked to G-proteins, have emerged as key in this process include glutamate and the peptide
elements in the regulation of GnRH secretion (Gottsch kisspeptin, which exert neurotransmission and neuro-
et al. 2006; Tena-Sempere 2006a, 2006b, 2006c). Kiss- modulation processes, whereas the most important
peptins were originally identified as tumoral metastasis inhibitory signal is provided by GABAtergic and
suppressor peptides bound to the GPR54 receptor opiatergic neurons (Ojeda et al. 2006b).
linked to G-proteins (Ojeda et al. 2006b). Yet, muta- It has been proposed that glial cells and GnRH
tions in the GPR54 gene have been related to an neurons display a morphological and functional rela-
absence in the onset of puberty and hypogonadotrophic tionship which depends on growth factors that act
hypogonadism (Tena-Sempere 2006c). Furthermore, through serine-threonine-kinase receptors as the trans-
the proteolytic cleavage of the KiSS1 primary product forming growth factor (TGFB1). Those growth factors
generates the decapeptide kispeptin-10, which is a very send their signals through receptors with tyrosine-kinase
potent agent promoting LH release (Ojeda et al. activity such as the insulin-like growth factors (IGF-1),
2006b). the fibroblast growth factor, members of the epidermal
SynCAM, an element involved in synapse formation, growth factor family, the TGFalfa and the neuregulins.
is an immunoglobulin-like adhesion molecule previously A complete discussion on the roles of these growth
recognized as a tumour suppressor molecule in lung factors has been previously presented by Garcia-Segura
cancer. It has been proposed that both SynCAM and and McCarthy (2004), Gill et al. (2004) and Ojeda et al.
kisspeptin conform a network of genes, which, besides (2006a,b).

 2009 Blackwell Verlag GmbH


e500 CA Meza-Herrera, A Gonzalez-Bulnes, RT Kridli, M Mellado, CF Arechiga-Flores, H Salinas and JM Luginbuhl

The Hierarchical Expression of Genes This regulatory mechanism is important to initiate


Modulating the Onset of Puberty: puberty in females because: (i) The hypothalamic
OCT-2, TTF-1 and EAP-1 mRNA levels of OCT-2 are increased during the
The establishment of the nerve function of GnRH prepubertal development in an independent format
neurons requires the ordered and hierarchical comple- from gonadal action, (ii) Blockage in the OCT-2
mentation of a set of genes whose primordial objective is synthesis reduces TGFalfa and delays the age at first
to establish the required conditions for a productive ovulation and (iii) Hypothalamic lesions inducing sexual
interaction between neurons and glial cells. In addition precocity activate both OCT-2 and TGFalfa in the
to residing at the centre of a complex regulatory astrocytes close to the place of injury.
network, these genes should maintain a hierarchical The second candidate gene is the TTF-1 (thyroid
structure in such network to assure that the neuronal transcription factor-1), which is required for dience-
system includes redundancy and combinatory diversity phalic morphogenesis. After birth, it is expressed in
(Ojeda et al. 2006a,b; Tena-Sempere 2006c). selected neuron populations and hypothalamic glial
Ojeda et al. (2006b) proposed a potential role of three cells, in GnRH neurons, in proopioenkephalinergic
candidate genes as controllers in the hierarchy of genic neurons, as well as in the ME. Such gene acts on these
expression when controlling the pubertal process: OCT- cellular groups to promote specific functions in each
2, TTF-1 and EAP-1. The OCT-2 proteins are more cellular type. Not only DNA arrays but also quantita-
abundant in astrocytes than in cultivated neurons, tive PCR analyses in the hypothalamic regions revealed
suggesting that the OCT-2 gene can be important in a pubertal increase in the TTF-1 expression. Elimination
the transregulation of the transcription of astroglial of the TTF-1 gene from the hypothalamic sub-networks,
genes. Both SynCAM and TGFalfa have been identified where it is commonly expressed, generated a delay in
as targets of this gene because the promoter of SynCAM puberty, a disruption in the initial oestrous cyclicity, as
and TGFalfa possess binding sites for the OCT-2 gene. well as a decreased reproductive function. Such

Fig. 1. Proposed interactions between neuroendocrine, metabolic and genomic cues signalling the onset of puberty. Nutritional state may
regulate both leptin, in a long-term fashion while insulin release may be affected in a real-time ⁄ short-term fashion. These two metabolic hormones
may act on appetite and reproductive centres in the hypothalamus, and activate ⁄ deactivate a set of genes (OCT2, TTF1 and EAP1), whose
primordial objective is to establish conditions at hypothalamic level for the establishment of nervous function of GnRH neurons. In addition, the
hypothalamic gene KiSS1, its product kisspeptin and the activation of its G-protein coupled GPR54 receptor have also been involved in the
activation of GnRH neurons. Besides that, a set of neurotransmitters and growth factors have been identified as positive or negative modulators
of glutamatergic neurons co-expressing such neurotransmitters and growth factors, whose actions activate the hypothalamic release of the
hypothalamic decapeptide GnRH. Activation of glutamatergic neurons has been involved as a critical key component in the establishment of
puberty acting via the release of the gaseous neurotransmitter nitric oxide. This neurotransmitter stimulates GnRH release through activation of
the enzyme guanylate cyclase, which in turn promotes an increase in the production of cGMP and the posterior release of GnRH in the portal
circulation, stimulating, in turn, the release of LH and FSH from the pituitary and initiating a complete activation in the communication among
the key elements involved in the onset of puberty: the hypothalamus, the hypophysis and the ovary. Recognition of additional genes and
neurological pathways modulating both the timing of pubertal onset and its tempo are warranted

 2009 Blackwell Verlag GmbH


Signalling Mechanisms Controlling Puberty e501

deficiencies were accompanied by an increase in the one another than genes of high hierarchy, yet having a
genic expression of pre-proenkephalin and by a hypo- notable redundancy and high diversity in such system.
thalamic suppression of GnRH as well as in the mRNA The existence of a network of hypothalamic genes
levels of KiSS1. located in different but interacting hierarchical levels is
The third candidate gene is EAP-1 (early at puberty), consistent with the proposed idea that the onset of
which is located as the TTF-1 gene, in chromosome 14 puberty is genetically determined and neuromodulated
in humans. The hypothalamic levels of EAP-1 increase through a polygenic strategy highly hierarchical in the
during puberty in females, suggesting its involvement in command of the genic expression (Fig. 1).
the control of puberty. The EAP-1 gene codifies a
nuclear protein which is expressed in neuronal subsets
involved in both stimulatory and inhibitory secretion of Author contributions
GnRH, such as the glutamatergic, GABAergic, pro- CA Meza-Herrera and A Gonzalez-Bulnes, designed the outline and
enkephalinergic neurons, the KiSS-1 neurons as well as the structure of the review. CA Meza-Herrera, A Gonzalez-Bulnes,
and RT Kridli made an in-depth search of information, and
the GnRH neurons. As in the case with the TTF-1 gene, substantially contributed to the discussions and the review of the
EAP-1 transactivates the promoter in those genes document. JM Luginbuhl, M Mellado, CF Arechiga-Flores and
involved in the onset of puberty (i.e. GnRH), while it H Salinas made specific topic contributions in the development of the
suppresses the expression of those related with inhibi- review.
tory routes (i.e. pre-proenkephalin). Suppression of the
hypothalamic EAP-1 expression, as that for TTF-1,
delays puberty and disrupts the oestrous cycle, high-
References
lighting the importance of EAP-1 as a gene of high Apter D, 1997: Development of the hypothalamic-pituitary-
ovarian axis. Ann N Y Acad Sci 816, 9–21.
hierarchy in the neuron-to-neuron regulation of the
Apter D, 2003: The role of leptin in female adolescente. Ann
GnRH secretion at puberty (Ojeda et al. 2006b). N Y Acad Sci 997, 64–76.
Apter D, Hermanson E, 2002: Update on female pubertal
development. Curr Opin Obstet Gynecol 14, 475–481.
Final Considerations Budak E, Fernandez-Sanchez M, Bellver J, Cervero A, Simon
The mechanisms involved in the initiation of puberty are C, Pellicer A, 2006: Interactions of the hormones leptin,
quite complex. It has been proposed that GnRH gherlin, adiponectin, resistin and PYY3-36 with the repro-
neurons are reasonably mature at birth. As the growth ductive system. Fertil Steril 85, 1563–1581.
process starts, yet, the pulsatile release of GnRH is Carbone S, Szwarcfarb B, Reynoso R, Bollero G, Ponzo O,
suppressed. Whereas the most prevailing invoked excit- Rondina D, Scacchi P, Moguilevinsky J, 2005: Leptin
stimulates LH secretion in peripubertal male rats through
atory neuronal systems consider glutamate and the
NMDA receptors. Endocr Res 31, 387–396.
kisspeptins with their GPR54 receptors for neurotrans- Cheung CC, Thornton JE, Kuijper JL, Weigle DS, Clifton
mission and neuromodulatory processes, the most DK, Steiner RA, 1997: Leptin is a metabolic gate for the
important inhibitory systems include GABAergic and onset of puberty in the female. Endocrinology 138,
opiatergic neuronal systems. As the animal approaches 855–858.
puberty, inhibitory signals diminish until they are Clarkson J, Herbison AE, 2006: Development of GABA and
removed. This generates an increase in the pulsatile glutamate signaling at the GnRH neurin in relation to
release of GnRH as a result of increasing neuronal puberty. Mol Cell Endocrinol 25, 32–38.
signals, mainly glutamatergic. The increase in the Dhandapani KM, Brann DW, 2000: The role of glutamate and
pulsatile release of GnRH results in a cascade of events nitric oxide in the reproductive neuroendocrine system.
Biochem Cell Biol 78, 165–179.
leading to an increase in the synthesis and release of LH
Ebling FJ, Schwarta ML, Foster DL, 1989: Endogenous
and FSH, as well as an increase in both steroidogenesis opioid regulation of pulsatile hormone secretion during
and gametogenesis. sexual maturation in the female sheep. Endocrinology 125,
It is well-established that GnRH pulsatility is signif- 369–383.
icantly reduced under negative energy balance observed Fernandez-Fernandez R, Martini AC, Navarro VM, Castel-
with suboptimal nutritional scenarios and disturbance lano JM, Dieguez C, Aguilar E, Pinilla L, Tena-Sempere M,
of metabolic conditions. In that respect, the role of some 2006: Novel signals for the integration of energy balance and
metabolic hormones such as leptin, ghrelin and insulin reproduction. Mol Cell Endocrinol 25, 127–132.
as body reserve markers and adequate energy level is to Foster DL, Olster DH, 1985: Effect of restricted nutrition on
be noted. Metabolic hormones act positively on the puberty in the lamb: patterns of tonic luteinizing hormone
secretion and competency of the LH surge system.
central nervous system to increase pulsatility and
Endocrinology 116, 375–381.
frequency of GnRH release. As a plethora of genes are Foster DL, Ryan KD, 1979: Endocrine mechanisms governing
activated and inactivated in the brain, the series of transition into adulthood: a marked decrease in inhibitory
complex events determining the tempo of puberty must feedback action of estradiol on tonic secretion of luteinizing
be regulated by a certain number of master genes. hormone in the lamb during puberty. Endocrinology 105,
Presently, neuroendocrine control of puberty gov- 896–904.
erned by a network of genes with a high hierarchical Foster DL, Jackson LM, Padmanablhan V, 2006: Program-
nature has been suggested (OCT-2, TTF-1 & EAP-1). ming of GnRH feedback controls timing puberty and adult
These genes are highly dominant and closely connected reproductive activity. Mol Cell Endocrinol 25, 109–119.
in the command of genic regulation. In addition, a Garcia-Segura LM, McCarthy MM, 2004: Minireview: role
of glia in neuroendocrine function. Endocrinology 145,
partial overlap of a second sub-network of subordinated
1082–1086.
genes has been proposed. They are less connected with

 2009 Blackwell Verlag GmbH


e502 CA Meza-Herrera, A Gonzalez-Bulnes, RT Kridli, M Mellado, CF Arechiga-Flores, H Salinas and JM Luginbuhl

Gill JC, Moenter SM, Tsai PS, 2004: Developmental regula- mitter amino acids during sexual maturation in female rats.
tion of gonadotropin-releasing hormone neurons by Exp Clin Endocrinol Diabetes 111, 274–277.
fibroblast growth factor signaling. Endocrinology 145, Roth C, Schricker M, Lakomek M, Strege A, Heiden I, Luft
3830–3839. H, Munzel U, Wuttke W, Jarry H, 2001: Autoregulation of
Gottsch ML, Clifton DK, Steiner RA, 2006: Kisspeptin- the gonadotropin-releasing hormone (GnRH) system during
GPR54 signaling in the neuroendocrine reproductive axis. puberty: effects of antagonistic versus agonistic GnRH
Mol Cell Endocrinol 25, 91–96. analogs in a female rat model. J Endocrinol 169, 361–371.
Huffman LJ, Inskeep EK, Goodman RL, 1987: Changes in Roth C, McCormack AL, Lomnizci A, Mungenast AE, Ojeda
episodic luteinizing hormone secretion leading to puberty in SR, 2006: Quantitative proteomics identifies a change in
the lamb. Biol Reprod 37, 755–761. glial glutamate metabolism at the time of female puberty.
Hughes IA, Kumanan M, 2006: A wider perspective on Mol Cell Endocrinol 254, 51–59.
puberty. Mol Cell Endocrinol 25, 1–7. Suttie JM, Kostyo JL, Ebling FJ, Wood RI, Bucholtz DC,
Land RB, 1978: Reproduction in young sheep: some genetic Skottner A, Adel TE, Towns RJ, Foster DL, 1991: Metabolic
and environmental sources of variation. J Reprod Fertil 53, interfases between growth and reproduction. IV. Chronic
427–436. pulsatile administration of growth hormone and the timing
Mahdi D, Khallili K, 2008: Relationship between follicle growth of puberty in sheep. Endocrinology 129, 2024–2032.
and circulating gonadotropihin levels during postnatal devel- Tena-Sempere M, 2006a: The roles of kisspeptins and
opment of sheep. Anim. Reprod. Sci. 106, 100–112. G-protein-coupled receptor 54 in pubertal development.
Mahesh VB, Brann DW, 2005: Regulatory role of excitatory Curr Opin Pediatr 18, 442–447.
amino acids in reproduction. Endocr 28, 271–280. Tena-Sempere M, 2006b: KISS-1 and reproduction: focus on
Messinis IE, 2006: From menarche to regular menstruation: its role in the metabolic regulation of fertility. Neuroendo-
endocrinological background. Ann N Y Acad Sci 1042, crinology 83, 275–281.
49–56. Tena-Sempere M, 2006c: GPR54 and kisspeptin in reproduc-
Moguilevsky JA, Wuttke W, 2001: Changes in the control of tion. Hum Reprod Update 12, 631–639.
gonadotrophin secretion by neurotransmitters during sexual Teresawa E, 2005: Role of GABA in the mechanism of the
development in rats. Exp Clin Endocrinol Diabetes 109, onset of puberty in non-human primates. Int Rev Neurobiol
188–195. 71, 113–129.
Ojeda SR, Roth C, Mungenast A, Heger S, Mastronardi C, Wankowska M, Lerrant Y, Wojcik-Gladysz A, Sterzec A,
Parent AS, Lomniczi A, Jung H, 2006a: Neuroendocrine Counis R, Polkowska J, 2002: Intracerebroventricular
mechanisms controlling female puberty: new approaches, infusion of neuropeptide Y up-regulates synthesis and
new concepts. Int J Androl 29, 286–290. accumulation of luteinizing hormone but not follicle stim-
Ojeda SR, Lomniczi A, Mastronardi C, Heger S, Roth C, ulating hormone in the pituitary cells of prepubertal female
Parent AS, Matagne V, Mungenast AE, 2006b: The neuro- lambs. J Chem Neuroanat 23, 133–142.
endocrine regulation of puberty: Is time ripe for a systems Whitlock KE, Illing N, Brideau NJ, Smith KM, Twomey S,
biology approach? Endocrinology 147, 1166–1174. 2006: Development of GnRH cells: setting the stage for
Parent AS, Matagne V, Bourguignon JP, 2005: Control of puberty. Mol Cell Endocrinol 25, 39–50.
puberty by excitatory aminoacid neurourotransmitters and Wojcik-Gladysz A, Polkowska J, 2006: Neuropeptide Y, a
its clinical applications. Endocr 28, 281–286. neuromodulatory link between nutrition and reproduction
Polkowska J, Lerrant Y, Wankowska M, Wojcik-Gladysz A, at the central nervous system level. Reprod Biol 6, 21–28.
Starzec A, Counis R, 2003: The effect of dietary protein Zamorano PL, Mahesh VB, De Sevilla L, Brann DW, 1998:
restriction on the secretion of LH and FSH in pre-pubertal Excitatory amino acids receptors and puberty. Steroids 63,
female lambs. Anim Reprod Sci 76, 53–56. 268–270.
Ponzo OJ, Reynoso R, Rimoldi G, Rondina D, Szwarcfarb B, Zapatero-Caballero H, Sanchez-Franco F, Fernandez-Mendez
Carbone S, Scacchi P, Moguilevsky JA, 2005: Leptin C, Garcia-Sanrutos M, Botella-Cubells LM, Fernandez-
stimulates the reproductive male axis in rats during sexual Vazquez G, 2004: Gonadotropin-releasing hormone expres-
maturation by acting on hypothalamic excitatory amino sion during pubertal development in rats. Biol Reprod 70,
acids. Exp Clin Endocrinol Diabetes 113, 135–138. 348–355.
Ramirez DV, McCann SM, 1963: Comparison of the regula- Zarazaga LA, Guzman JL, Dominguez C, Perez MC, Prieto
tion of luteinizing hormone (LH) secretion in immature and R, Sanchez J, 2009: Nutrition level and season of birth do
adult rats. Endocrinology 72, 452–464. not modify puberty of Papoya goat kids. Animal 3, 79–86.
Rawlings NC, Evans AC, Hoaramooz A, Bartlewski PM,
2003: Antral follicle growth and endocrine changes in
prepuberal cattle, sheep and goats. Anim Reprod Sci 78, Submitted: 15 Sep 2008
259–270.
Reynoso R, Ponzo OJ, Szwarcfarb B, Rondina D, Carbone S, Author’s address (for correspondence): Dr. Cesar A. Meza-Herrera,
Rinoldi G, Scacchi P, Moguilevisky JA, 2003: Effect of Galeana 585 Poniente, Colonia Centro, Lerdo, Durango, Mexico
leptin on hypothalamic release of GnRH and neurotrans- 35150. E-mail: cmeza2000@hotmail.com

 2009 Blackwell Verlag GmbH

Potrebbero piacerti anche