You are on page 1of 8

Hormones and Behavior 64 (2013) 187–194

Contents lists available at ScienceDirect

Hormones and Behavior


journal homepage: www.elsevier.com/locate/yhbeh

Review

Metabolic control of puberty: Roles of leptin and kisspeptins


Miguel A. Sanchez-Garrido, Manuel Tena-Sempere ⁎
Department of Cell Biology, Physiology and Immunology, University of Córdoba, CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
Instituto Maimónides de Investigaciones Biomédicas (IMIBIC)/Hospital Universitario Reina Sofía, 14004 Córdoba, Spain

a r t i c l e i n f o a b s t r a c t

Keywords: This article is part of a Special Issue “Puberty and Adolescence”.


Puberty
Leptin Reproduction is an energy-demanding function. Accordingly, puberty is metabolically gated, as a means to
Kisspeptins prevent fertility in conditions of energy insufficiency. In addition, obesity has been shown to impact the timing
Energy homeostasis of puberty and may be among the causes for the earlier trends of pubertal age reported in various countries.
Metabolism
The metabolic control of puberty in such a spectrum of situations, ranging from energy deficit to extreme
overweight, is the result of the concerted action of different peripheral hormones and central transmitters
that sense the metabolic state of the organism and transmit this information to the various elements of the
reproductive axis, mainly the GnRH neurons. Among the peripheral signals involved, the adipose hormone,
leptin, is known to play an essential role in the regulation of puberty, especially in females. Yet, although
it is clear that the effects of leptin on puberty onset are predominantly permissive and mainly conducted at
central (hypothalamic) levels, the primary sites and mechanisms of action of leptin within the reproductive
brain remain unsolved. In this context, neurons expressing kisspeptins, the products of the Kiss1 gene
that have emerged recently as essential upstream regulators of GnRH neurons, operate as key sensors of
the metabolic state and funnel of the reproductive effects of leptin. Yet, much debate has arisen recently on
whether the putative actions of leptin on the Kiss1 system are actually indirect and/or may primarily target
Kiss1-independent pathways, such as those originating from the ventral premmamilary nucleus. Moreover,
evidence has been presented for extra-hypothalamic or peripheral actions of leptin, including direct gonadal
effects, which may contribute to the metabolic control of reproduction in extreme body weight conditions.
In this work, we will critically review the experimental evidence supporting a role of leptin, kisspeptin and
putatively related pathways in the concerted control of puberty by energy balance and metabolism.
© 2013 Elsevier Inc. All rights reserved.

Contents

Introduction: Puberty is metabolically gated . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 187


Leptin as major determinant of mammalian puberty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188
Kisspeptins as essential gatekeepers of mammalian puberty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 189
Kisspeptins and the metabolic control of puberty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
Interplay of leptin and kisspeptins in the metabolic control of puberty: direct or indirect? . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
Other sites of action of leptin in the metabolic control of puberty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
Final remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
Disclosure statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193

Introduction: Puberty is metabolically gated

⁎ Corresponding author at: Department of Cell Biology, Physiology and Immunology,


Faculty of Medicine, University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba,
Puberty is a complex developmental event within the continuum of
Spain. Fax: +34 957 218288. changes leading to sexual and somatic maturation of the individual, dur-
E-mail address: fi1tesem@uco.es (M. Tena-Sempere). ing which reproductive competence is achieved and multiple growth,

0018-506X/$ – see front matter © 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.yhbeh.2013.01.014
188 M.A. Sanchez-Garrido, M. Tena-Sempere / Hormones and Behavior 64 (2013) 187–194

behavioral and psychological changes occur (Ojeda and Skinner, 2006; gonadotropin levels in female rats (True et al., 2011). Yet, the physiolog-
Ojeda et al., 2010; Parent et al., 2003). The timing of puberty is precisely ical relevance of this observation needs further evaluation.
controlled by a plethora of endogenous signals and environmental cues While detailed description of the biological features of leptin is be-
that impinge at different levels of the hypothalamic–pituitary–gonadal yond the scope of this review, it is important to stress that leptin operates
(HPG or reproductive) axis (Parent et al., 2003). This phenomenon, as a major signal of energy sufficiency, thus allowing different physiolog-
which ensures that the tempo of puberty is adjusted to the internal ical systems (not only those controlling body weight) to sense the mag-
and external conditions of the body, is also responsible for perturbations nitude of energy reserves. In fact, leptin is known to inform quite diverse
of its timing when such conditions are not optimal. Accordingly, puberty axes, from the immune system to the growth axis, of the existence of ad-
has been regarded not only as a fundamental developmental event, but equate energy stores to face these various body functions (Casanueva
also as sensor of the proper interplay between genes and environment and Dieguez, 1999). Accordingly, conditions of leptin insufficiency, if per-
along development (Tena-Sempere, 2010a). Hence, the recent trends sistent in time, are coupled to perturbed function of numerous systems.
for the change (mainly, advance) in the age of puberty, as reported in This is the case also of the reproductive axis, as illustrated in various an-
different populations, may be a sentinel for the deterioration of repro- imal models and human pathologies associated with low or null leptin
ductive and/or environmental health and, as such, have raised concerns levels, which are usually linked to a delay or absence of puberty onset
in the medical community and lay-public worldwide (Aksglaede et al., and perturbed fertility (Roa et al., 2010).
2009a, 2009b). Although the important roles of leptin in the control of puberty were
Among its different regulators, it is well established that metabolic well settled soon after its identification in the mid 90's, the precise na-
conditions and the amount of energy reserves of the organism play an ture of leptin effects on pubertal control was the subject of some debate.
essential role in the modulation of the timing of puberty (Fernandez- Thus, initial work in rodents suggested that leptin administration might
Fernandez et al., 2006; Martos-Moreno et al., 2010). This makes good actually induce or advance the onset of female puberty, therefore im-
biological sense, especially in the female, so that the reproductive plying a net stimulatory action (Ahima et al., 1997; Chehab et al.,
capacity, which implies the potential metabolic drainage of pregnancy 1997). This possibility was considered relevant not only from a physio-
and lactation, is only acquired when threshold energy stores and optimal logic standpoint but also from a therapeutic perspective; if leptin was to
metabolic conditions are achieved. In any event, evidence for the influ- be used to treat forms of morbid obesity of early origin (especially in
ence of metabolic signals on puberty and fertility is also found in the leptin-null individuals), any potential action of leptin as trigger of
male (Castellano et al., 2009b), in which energy demands for proper re- precocious puberty should be regarded as an important side-effect.
production are not so evident but probably required as well (e.g., for ter- However, pharmacological studies in humans and rodents with leptin
ritoriality and partner selection) (Elias, 2012; Elias and Purohit, 2013). deficiency clearly documented that, while leptin is indispensable for
Moreover, not only conditions of metabolic distress associated with puberty to proceed, leptin alone cannot trigger early puberty, therefore
energy insufficiency but also with its excess, such as morbid obesity, are supporting a major permissive role of this hormone in the metabolic
linked to reproductive perturbations, such as precocious puberty in the gating of pubertal maturation (Barash et al., 1996; Castellano et al.,
female (Castellano et al., 2011) and trends for hypogonadism of central 2009b; Cheung et al., 1997; Roa et al., 2010). This permissive function
origin in the male (Tajar et al., 2010). Moreover, early forms of nutritional has been better characterized in females, while leptin's role in male pu-
or metabolic challenge, such as under- or over-nutrition taking place dur- berty remains less clear. In fact, circulating leptin has shown to rise dur-
ing the fetal and neonatal life, have been also shown to alter the timing of ing the pubertal transition in girls (Garcia-Mayor et al., 1997), while
puberty in rodents and humans (Caron et al., 2012; Castellano et al., 2011; puberty onset in boys and male monkeys does not seem to require a
Ibanez and de Zegher, 2006). Altogether, these findings illustrate not only similar increase in leptin levels (Garcia-Mayor et al., 1997; Plant and
the fact that the metabolic status has a clear impact on the timing of Durrant, 1997).
puberty but also that the mechanisms underlying this phenomenon are Another issue that has remained contentious for years is how and
complex and likely involve different regulatory pathways (Roa and where leptin conducts its reproductive actions (Elias, 2012). In one
Tena-Sempere, 2010; Roa et al., 2010). hand, solid evidence has demonstrated that the permissive actions of
leptin are conducted primary at central (hypothalamic) levels, where
leptin is able to ultimately modulate the function of the GnRH neuronal
Leptin as major determinant of mammalian puberty system (Cunningham et al., 1999). Notwithstanding this, evidence for
additional actions of leptin at other levels of the HPG axis has been
Despite historical intuitions about the metabolic gating of puberty presented (Caprio et al., 2001; Tena-Sempere, 2007). Indeed, at high
that existed since old ages, the basis for such a close relationship between concentrations, as those expected in obesity, leptin can directly inhibit
the magnitude of energy (fat) stores and pubertal timing became scien- some gonadal functions, such as sex steroid secretion (Caprio et al.,
tifically formulated only in the 1960's and 70's. This was mainly due to 2001; Tena-Sempere et al., 1999). Although this was initially considered
the pioneering work of Kennedy and Mitra in rodents, and Frisch and counterintuitive given its primarily permissive/positive roles on the
co-workers in humans (Fernandez-Fernandez et al., 2006; Pralong, HPG axis, such inhibitory actions of leptin may explain part of the
2010). This work resulted in the so-called critical fat mass hypothesis hypogonadal state frequently observed in morbid obese patients
that, although partially challenged later, set the scene for the search of (Caprio et al., 2001; Tena-Sempere and Barreiro, 2002). On the other
the factor(s) responsible for the integral control of energy metabolism, hand, even assuming a primary permissive action at central levels, lep-
puberty onset and fertility. The identification in 1994 of the adipose tin effects on the GnRH system appear to be conducted indirectly, via
hormone, leptin, as an essential signal in the control of body weight ho- leptin-sensitive afferents that project to GnRH neurons, which are ap-
meostasis, whose levels in circulation are proportional to the size of fat parently devoid of functional leptin receptors. This contention, which
stores, led to specific analyses of the reproductive roles of this hormone. was originally questioned by the detection of leptin receptors in immor-
These studies settled the contention that leptin plays a central function in talized GT1-7 cells, which are parentally derived from GnRH neurons, is
the metabolic control of puberty and fertility (Ahima et al., 2000; now solidly supported by data from brain expression studies in rodents
Casanueva and Dieguez, 1999; Cheung et al., 1997; Fernandez- and primates (Finn et al., 1998; Hakansson et al., 1998), and recent
Fernandez et al., 2006; Tena-Sempere, 2007). Nonetheless, recent exper- functional genomic approaches that showed that congenital elimina-
imental evidence has cast doubts on the actual roles of leptin in some tion of putative leptin receptors from GnRH neurons was compatible
particular aspects of the metabolic control of the gonadotropic axis. For with preserved puberty or fertility (Quennell et al., 2009). Although
instance, normalization of mean leptin concentrations in conditions the occurrence of developmental compensation cannot be excluded in
of negative energy balance, such as acute fasting, failed to restore the later model, the experimental evidence available strongly suggests
M.A. Sanchez-Garrido, M. Tena-Sempere / Hormones and Behavior 64 (2013) 187–194 189

that the physiological actions of leptin on GnRH secretion and hence on described (Clarkson et al., 2009; Pinilla et al., 2012). The importance
puberty onset are conducted indirectly (Quennell et al., 2009). This as- of kisspeptins in reproductive function is not only illustrated by the
sumption has led to the search for the primary targets of leptin within HH phenotypes of humans and mice with inactivating mutations of
the reproductive brain. Recent developments in this area will be the Kiss1 or Gpr54 genes, but also by solid neuroanatomical, electro-
discussed in the following sections. physiological, pharmacological and hormonal data. Collectively, these
As final note to this section, the molecular mediators and signaling data have documented the involvement of the so-called Kiss1 system
pathways for the central reproductive actions of leptin have been also in the regulation of virtually all aspects of reproductive maturation
the subject of active investigation. Based on the known properties of and function, from brain sex differentiation to the neuroendocrine con-
functional leptin receptors (LepR-b or Ob-Rb), the role of leptin- trol of ovulation and sex steroid feedback.
induced STAT3 signaling has been studied (Elias and Purohit, 2013). Ge- In the above scenario, the possibility that the Kiss1 system may play a
netic models have demonstrated that, while elimination of STAT3 signal- physiological role in the control of puberty has drawn considerable
ing from the brain results in lack of pubertal maturation and infertility, attention and has been the subject of thorough analyses in numerous
selective ablation of STAT3 from LepR-expressing neurons (hence, neu- (mammalian and nonmammalian) species (Pineda et al., 2010;
rons responsive to the actions of leptin) is compatible with proper devel- Tena-Sempere, 2010b). Indeed, the findings of the lack of pubertal mat-
opment of reproductive organs and cyclic ovarian function in mice (Bates urations in patients or mice with null mutations in Gpr54 or Kiss1 genes
et al., 2003). This would suggest that STAT3-independent pathways are suggest a prominent, indispensable role of kisspeptin signaling in the
recruited by leptin in the brain to conduct, at least part of, their effects control of puberty. Admittedly, however, the fact that congenital inacti-
on the reproductive axis. These pathways may involve PI3K, a molecular vation of Gpr54 or Kiss1 perturbs puberty does not necessarily imply an
complex that is known to mediate part of the central effects of leptin in activational role of the Kiss1 system in puberty itself, as the above pheno-
terms of metabolic control (Elias and Purohit, 2013). In this sense, selec- type (lack of puberty) may derive from the alteration of early develop-
tive elimination of key catalytic subunits of PI3K from LepR-expressing mental phenomena in the absence of kisspeptin signaling. In any event,
cells prevents leptin effects on key cellular functions in POMC and the available experimental evidence does suggest that kisspeptins
AgRP neurons in the arcuate nucleus (ARC) (Al-Qassab et al., 2009), as participate in the activational program responsible for puberty, as docu-
well as in the ventral premmamilary nucleus (PMV) (Williams et al., mented by a combination of neuroanatomical and functional analyses,
2011). However, whether blockade of leptin-induced activation of PI3K conducted mainly in rodent species, especially in the female (Roa and
has deleterious effects on puberty onset and reproductive function has Tena-Sempere, 2010; Tena-Sempere, 2010b).
not been reported to date. Finally, central signaling pathways involving Initial expression studies documented an increase in the hypotha-
the mammalian target of rapamycin (mTOR) have been also implicated lamic expression of the Kiss1 gene during pubertal maturation in the
in mediating not only the metabolic actions of leptin (e.g., in terms of rat and monkey, therefore suggesting a rise of the kisspeptin tone at
food intake control), but also the permissive effects of leptin on puberty the hypothalamus during puberty (Navarro et al., 2004a; Shahab et al.,
onset (Roa and Tena-Sempere, 2010). Thus, central blockade of mTOR 2005). Of note, pharmacological analyses demonstrated the functional
blunted the ability of leptin to rescue puberty in female rats subjected relevance of such a phenomenon, since repeated administration of
to chronic subnutrition (Roa et al., 2009). Despite this compelling evi- kisspeptin-10 was sufficient to advance the occurrence of different indi-
dence, whether these effects of mTOR are conducted directly in Kiss1 ces of puberty onset in immature female rats (Navarro et al., 2004b).
neurons or in upstream afferents, sensitive to the regulatory actions of Likewise, repeated kisspeptin administration to juvenile monkeys
leptin, remains unknown. evoked a pattern of GnRH secretion similar to that of (post)pubertal an-
imals (Plant et al., 2006). In addition, rodent studies have shown that
Kisspeptins as essential gatekeepers of mammalian puberty during puberty there is an increase in the sensitivity to the stimulatory
effects of kisspeptin in terms of GnRH/LH responses, as described in rats
Our knowledge of the neurobiological basis of puberty in general, and mice (Castellano et al., 2006a; Han et al., 2005). Likewise, in female
and of the mechanisms for its metabolic regulation in particular, has mice, the number of GnRH neurons expressing Gpr54 increases before
substantially enlarged in recent years. Probably, the most important de- puberty (Herbison et al., 2010), as does the Gpr54 signaling efficiency
velopment in this front was the recognition of the essential roles of (Han et al., 2005). Importantly, Gpr54 seems to be less prone to
kisspeptins in the central control of different aspects of reproductive desensitization to the stimulatory effects of continuous kisspeptin stim-
function, including puberty onset (Clarkson et al., 2010; Oakley et al., ulation during the pubertal transition in female rats (Roa et al., 2008b).
2009; Pinilla et al., 2012; Roa et al., 2008a). Due to the astonishingly This phenomenon may help to explain why at this period the rise of the
rapid progress of this field, a detailed recapitulation of the major fea- endogenous kisspeptin tone is compatible with the progressive activa-
tures of the physiology of kisspeptins is beyond the scope of this review tion of the gonadotropic axis, without any sign of receptor desensitiza-
and can be found elsewhere (Pinilla et al., 2012). Nonetheless, in order tion. Finally, detailed neuro-anatomical studies in female mice have
to introduce later sections of this review, a brief account of the proposed demonstrated that during puberty there is a sharp rise in the number
roles of kisspeptins as putative gatekeepers of puberty is presented of kisspeptin neurons, as documented by immunohistochemical
here. analyses (Clarkson et al., 2010). In addition, the number of projections/
Kisspeptins (Kp), encoded by the Kiss1 gene, are a family of appositions of Kiss1 neurons to GnRH neurons also increases during pu-
structurally-related peptides with the ability to activate the G protein- berty. This phenomenon seems to require, at least in female rodents, the
coupled receptor, Gpr54 – or Kiss1R – (Oakley et al., 2009; Pinilla et permissive action of some degree of estrogenic input of ovarian origin
al., 2012; Roa et al., 2008a). Departing from the seminal finding of (Clarkson et al., 2010). This would imply that the early stages of ovarian
forms of hypogonadotropic hypogonadism (HH) in patients with activation during puberty take place before full activation of the hypo-
inactivating mutations of GPR54 (de Roux et al., 2003; Seminara et al., thalamic Kiss1 system, which would rather operate as an essential am-
2003), a growing number of studies, conducted in different model spe- plifier of the GnRH neurosecretory activity along pubertal maturation.
cies as well as in humans, have now set the contention that kisspeptins Altogether, the above experimental evidence demonstrates that
are key transmitters in the reproductive brain. These originate from dis- during puberty the hypothalamic Kiss1 system undergoes an extensive
crete neuronal populations in the hypothalamus and are able to stimu- and complex activational program that seems to be essential for proper
late the secretory activity of GnRH neurons. The neuro-anatomy of such pubertal timing. In good agreement, studies from our group have dem-
populations has been well characterized in rodents, where two major onstrated that pharmacological blockade of Gpr54 is sufficient to delay
sets of Kiss1 neurons, located in the arcuate nucleus (ARC) and the puberty onset in female rats (Pineda et al., 2010). Yet, in spite of the
rostral periventricular area of the 3rd ventricle (RP3V), have been above compelling evidence, recent data have challenged the view that
190 M.A. Sanchez-Garrido, M. Tena-Sempere / Hormones and Behavior 64 (2013) 187–194

intact kisspeptin signaling is indispensable for puberty onset in rodents, lipopolysaccharide S (LPS) injection, not only provoked metabolic stress
as it has been shown, by the use of elegant genetically-modified mouse and hypogonadotropism but also reduced hypothalamic Kiss1 mRNA ex-
models, that fertility can be apparently achieved even after the congen- pression in male rats (Castellano et al., 2010b).
ital ablation of Kiss1 neurons in female mice (Mayer and Boehm, 2011). Evidence for the metabolic control of the Kiss1 system does not only
Quite expectedly, this set of data has drawn enormous interest, as it ar- come from studies in rodent species under negative energy balance
gues against the consensus notion that kisspeptins are essential gate- conditions. A recent study also documented that hypogonadotropic
keepers of puberty. Some reconciliation with previous literature can lean ewes display lower Kiss1 mRNA levels in the ARC than their corre-
be obtained, however, by careful analysis of those results, as the same sponding normal weight counterparts (Backholer et al., 2010). This
publication clearly showed that timed elimination of Kiss1 neurons dur- finding suggests that a reduced kisspeptin tone in the hypothalamus
ing the early juvenile period (i.e., before puberty onset) did prevent is common to conditions of energy deficit in various mammalian
normal pubertal maturation and caused infertility (Mayer and Boehm, species. The functional relevance of this phenomenon is confirmed by
2011). The most plausible explanation for these two, apparently contra- experimental studies in rodents, where repeated administration of
dictory sets of data is that a residual group of Kiss1 neurons remaining kisspeptin in models of negative energy balance was sufficient to ame-
after congenital ablation would be sufficient to drive puberty onset, liorate the state of reproductive failure. This was the case in pubertal fe-
due to developmental compensatory mechanisms. In fact, such a devel- male rats submitted to chronic sub-nutrition (30% reduction in daily
opmental compensation has been previously documented in models of calorie intake from day 23 onwards that prevented normal puberty
early, nearly complete depletion of GnRH neurons (Herbison et al., onset), in which repeated central injections of kisspeptin-10 rescued
2008). In contrast, depletion of Kiss1 neurons at later stages of develop- vaginal opening as an external sign of puberty, in a notable proportion
ment would not cause similar compensatory adaptation and, hence, of cases and evoked very potent gonadotropic and estrogenic responses,
lowering of the endogenous kisspeptin tone would prevent the normal despite the persistent reduction in body weights (Castellano et al.,
timing of puberty. All in all, genomic, expression and functional data 2005). Analogous observations have been obtained in other models of
jointly point out that, despite the potential existence of fail-safe mech- metabolic stress in adulthood, such as uncontrolled diabetic male and
anisms to compensate for the early loss of kisspeptin signaling, Kiss1 female rats. In these animals, kisspeptin-10 induced robust gonadotrop-
neurons do play an indispensable role in the activational mechanisms ic responses in spite of the lowering of the prevailing gonadotropin
leading to puberty onset. levels due to the diabetic state (Castellano et al., 2009a, 2006b), and
its repeated administration substantially improved different indices of
Kisspeptins and the metabolic control of puberty gonadotropic function (e.g., testicular and prostate weights) and testos-
terone levels (Castellano et al., 2006b). All in all, these data indirectly
Considering the important roles of kisspeptin signaling in the con- support the involvement of suppressed kisspeptin tone in the genera-
trol of puberty and reproduction, and the relevance of energy homeo- tion of the state of hypo-gonadotropism associated with conditions
stasis and metabolic cues in pubertal regulation, especially in females, of energy insufficiency, and emphasize the functional relevance of cen-
it is not surprising that the possibility of specific functions of Kiss1 tral kisspeptin pathways in the metabolic control of puberty and
neurons in the metabolic control of puberty and the gonadotropic reproduction.
axis have been thoroughly analyzed in recent years using a number In addition, very recent studies in models of metabolic stress in pu-
of experimental paradigms. As a potential call of caution, the majority bertal female rats have demonstrated that the hypothalamic neurokinin
of those studies have focused on the consequences, in terms of Kiss1 B (NKB) system is also sensitive to conditions of negative energy bal-
expression and/or function, of conditions of negative energy balance ance. NKB is co-expressed with kisspeptins in ARC Kiss1 neurons and
that causes some degree of pubertal suppression and/or hypogonadism, defects of NKB signaling are incompatible with normal puberty onset
and hence have addressed rather extreme experimental conditions and fertility, as is also the case for inactivation of kisspeptin signaling
(Castellano et al., 2010a). Notwithstanding this potential limitation, (Navarro and Tena-Sempere, 2012). For instance, by using acute fasting
the available evidence does suggest that Kiss1 neurons are sensitive to as a model of metabolic stress, it has been recently shown that condi-
the metabolic/energy state of the organism and likely participate in tions of energy deficit coupled to hypogonadotropism do not only sup-
the modulation of the reproductive axis by metabolic cues, at least press Kiss1 mRNA expression in the ARC and RP3V, but inhibited also
under certain circumstances. As side comment, most of the supportive the hypothalamic expression of the genes encoding NKB and its recep-
data has been obtained in adult models; hence, extrapolation of adult tor (NK3R) in pubertal female rats (Navarro et al., 2012). Importantly,
results to puberty must be made with caution. the delay of female puberty caused by 30% under-nutrition during the
A wealth of experimental data has now documented that extreme juvenile period was partially prevented by repeated administration of
conditions of negative energy balance induce a suppression of the hypo- an NKB agonist (Navarro et al., 2012). This response is very similar to
thalamic Kiss1 system. This has been well characterized, at the mRNA that obtained after kisspeptin-10 injection in pubertal female rats sub-
and protein levels, in pubertal rats, in which fasting decreased hypotha- mitted to similar degree of sub-nutrition (see above and Castellano et
lamic Kiss1 expression and kisspeptin immunoreactivity, in association al., 2005). As a whole, these observations support the view that NKB sig-
with a significant lowering of circulating LH levels (Castellano et al., naling may cooperate with kisspeptins in the metabolic control of pu-
2005, 2010a). A roughly similar response to fasting has been also berty, at least in the female (Navarro et al., 2012). In good agreement,
reported in adult female rats (Brown et al., 2008; Kalamatianos et al., 50% caloric restriction in adult rats has been shown to suppress NKB
2008) and adult male mice, in which a decrease in hypothalamic Kiss1 mRNA expression in the ARC (True et al., 2011).
mRNA was detectable as soon as 12-h after beginning of food deprivation As final note in this section, the impact upon the hypothalamic Kiss1
(Luque et al., 2007). Other conditions of negative energy balance and system of other forms of metabolic stress due to persistent energy ex-
metabolic distress causing suppression of gonadotropic function display cess, such as in morbid obesity, remains less well characterized. Yet,
also analogous inhibitory responses in terms of hypothalamic Kiss1 the translational relevance of such a putative influence is high, due to
expression. For instance, 50% caloric restriction induced a significant (a) the escalating prevalence of obesity worldwide; and (b) the fre-
suppression of Kiss1 mRNA expression in the ARC and RP3V in adult quent reproductive co-morbidities associated to obesity, which include
female rats (True et al., 2011). Similarly, uncontrolled experimental dia- male and female sub/infertility, as well as altered timing of puberty
betes in rats, which is associated to hypogonadotropic hypogonadism, (Ahmed et al., 2009; Biro and Wien, 2010; Loret de Mola, 2009;
causes also a lowering of Kiss1 mRNA levels at the hypothalamus in Rachon and Teede, 2010). Although the available data in this front are
both males and females (Castellano et al., 2006b, 2009a). Likewise, still limited, there is indirect evidence supporting the possibility that
acute inflammatory challenges, such as those induced by bacterial obesity has an impact on the Kiss1 system. First, while initial studies
M.A. Sanchez-Garrido, M. Tena-Sempere / Hormones and Behavior 64 (2013) 187–194 191

in diet-induced obese male mice of the C57/B6 strain did not show sig- physiological levels would be insufficient to normalize Kiss1 expression
nificant changes in relative Kiss1 mRNA levels in whole hypothalamic and LH levels in situations of energy deficit (True et al., 2011). Whether
fragments (Luque et al., 2007), these animals had dramatically the above protocols of leptin replacement fully mimicked physiological
suppressed testosterone levels, and hence their hypothalamic Kiss1 ex- conditions and, if so, whether the same phenomenon applies to puberty
pression should be considered inappropriately low (Luque et al., 2007). is yet to be defined.
In good agreement, subsequent analyses in other mouse strains, such as In spite of the compelling evidence summarized above, recent ex-
the DBA/2J, more prone to develop obesity-induced infertility demon- perimental data have questioned the consensus of a predominant direct
strated a significant suppression of kisspeptin expression in the ARC mode of action of leptin on Kiss1 neurons. Thus, by the use of functional
and RP3V of females in conditions of diet-induced obesity (Quennell genomics, Donato and co-workers have recently reported that congen-
et al., 2011). On the other hand, we have recently shown that early ital selective elimination of LepR from Kiss1-expressing cells seems to
onset overweight in female rats, which is linked to precocious occur- be compatible with preserved puberty onset and fertility (Donato et
rence of puberty, is associated with detectable changes in the hypotha- al., 2011). It must be stressed, however, that the Kiss1 gene appears to
lamic expression of Kiss1 and a trend for increased numbers of be widely expressed in other brain and other peripheral tissues at
kisspeptin fibers in the RP3V immediately prior the onset of puberty early stages of development. Thus, it is possible that the selectivity of
(Castellano et al., 2011). The latter observations may be of help to ex- LepR elimination in postnatal Kiss1 neurons in the hypothalamus is
plain the underlying mechanisms for the association between child compromised in that model. Moreover, it is also plausible that congen-
obesity and earlier puberty. In addition, they stress the importance of ital ablation of LepR from Kiss1 cells may have caused compensatory
early developmental/metabolic challenges in the timing of puberty changes that would obscure the impact of elimination of leptin signal-
onset, which manifests not only in conditions of overfeeding but also ing in this neuronal population. In any event, the possibility of a pre-
of early malnutrition. In fact, our studies in undernourished rats during dominantly indirect mode of action of leptin in Kiss1 neurons has
the postnatal period evidenced a suppression of the hypothalamic Kiss1 been reinforced by a recent independent study mapping the distribu-
system coupled to delayed puberty (Castellano et al., 2011). This con- tion of the functional LepRb in various hypothalamic areas. That study
tention has been recently confirmed in mice (Caron et al., 2012). could not find evidence for expression of functional LepR in GnRH or
Kiss1 neurons, except for a small population of Kiss1 cells in the ARC
Interplay of leptin and kisspeptins in the metabolic control of (Louis et al., 2011). Of note, in the same work, an uncharacterized pop-
puberty: direct or indirect? ulation of LepRb-expressing neurons was identified in the ARC and
RP3V, in the close vicinity of Kiss1 neurons (Louis et al., 2011). All in
Considering the individual roles of leptin and kisspeptin pathways all, these data suggest that a (substantial) part of the effects of leptin
in the metabolic control of puberty, it is not surprising that it was ini- on the hypothalamic expression of Kiss1/kisspeptin may be conducted
tially hypothesized that one of the major mechanisms whereby leptin indirectly and transmitted via intermediate pathways of as yet un-
influences the timing of puberty is via its ability to regulate the hypo- known nature (Louis et al., 2011). The physiological significance of
thalamic Kiss1 system. Such a potential regulatory mechanism, such an intricate, indirect mode of regulation of Kiss1/GnRH neurons
which has been experimentally tested using different paradigms, is by leptin is yet to be deciphered.
supported by a wealth of complementary data, which can be summa-
rized as follows: (a) the mRNA encoding the functional leptin recep- Other sites of action of leptin in the metabolic control of puberty
tor, namely LepR or Ob-Rb, has been detected in ARC Kiss1 neurons in
the mouse and sheep (Backholer et al., 2010; Smith et al., 2006), al- Despite the central roles of kisspeptin pathways in the control of pu-
though only a relatively modest sub-population of Kiss1 neurons berty and their interplay with leptin (see above), experimental evi-
(ranging from ~ 40 to b10%, depending on the studies) does apparent- dence has been presented in recent years to document that the
ly express LepR in the mouse (Cravo et al., 2011); (b) leptin can di- reproductive effects of leptin are, at least partially, mediated by primary
rectly excite ARC Kiss1 neurons in the guinea pig by activation of actions conducted in hypothalamic areas that do not apparently contain
TRPC channels (Qiu et al., 2011); (c) models of leptin deficiency dis- Kiss1 neurons. Importantly, this evidence does not refute a critical role
play lower Kiss1 mRNA expression, especially in the ARC (Quennell of kisspeptins in the metabolic control of reproduction, neither does it
et al., 2011; Smith et al., 2006), and decreased numbers of kisspeptin rule out the possibility of interactions between these alternative circuits
neurons in the RP3V (Quennell et al., 2011); and (d) leptin adminis- and Kiss1 pathways. In fact, the latter possibility has gained momentum
tration in rodent models of leptin deficiency, due to genetic inactiva- given the assumption of the indirect mode of action of leptin upon Kiss1
tion (as the ob/ob) or metabolic stress (as the uncontrolled diabetic neurons. Nonetheless, recognition and detailed characterization of such
rats), was capable to increase the hypothalamic expression of Kiss1 alternative leptin-sensitive circuits are essential to obtain a complete
gene (Backholer et al., 2010; Castellano et al., 2006b; Luque et al., view of how leptin modulates the reproductive brain.
2007; Smith et al., 2006). The later phenomenon has been also observed A paradigmatic example of a target area for the reproductive effects
after leptin injection in a sheep model of central hypogonadism of leptin outside Kiss1-enriched hypothalamic nuclei is the PMV. In-
associated to leanness, as well as in murine and human neuronal cell deed, this nucleus had been pointed out as node for the transmission
lines endogenously expressing Kiss1 following leptin stimulation of environmental cues to the reproductive centers well before the iden-
(Backholer et al., 2010; Luque et al., 2007; Morelli et al., 2008). Altogeth- tification of the roles of leptin and kisspeptins (Elias and Purohit, 2013).
er, these data strongly suggested a direct mode of action of leptin in the Recently, the PMV has emerged as an important primary target and
control of Kiss1 neurons; a view that has dominated the field until very transmitting hypothalamic area for the permissive/stimulatory effects
recently (see below). It is stressed, though, that most of the above of leptin on puberty and fertility (Donato et al., 2009, 2011). The main
experimental evidence has not been obtained in specific models of pu- experimental evidence supporting this role can be summarized as fol-
berty. Yet, our data correlating the timing of puberty, circulating levels lows: (a) a subset of neurons in the PMV express LepR and are
of leptin and hypothalamic expression of the Kiss1 system indirectly depolarized (excited) after leptin stimulation; (b) bilateral excitotoxic
suggest a close association between higher leptin levels, increased lesions of PMV lowered LH and estradiol levels, and prevented the
Kiss1/kisspeptin expression and earlier puberty onset (or vice versa) positive effects of leptin on the gonadotropic axis in fasted animals;
in the female rat (Castellano et al., 2011). As a note of caution, recent and (c) rescue of expression of LepR selectively in PMV neurons in a
data from adult models of negative energy balance suggested that LepR-null background was sufficient to induce puberty and improve
while supra-physiological levels of leptin can prevent the state of fertility, selectively in female mice (Elias, 2012; Elias and Purohit,
hypogonadotropism associated to fasting, leptin replacement to 2013). Intriguingly, a similar procedure of LepR rescue in male mice
192 M.A. Sanchez-Garrido, M. Tena-Sempere / Hormones and Behavior 64 (2013) 187–194

was ineffective in terms of pubertal restoration, thus suggesting a Among those, puberty is influenced by the metabolic and energy status
marked sexual dimorphism in the role of PMV in mediating the repro- of the organism, so that deregulation of energy homeostasis and metab-
ductive actions of leptin. As a call of caution, only a fraction of PMV olism affects puberty, while puberty itself, and its deviations, may have
neurons do express LepR, so it remains possible that part of the an impact of different metabolic indices and body weight. Such a tight
reproductive phenotypes of the excitotoxic lesions described above connection is conveyed by the concerted actions of numerous neurohor-
may derive from the damage of leptin-independent pathways. In any monal signals. In this work, we have highlighted the important contribu-
event, the above observations would suggest the existence of Kiss1- tion of the adipose hormone, leptin, and the central (hypothalamic)
independent circuits for the transmission of leptin effects onto GnRH circuits involving kisspeptins in the metabolic control of puberty onset.
neurons. Notwithstanding, neuroanatomical data suggest that neurons In doing so, we have intended not only to critically review the available
from the PMV innervate Kiss1 and GnRH neurons in the preoptic area; evidence suggesting an important role of leptin and kisspeptins in this
therefore, it is highly plausible that leptin-sensitive pathways origi- phenomenon, but also to stress the unsolved questions and the experi-
nating from the PMV may impinge onto Kiss1 circuits for transmitting mental data challenging to some extent this consensus view. Certainly,
part of the reproductive actions of leptin. A tentative model illustrating the basis for the metabolic regulation of puberty is much more complex
the putative direct, indirect and independent actions of leptin on Kiss1 than merely leptin–kisspeptin interactions, and involves the important
circuits, and thereby GnRH neurons, is provided in Fig. 1. contribution of additional peripheral signals (e.g., ghrelin, insulin) and
As additional note to this section, it is stressed that the experimental central neuropeptides (e.g., NPY, melanocortins). Notwithstanding this,
evidence available suggests that leptin can act at levels of the gonado- it is clear that identification of the essential reproductive roles of leptin
tropic axis other than the hypothalamus, including the pituitary and and kisspeptins, and of their ways of interaction, has revolutionized our
the gonads. At the pituitary level, leptin has been shown to modestly understanding of how reproductive maturity is attained and of the
stimulate gonadotropin secretion. In the gonads, both direct stimulato- mechanisms whereby this complex phenomenon is finely regulated by
ry and inhibitory actions of leptin on different aspects of gonadal func- (and tightly coupled to) the metabolic/energy state of the organism.
tion have been reported (Tena-Sempere, 2007; Tena-Sempere and
Barreiro, 2002). As noted earlier in this review, at high concentrations, Disclosure statement
leptin has been documented to (paradoxically) inhibit testicular and
ovarian steroidogenesis. This phenomenon may contribute to the state The authors have nothing to disclose.
of hypogonadism frequently associated to conditions of persistent
hyperleptinemia, such as morbid obesity (Tena-Sempere, 2007). In Acknowledgments
any event, pituitary and gonadal effects of leptin have been character-
ized mainly in adulthood; hence, the contribution of such peripheral ac- The authors are indebted with the members of the research team at
tions to the metabolic control of puberty is yet to be defined. the Physiology Section of the University of Cordoba, and especially
with J.M. Castellano, V.M. Navarro, J. Roa and L. Pinilla, who actively par-
Final remarks ticipated in the generation of experimental data discussed herein. The
work from the authors' laboratory summarized in this article was
Puberty is a complex developmental phenomenon that is sensitive to supported by grants BFU 2008-00984 and BFU2011-25021 (Ministerio
the regulatory actions of many endogenous and environmental signals. de Economía y Competitividad, Spain; co-funded with EU FEDER

CNS

C PMV

A
Leptin

Kiss1
GnRH
WAT
T Neuron
Neuron
B
Intermed
Intermediate
diate
Neuron

Hypothalamus

Fig. 1. Tentative diagram for the potential mode of action of leptin on Kiss1 and/or GnRH neurons. Direct (A), indirect (B) and independent (C) actions of leptin on Kiss1 pathways,
as putative mechanism for transmission of the regulatory effects of the adipose hormone on GnRH neurons, are depicted. Note, however, that primary actions of leptin in hypotha-
lamic nuclei, such as the ventral premmamilary nucleus (PMV), initially considered as kisspeptin-independent, do not necessarily exclude the possibility of some interplay with
Kiss1 circuits, which is represented as a dotted line projection. For further details, see Interplay of leptin and kisspeptins in the metabolic control of puberty: direct or indirect?
and Other sites of action of leptin in the metabolic control of puberty sections. WAT: White adipose tissue; CNS: Central Nervous System.
M.A. Sanchez-Garrido, M. Tena-Sempere / Hormones and Behavior 64 (2013) 187–194 193

funds), Project PI042082 (Ministerio de Sanidad, Spain), Project Elias, C.F., Purohit, D., 2013. Leptin signaling and circuits in puberty and fertility. Cell.
Mol. Life Sci. 70, 841–862.
P08-CVI-03788 (Junta de Andalucía, Spain), and EU research contract Fernandez-Fernandez, R., et al., 2006. Novel signals for the integration of energy bal-
DEER FP7-ENV-2007-1. CIBER is an initiative of Instituto de Salud Carlos ance and reproduction. Mol. Cell. Endocrinol. 254–255, 127–132.
III (Ministerio de Sanidad, Spain). Finn, P.D., et al., 1998. The stimulatory effect of leptin on the neuroendocrine reproduc-
tive axis of the monkey. Endocrinology 139, 4652–4662.
Garcia-Mayor, R.V., et al., 1997. Serum leptin levels in normal children: relationship to
age, gender, body mass index, pituitary–gonadal hormones, and pubertal stage.
References J. Clin. Endocrinol. Metab. 82, 2849–2855.
Hakansson, M.L., et al., 1998. Leptin receptor immunoreactivity in chemically defined
Ahima, R.S., et al., 1997. Leptin accelerates the onset of puberty in normal female mice. target neurons of the hypothalamus. J. Neurosci. 18, 559–572.
J. Clin. Invest. 99, 391–395. Han, S.K., et al., 2005. Activation of gonadotropin-releasing hormone neurons by kisspeptin
Ahima, R.S., et al., 2000. Leptin regulation of neuroendocrine systems. Front. as a neuroendocrine switch for the onset of puberty. J. Neurosci. 25, 11349–11356.
Neuroendocrinol. 21, 263–307. Herbison, A.E., et al., 2008. Gonadotropin-releasing hormone neuron requirements for
Ahmed, M.L., et al., 2009. Childhood obesity and the timing of puberty. Trends puberty, ovulation, and fertility. Endocrinology 149, 597–604.
Endocrinol. Metab. 20, 237–242. Herbison, A.E., et al., 2010. Distribution and postnatal development of Gpr54 gene expres-
Aksglaede, L., et al., 2009a. Age at puberty and the emerging obesity epidemic. PLoS sion in mouse brain and gonadotropin-releasing hormone neurons. Endocrinology
One 4, e8450. 151, 312–321.
Aksglaede, L., et al., 2009b. Recent decline in age at breast development: the Copenha- Ibanez, L., de Zegher, F., 2006. Puberty and prenatal growth. Mol. Cell. Endocrinol.
gen Puberty Study. Pediatrics 123, e932–e939. 254–255, 22–25.
Al-Qassab, H., et al., 2009. Dominant role of the p110beta isoform of PI3K over Kalamatianos, T., et al., 2008. Fasting reduces KiSS-1 expression in the anteroventral
p110alpha in energy homeostasis regulation by POMC and AgRP neurons. Cell periventricular nucleus (AVPV): effects of fasting on the expression of KiSS-1 and
Metab. 10, 343–354. neuropeptide Y in the AVPV or arcuate nucleus of female rats. J. Neuroendocrinol.
Backholer, K., et al., 2010. Kisspeptin cells in the ewe brain respond to leptin and com- 20, 1089–1097.
municate with neuropeptide Y and proopiomelanocortin cells. Endocrinology 151, Loret de Mola, J.R., 2009. Obesity and its relationship to infertility in men and women.
2233–2243. Obstet. Gynecol. Clin. North Am. 36, 333–346 (ix).
Barash, I.A., et al., 1996. Leptin is a metabolic signal to the reproductive system. Endo- Louis, G.W., et al., 2011. Molecular mapping of the neural pathways linking leptin to
crinology 137, 3144–3147. the neuroendocrine reproductive axis. Endocrinology 152, 2302–2310.
Bates, S.H., et al., 2003. STAT3 signalling is required for leptin regulation of energy bal- Luque, R.M., et al., 2007. Regulation of hypothalamic expression of KiSS-1 and GPR54
ance but not reproduction. Nature 421, 856–859. genes by metabolic factors: analyses using mouse models and a cell line. Endocri-
Biro, F.M., Wien, M., 2010. Childhood obesity and adult morbidities. Am. J. Clin. Nutr. nology 148, 4601–4611.
91, 1499S–1505S. Martos-Moreno, G.A., et al., 2010. Metabolic signals in human puberty: effects of over
Brown, R.E., et al., 2008. KiSS-1 mRNA in adipose tissue is regulated by sex hormones and undernutrition. Mol. Cell. Endocrinol. 324, 70–81.
and food intake. Mol. Cell. Endocrinol. 281, 64–72. Mayer, C., Boehm, U., 2011. Female reproductive maturation in the absence of
Caprio, M., et al., 2001. Leptin in reproduction. Trends Endocrinol. Metab. 12, 65–72. kisspeptin/GPR54 signaling. Nat. Neurosci. 14, 704–710.
Caron, E., et al., 2012. Alteration in neonatal nutrition causes perturbations in hypothalam- Morelli, A., et al., 2008. Sex steroids and leptin regulate the “first Kiss” (KiSS 1/G-protein-
ic neural circuits controlling reproductive function. J. Neurosci. 32, 11486–11494. coupled receptor 54 system) in human gonadotropin-releasing-hormone-secreting
Casanueva, F.F., Dieguez, C., 1999. Neuroendocrine regulation and actions of leptin. neuroblasts. J. Sex. Med. 5, 1097–1113.
Front. Neuroendocrinol. 20, 317–363. Navarro, V.M., Tena-Sempere, M., 2012. Neuroendocrine control by kisspeptins: role in
Castellano, J.M., et al., 2005. Changes in hypothalamic KiSS-1 system and restoration of metabolic regulation of fertility. Nat. Rev. Endocrinol. 8, 40–53.
pubertal activation of the reproductive axis by kisspeptin in undernutrition. Endo- Navarro, V.M., et al., 2004a. Developmental and hormonally regulated messenger
crinology 146, 3917–3925. ribonucleic acid expression of KiSS-1 and its putative receptor, GPR54, in rat hypo-
Castellano, J.M., et al., 2006a. Ontogeny and mechanisms of action for the stimulatory thalamus and potent luteinizing hormone-releasing activity of KiSS-1 peptide. En-
effect of kisspeptin on gonadotropin-releasing hormone system of the rat. Mol. docrinology 145, 4565–4574.
Cell. Endocrinol. 257–258, 75–83. Navarro, V.M., et al., 2004b. Advanced vaginal opening and precocious activation of the
Castellano, J.M., et al., 2006b. Expression of hypothalamic KiSS-1 system and rescue of reproductive axis by KiSS-1 peptide, the endogenous ligand of GPR54. J. Physiol.
defective gonadotropic responses by kisspeptin in streptozotocin-induced diabetic 561, 379–386.
male rats. Diabetes 55, 2602–2610. Navarro, V.M., et al., 2012. Role of neurokinin B in the control of female puberty and its
Castellano, J.M., et al., 2009a. Alterations in hypothalamic KiSS-1 system in experimen- modulation by metabolic status. J. Neurosci. 32, 2388–2397.
tal diabetes: early changes and functional consequences. Endocrinology 150, Oakley, A.E., et al., 2009. Kisspeptin signaling in the brain. Endocr. Rev. 30, 713–743.
784–794. Ojeda, S.R., Skinner, M.K., 2006. Puberty in the rat. In: Neill, J.D. (Ed.), The Physiology of
Castellano, J.M., et al., 2009b. KiSS-1/kisspeptins and the metabolic control of reproduc- Reproduction. Academic Press/Elsevier, San Diego, pp. 2061–2126.
tion: physiologic roles and putative physiopathological implications. Peptides 30, Ojeda, S.R., et al., 2010. New concepts on the control of the onset of puberty. Endocr.
139–145. Dev. 17, 44–51.
Castellano, J.M., et al., 2010a. Kisspeptins: bridging energy homeostasis and reproduc- Parent, A.S., et al., 2003. The timing of normal puberty and the age limits of sexual pre-
tion. Brain Res. 1364, 129–138. cocity: variations around the world, secular trends, and changes after migration.
Castellano, J.M., et al., 2010b. Acute inflammation reduces kisspeptin immunoreactivity Endocr. Rev. 24, 668–693.
at the arcuate nucleus and decreases responsiveness to kisspeptin independently Pineda, R., et al., 2010. Critical roles of kisspeptins in female puberty and preovulatory go-
of its anorectic effects. Am. J. Physiol. Endocrinol. Metab. 299, E54–E61. nadotropin surges as revealed by a novel antagonist. Endocrinology 151, 722–730.
Castellano, J.M., et al., 2011. Early metabolic programming of puberty onset: Impact of Pinilla, L., et al., 2012. Kisspeptins and reproduction: physiological roles and regulatory
changes in postnatal feeding and rearing conditions on the timing of puberty and de- mechanisms. Physiol. Rev. 92, 1235–1316.
velopment of the hypothalamic kisspeptin system. Endocrinology 152, 3396–3408. Plant, T.M., Durrant, A.R., 1997. Circulating leptin does not appear to provide a signal
Chehab, F.F., et al., 1997. Early onset of reproductive function in normal female mice for triggering the initiation of puberty in the male rhesus monkey (Macaca
treated with leptin. Science 275, 88–90. mulatta). Endocrinology 138, 4505–4508.
Cheung, C.C., et al., 1997. Leptin is a metabolic gate for the onset of puberty in the fe- Plant, T.M., et al., 2006. Repetitive activation of hypothalamic G protein-coupled recep-
male rat. Endocrinology 138, 855–858. tor 54 with intravenous pulses of kisspeptin in the juvenile monkey (Macaca
Clarkson, J., et al., 2009. Distribution of kisspeptin neurons in the adult female mouse mulatta) elicits a sustained train of gonadotropin-releasing hormone discharges.
brain. J. Neuroendocrinol. 21, 673–682. Endocrinology 147, 1007–1013.
Clarkson, J., et al., 2010. Neurobiological mechanisms underlying kisspeptin activation Pralong, F.P., 2010. Insulin and NPY pathways and the control of GnRH function and pu-
of gonadotropin-releasing hormone (GnRH) neurons at puberty. Mol. Cell. berty onset. Mol. Cell. Endocrinol. 324, 82–86.
Endocrinol. 324, 45–50. Qiu, J., et al., 2011. Guinea pig kisspeptin neurons are depolarized by leptin via activa-
Cravo, R.M., et al., 2011. Characterization of Kiss1 neurons using transgenic mouse tion of TRPC channels. Endocrinology 152, 1503–1514.
models. Neuroscience 173, 37–56. Quennell, J.H., et al., 2009. Leptin indirectly regulates gonadotropin-releasing hormone
Cunningham, M.J., et al., 1999. Leptin's actions on the reproductive axis: perspectives neuronal function. Endocrinology 150, 2805–2812.
and mechanisms. Biol. Reprod. 60, 216–222. Quennell, J.H., et al., 2011. Leptin deficiency and diet-induced obesity reduce hypotha-
de Roux, N., et al., 2003. Hypogonadotropic hypogonadism due to loss of function of lamic kisspeptin expression in mice. Endocrinology 152, 1541–1550.
the KiSS1-derived peptide receptor GPR54. Proc. Natl. Acad. Sci. U. S. A. 100, Rachon, D., Teede, H., 2010. Ovarian function and obesity—interrelationship, impact on
10972–10976. women's reproductive lifespan and treatment options. Mol. Cell. Endocrinol. 316,
Donato Jr., J., et al., 2009. The ventral premammillary nucleus links fasting-induced 172–179.
changes in leptin levels and coordinated luteinizing hormone secretion. J. Neurosci. Roa, J., Tena-Sempere, M., 2010. Energy balance and puberty onset: emerging role of
29, 5240–5250. central mTOR signaling. Trends Endocrinol. Metab. 21, 519–528.
Donato Jr., J., et al., 2011. Leptin's effect on puberty in mice is relayed by the ventral Roa, J., et al., 2008a. New frontiers in kisspeptin/GPR54 physiology as fundamental
premammillary nucleus and does not require signaling in Kiss1 neurons. J. Clin. Invest. gatekeepers of reproductive function. Front. Neuroendocrinol. 29, 48–69.
121, 355–368. Roa, J., et al., 2008b. Desensitization of gonadotropin responses to kisspeptin in the fe-
Elias, C.F., 2012. Leptin action in pubertal development: recent advances and unan- male rat: analyses of LH and FSH secretion at different developmental and meta-
swered questions. Trends Endocrinol. Metab. 23, 9–15. bolic states. Am. J. Physiol. Endocrinol. Metab. 294, E1088–E1096.
194 M.A. Sanchez-Garrido, M. Tena-Sempere / Hormones and Behavior 64 (2013) 187–194

Roa, J., et al., 2009. The mammalian target of rapamycin as novel central regulator of puberty Tena-Sempere, M., 2010a. Neuroendocrinology of puberty: recent milestones and new
onset via modulation of hypothalamic Kiss1 system. Endocrinology 150, 5016–5026. challenges. Mol. Cell. Endocrinol. 324, 1–2.
Roa, J., et al., 2010. Metabolic control of puberty onset: new players, new mechanisms. Tena-Sempere, M., 2010b. Roles of kisspeptins in the control of hypothalamic–
Mol. Cell. Endocrinol. 324, 87–94. gonadotropic function: focus on sexual differentiation and puberty onset. Endocr.
Seminara, S.B., et al., 2003. The GPR54 gene as a regulator of puberty. N. Engl. J. Med. Dev. 17, 52–62.
349, 1614–1627. Tena-Sempere, M., Barreiro, M.L., 2002. Leptin in male reproduction: the testis para-
Shahab, M., et al., 2005. Increased hypothalamic GPR54 signaling: a potential mechanism digm. Mol. Cell. Endocrinol. 188, 9–13.
for initiation of puberty in primates. Proc. Natl. Acad. Sci. U. S. A. 102, 2129–2134. Tena-Sempere, M., et al., 1999. Leptin inhibits testosterone secretion from adult rat tes-
Smith, J.T., et al., 2006. KiSS-1 neurones are direct targets for leptin in the ob/ob mouse. tis in vitro. J. Endocrinol. 161, 211–218.
J. Neuroendocrinol. 18, 298–303. True, C., et al., 2011. Leptin is not the critical signal for kisspeptin or luteinising hor-
Tajar, A., et al., 2010. Characteristics of secondary, primary, and compensated mone restoration during exit from negative energy balance. J. Neuroendocrinol.
hypogonadism in aging men: evidence from the European Male Ageing Study. 23, 1099–1112.
J. Clin. Endocrinol. Metab. 95, 1810–1818. Williams, K.W., et al., 2011. The acute effects of leptin require PI3K signaling
Tena-Sempere, M., 2007. Roles of ghrelin and leptin in the control of reproductive func- in the hypothalamic ventral premammillary nucleus. J. Neurosci. 31,
tion. Neuroendocrinology 86, 229–241. 13147–13156.

You might also like