Rev Esp Endocrinol Pediatr

Rev Esp Endocrinol Pediatr 2017;8(2):8-14 | Doi. 10.3266/RevEspEndocrinolPediatr.pre2017.Oct.429
Kisspeptins and Puberty

Sent for review: 17 Oct. 2017 | Accepted: 17 Oct. 2017  | Published: 14 Nov. 2017
JM Castellano, M Tena-Sempere
Inst. Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Dep. of Cell Biology, Physiology and Immunology. Univ. of Córdoba; Hosp. Universitario Reina Sofía; CIBER Fisiopatología de la Obesidad y Nutrición, Inst. de Salud Carlos III. Córdoba
Correspondence:M Tena-Sempere, Inst. Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Dep. of Cell Biology, Physiology and Immunology, Univ. of Córdoba; Hosp. Universitario Reina Sofía; CIBER Fisiopatología de la Obesidad y Nutrición, Inst. de Salud Carlos III, Córdoba
E-mail: fi1tesem@uco.es
Resumen

Puberty is a complex developmental phenomenon, driven by brain pathways under the modulation of external and internal cues, which culminates with the acquisition of reproductive competence and sexual maturity. From a neurobiological perspective, this process is incumbent to the timed activation of the population of hypothalamic neurons producing gonadotropin-releasing hormone (GnRH); the enhancement of GnRH neurosecretory activity being ultimately responsible for the triggering of puberty. In the last decade, kisspeptins have emerged as pivotal upstream regulators of GnRH neurons, with prominent roles in their activation during the pubertal transition and its (direct or indirect) modulation by different regulatory signals, including metabolic cues. We will briefly review herein the major features of kisspeptins and their producing neurons, the so-called Kiss1 neurons, as major gatekeepers and fine regulators of reproductive maturation and puberty onset in mammals.

INTRODUCTION

Puberty is major developmental event in the lifespan of any individual, which culminates with the attainment of sexual (somatic, psychological) maturity and reproductive capacity1. This intricate maturational phenomenon is grounded on early differentiation events (starting in utero), and involves a complex series of morphological, functional, endocrine, behavioral and psychological changes, which ultimately lead to the acquisition of a complete adult phenotype1. Accordingly, puberty is regarded not only as a specific, relatively narrow stage of development, but rather considered as the final output of a maturational continuum that leads to reproductive competence.

The tempo of puberty is dictated by the dynamic interplay between genetic and environmental factors1, so that perturbation of such dialogue often results in the inappropriate development of the reproductive axis that commonly leads to alterations of the timing of puberty (precocious, delayed or absent). In this sense, beyond its paramount biological relevance, puberty may be considered as putative sentinel for perturbations of the gene-environment interactions along early stages of development, whose alterations may lead to deregulation of key homeostatic systems. In fact, changes in the timing of puberty, especially earlier puberty, might impact important development events, including somatic and psychological maturation, and appear to be linked to numerous adverse health outcomes2, as well as reduced life expectancy3. This is specially worrying given the reported trends for changes in the age of puberty (mostly earlier), which seem to be more frequent in girls but appear to occur also in boys4-6. These observations urge for a better understanding of the physiological control of puberty, and of the pathophysiological basis of its alterations.

 

NEUROENDOCRINOLOGY OF PUBERTY: THE GnRH SECRETRORY DRIVE

The neuroendocrine system responsible for puberty onset is the so-called hypothalamic-pituitary-gonadal (HPG) axis, whose full activation permits the acquisition of reproductive competence at puberty7, 8. This neurohormonal system primarily integrates three major groups of factors, which are: (a) the decapeptide, GnRH, which is released in a pulsatile manner to the portal circuit connecting the medial-basal hypothalamus and the pituitary; (b) the gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), produced by gonadotropic cells of the anterior pituitary; and (c) the gonadal hormones, mainly sex steroids and several peptides, which operate via feedback loops at upper levels of the HPG axis to conduct its homeostatic regulation. In addition, the HPG axis is also modulated by other endogenous factors, e.g., metabolic hormones, and exogenous signals, e.g., nutrition and light conditions8, which contribute also to the dynamic control of puberty.

From its reproductive perspective, the initiation of puberty is founded on the heightening of the pulsatile release of hypothalamic GnRH9. Yet, the intimate mechanisms whereby this increase occurs remain ill defined. The current view, articulated in the so-called “central drive” hypothesis, concurs that the increase in the pulsatile GnRH secretion leading to the onset of puberty is the result of changes in the activity of the central pathways controlling GnRH neurons, with a switch in the balance between excitatory inputs (which increase) and inhibitory signals (which decrease at the time of puberty), thus causing the drive for the pubertal activation of the HPG axis9. Notably, the activator afferents of GnRH neurons include not only trans-synaptic inputs but also glial-born factors10.

 

KISSPEPTINS AND THE CENTRAL CONTROL OF PUBERTY

While the control of GnRH neurons is multifactorial, and numerous neuropeptides and transmitters have been shown in the last decades to regulate GnRH secretion, kisspeptins have emerged in recent years as fundamental regulatory signals, with an essential role in the control of puberty11. Kisspeptins are a family of structurally related peptides, encoded by the Kiss1 gene, which act via the G protein-coupled receptor, Gpr54, also termed Kiss1R or kisspeptin receptor11, 12. The reproductive facet of kisspeptins, and their role in the precise control of puberty, was surfaced by seminal observations, back in 2003, that inactivating mutations of GPR54 were linked to absence of puberty and hypogonadism of central origin (aka, hypogonadotropic hypogonadism) in humans13, 14. Such reproductive role was further demonstrated by the observation of similar phenotypes in patients with inactivating mutations of KISS1 and in null mice for Gpr54 or Kiss111.

These findings boosted enormous interest and prompted the analysis of the physiological roles of Kiss1 neurons in the regulation of GnRH neurosecretion and puberty onset. While detailed recapitulation of the results of these studies clearly exceeds the scope of this review, it is important to stress that such analyses have unambiguously documented a sophisticated developmental program of Kiss1 neurons, responsible for their activation during the pubertal maturation11, 15. This complex and multifaceted phenomenon includes, at least, the following major components: (a) an increase in the hypothalamic Kiss1 mRNA/kisspeptin content during the juvenile-pubertal transition that drives the full activation of the GnRH/gonadotropin system16-19; (b) a rise in the sensitivity to the excitatory actions of kisspeptins on GnRH secretion20, 21; (c) an enhancement of Gpr54 signaling efficiency in GnRH neurons; (d) a state of partial resistance to desensitization to kisspeptin stimulation21, 22; and (e) a rise of kisspeptin-positive neurons and their projections to GnRH neurons23, 24. In good agreement, pharmacological blockade of kisspeptin signaling has been reported to delay the onset of puberty in female rats25, whereas ablation of Kiss1 neurons in the juvenile period prevented pubertal maturation in female mice26.

Despite the solid clinical and experimental evidence suggesting a role of kisspeptins in the timing of puberty, one study using functional genomics to congenitally ablate Kiss1 neurons suggested that kisspeptin signaling seems to be dispensable for the attainment of female fertility26. In the same vein, another study in a mouse model causing a congenital 95% reduction in hypothalamic Kiss1 expression was compatible with roughly preserved reproductive function in male mice27; yet, a similar reduction in hypothalamic Kiss1 levels severely altered fertility in female mice. A tenable explanation is that these phenotypes might be due to incomplete neuronal/Kiss1 expression elimination, coupled to potential developmental compensation26, which can be activated by such congenital manipulations. Thus, these findings would not refute the essential role of hypothalamic kisspeptins in the physiological timing of puberty, but rather suggest that redundancy in Kiss1 signaling may help to safeguard reproduction in relatively adverse conditions, at least in rodents. 

 

PUTATIVE REGULATORS OF KISSPEPTINS: NEUROKININ B AND LEPTIN

Two major populations of Kiss1 neurons have been identified in the hypothalamus, as characterized in detail in rodents28. One is located in the rostral hypothalamus, mainly at the antero-ventral periventricular nucleus (AVPV), while the other is placed in the arcuate nucleus (ARC) or its equivalent infundibular region in humans28, 29. The latter, which is primarily involved in the tonic control of pulsatile gonadotropin secretion, have been shown to co-express the neuropeptides, neurokinin-B (NKB) and dynorphin (Dyn), which seemingly operate as co-regulators of kisspeptin neurons and eventual modulators of puberty onset30, 31. Therefore, this neuronal population has been named KNDy neurons, as they express Kiss1, NKB and Dyn. Admittedly, the level of co-expression of these neuropeptides varies depending on the species, the sex and the state of the HPG axis32, 33. In any event, compelling evidence suggest that NKB may operate as a major stimulatory signal for the auto-regulation of kisspeptin release by KNDy neurons, and hence an important component of the machinery responsible for pulsatile GnRH secretion. In turn, Dyn would conduct an opposite role, by inhibiting kisspeptin output from KNDy neurons, and thereby suppressing GnRH neurosecretion.

Suggestive, as yet incomplete evidence suggests a role of NKB in the control of puberty onset. Thus, it has been shown that central injection of the NKB agonist, senktide, in prepubertal female rats stimulates LH secretion34, while the hypothalamic expression of NKB gene increases before the rise of Kiss1 mRNA levels in female rodents35. In the same line, central blockade of NKB signaling caused a moderate delay of puberty onset in female rats34. Recent evidence has shown that the stimulation of NKB signaling or the inhibition of Dyn induces early puberty onset in female rats36. These findings are compatible with the view that the inhibitory effect of Dyn on kisspeptin neurosecretion decreases at the time of puberty, while the stimulatory effect of NKB gradually augments during this period. As a result of these sequential events, the pulsatile kisspeptin/GnRH/LH release would increase, thus leading to puberty onset37.

Another group of putative modulators of kisspeptin signaling are the metabolic factors, which are known to be potent modifiers of puberty onset. Among them, a major player is the adipose hormone, leptin38. The unequivocal essential role of leptin in the control of puberty is attested by the alterations in pubertal timing observed in conditions of leptin deficiency or (morbid) leptin excess, which are often associated with delayed or advanced puberty, respectively39, 40. In this context, experimental evidence has suggested that leptin is a positive modulator of Kiss1 neurons; a phenomenon that can contribute to transmitting the metabolic modulation puberty onset. Thus, conditions of leptin deficiency are known to suppress hypothalamic Kiss1 expression in rodents and sheep39, 40, while leptin administration activates kisspeptin neurons and/or increase Kiss1 expression in different species41-44, and cell lines45, 46. In addition, expression of the gene encoding the leptin receptor has been documented in a fraction of ARC kisspeptin neurons in mice and ewes42, 47. Nonetheless, whether leptin modulated Kiss1 neurons directly or indirectly has been the subject of intense debate, and studies involving selective elimination of leptin receptors from Kiss1 neurons congenitally, using the Cre-loxP technology, suggest that leptin signaling in this neuronal population is dispensable for attainment of reproductive competence48, while other studied have failed to detect a significant amount of functional leptin receptors in Kiss1 neurons before puberty 49, 50. These findings, together with the observations that leptin does influence Kiss1 neurons, would suggest a predominantly indirect mode of action, in which intermediary pathways and signals would play a relevant role. Among these, neuronal pathways originating from the ventral pre-mammillary nucleus (PMV)51, and others using nitric oxide as major transmitter52.

 

CONCLUSIONS AND FUTURE DIRECTIONS

Puberty, as fascinating developmental phenomenon, has drawn considerable attention, and numerous clinical and experimental studies, conducted in different mammalian species during the last decades, have contributed to substantially expand our understanding of the physiology of puberty and the basis of some of its major alterations. In this context, the discovery of kisspeptins, as essential regulatory elements for the central control of puberty in mammals, including humans, has revolutionized our knowledge about the intimate mechanisms of puberty, allowing to surface novel regulatory pathways whereby different factors, from environmental cues to endogenous hormones, contribute to the precise timing of puberty. While the progress has been astonishing, important aspects of kisspeptin and pubertal physiology remain ill defined and will likely focus considerable attention in the coming years. Among these facets, we foresee that elucidation of the molecular mechanisms for the precise control of Kiss1/kisspeptin expression and the relative role of the different Kiss1 neurons populations will be of special interest, as epitomized by recent exciting findings on the putative roles of epigenetic regulatory mechanisms and microRNAs in the central control of puberty53-55.    

 

Disclosure: no potential conflict of interest in relation to this presentation. 

References

1     Parent, A.S., Teilmann, G., Juul, A., Skakkebaek, N.E., Toppari, J. & Bourguignon, J.P. (2003) The timing of normal puberty and the age limits of sexual precocity: variations around the world, secular trends, and changes after migration. Endocr Rev 24, 668-693.

2     Day, F.R., Elks, C.E., Murray, A., Ong, K.K. & Perry, J.R. (2015) Puberty timing associated with diabetes, cardiovascular disease and also diverse health outcomes in men and women: the UK Biobank study. Sci Rep 5, 11208.

3     Lakshman, R., Forouhi, N.G., Sharp, S.J., Luben, R., Bingham, S.A., Khaw, K.T., Wareham, N.J. & Ong, K.K. (2009) Early age at menarche associated with cardiovascular disease and mortality. J Clin Endocrinol Metab 94, 4953-4960.

4     De Leonibus, C., Marcovecchio, M.L., Chiavaroli, V., de Giorgis, T., Chiarelli, F. & Mohn, A. (2013) Timing of puberty and physical growth in obese children: a longitudinal study in boys and girls. Pediatr Obes doi: https://dx.doi.org/10.1111/j.2047-6310.2013.00176.x.

5     Herman-Giddens, M.E., Steffes, J., Harris, D., Slora, E., Hussey, M., Dowshen, S.A., Wasserman, R., Serwint, J.R., Smitherman, L. & Reiter, E.O. (2012) Secondary sexual characteristics in boys: data from the Pediatric Research in Office Settings Network. Pediatrics 130, e1058-1068.

6     Aksglaede, L., Juul, A., Olsen, L.W. & Sorensen, T.I. (2009) Age at puberty and the emerging obesity epidemic. PLoS One 4, e8450.

7     Tena-Sempere, M. (2012) Deciphering puberty: novel partners, novel mechanisms. Eur J Endocrinol 167, 733-747.

8     Tena-Sempere, M., Huhtaniemi, I. (2003) Gonadotropins and gonadotropin receptors. In: Fauser, B.C.J.M. (Ed.), Reproductive Medicine –Molecular, Cellular and Genetic Fundamentals. Parthenon Publishing, New York., 225-244.

9     Ojeda, S.R., Skinner, M.K. (2006) Puberty in the rat. In: Neill, J.D. (Ed.), The Physiology of Reproduction. Academic Press/Elsevier, San Diego, pp. 2061–2126.

10   Ojeda, S.R., Roth, C., Mungenast, A., Heger, S., Mastronardi, C., Parent, A.S., Lomniczi, A. & Jung, H. (2006) Neuroendocrine mechanisms controlling female puberty: new approaches, new concepts. Int J Androl 29, 256-263; discussion 286-290.

11   Pinilla, L., Aguilar, E., Dieguez, C., Millar, R.P. & Tena-Sempere, M. (2012) Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol Rev 92, 1235-1316.

12   Oakley, A.E., Clifton, D.K. & Steiner, R.A. (2009) Kisspeptin signaling in the brain. Endocr Rev 30, 713-743.

13   Seminara, S.B., Messager, S., Chatzidaki, E.E., Thresher, R.R., Acierno, J.S., Jr., Shagoury, J.K., Bo-Abbas, Y., Kuohung, W., Schwinof, K.M., Hendrick, A.G., Zahn, D., Dixon, J., Kaiser, U.B., Slaugenhaupt, S.A., Gusella, J.F., O'Rahilly, S., Carlton, M.B., Crowley, W.F., Jr., Aparicio, S.A. & Colledge, W.H. (2003) The GPR54 gene as a regulator of puberty. N Engl J Med 349, 1614-1627.

14   de Roux, N., Genin, E., Carel, J.C., Matsuda, F., Chaussain, J.L. & Milgrom, E. (2003) Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A 100, 10972-10976.

15   Tena-Sempere, M. (2013) Keeping puberty on time: novel signals and mechanisms involved. Curr Top Dev Biol 105, 299-329.

16   Navarro, V.M., Castellano, J.M., Fernandez-Fernandez, R., Barreiro, M.L., Roa, J., Sanchez-Criado, J.E., Aguilar, E., Dieguez, C., Pinilla, L. & Tena-Sempere, M. (2004) Developmental and hormonally regulated messenger ribonucleic acid expression of KiSS-1 and its putative receptor, GPR54, in rat hypothalamus and potent luteinizing hormone-releasing activity of KiSS-1 peptide. Endocrinology 145, 4565-4574.

17   Navarro, V.M., Fernandez-Fernandez, R., Castellano, J.M., Roa, J., Mayen, A., Barreiro, M.L., Gaytan, F., Aguilar, E., Pinilla, L., Dieguez, C. & Tena-Sempere, M. (2004) Advanced vaginal opening and precocious activation of the reproductive axis by KiSS-1 peptide, the endogenous ligand of GPR54. J Physiol 561, 379-386.

18   Plant, T.M., Ramaswamy, S. & Dipietro, M.J. (2006) Repetitive activation of hypothalamic G protein-coupled receptor 54 with intravenous pulses of kisspeptin in the juvenile monkey (Macaca mulatta) elicits a sustained train of gonadotropin-releasing hormone discharges. Endocrinology 147, 1007-1013.

19   Shahab, M., Mastronardi, C., Seminara, S.B., Crowley, W.F., Ojeda, S.R. & Plant, T.M. (2005) Increased hypothalamic GPR54 signaling: a potential mechanism for initiation of puberty in primates. Proc Natl Acad Sci U S A 102, 2129-2134.

20   Castellano, J.M., Navarro, V.M., Fernandez-Fernandez, R., Castano, J.P., Malagon, M.M., Aguilar, E., Dieguez, C., Magni, P., Pinilla, L. & Tena-Sempere, M. (2006) Ontogeny and mechanisms of action for the stimulatory effect of kisspeptin on gonadotropin-releasing hormone system of the rat. Mol Cell Endocrinol 257-258, 75-83.

21   Han, S.K., Gottsch, M.L., Lee, K.J., Popa, S.M., Smith, J.T., Jakawich, S.K., Clifton, D.K., Steiner, R.A. & Herbison, A.E. (2005) Activation of gonadotropin-releasing hormone neurons by kisspeptin as a neuroendocrine switch for the onset of puberty. J Neurosci 25, 11349-11356.

22   Roa, J., Vigo, E., Garcia-Galiano, D., Castellano, J.M., Navarro, V.M., Pineda, R., Dieguez, C., Aguilar, E., Pinilla, L. & Tena-Sempere, M. (2008) Desensitization of gonadotropin responses to kisspeptin in the female rat: analyses of LH and FSH secretion at different developmental and metabolic states. Am J Physiol Endocrinol Metab 294, E1088-1096.

23   Bentsen, A.H., Ansel, L., Simonneaux, V., Tena-Sempere, M., Juul, A. & Mikkelsen, J.D. (2010) Maturation of kisspeptinergic neurons coincides with puberty onset in male rats. Peptides 31, 275-283.

24   Clarkson, J. & Herbison, A.E. (2006) Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons. Endocrinology 147, 5817-5825.

25   Pineda, R., Garcia-Galiano, D., Roseweir, A., Romero, M., Sanchez-Garrido, M.A., Ruiz-Pino, F., Morgan, K., Pinilla, L., Millar, R.P. & Tena-Sempere, M. (2010) Critical roles of kisspeptins in female puberty and preovulatory gonadotropin surges as revealed by a novel antagonist. Endocrinology 151, 722-730.

26   Mayer, C. & Boehm, U. (2011) Female reproductive maturation in the absence of kisspeptin/GPR54 signaling. Nat Neurosci 14, 704-710.

27   Popa, S.M., Moriyama, R.M., Caligioni, C.S., Yang, J.J., Cho, C.M., Concepcion, T.L., Oakley, A.E., Lee, I.H., Sanz, E., Amieux, P.S., Caraty, A., Palmiter, R.D., Navarro, V.M., Chan, Y.M., Seminara, S.B., Clifton, D.K. & Steiner, R.A. (2013) Redundancy in Kiss1 expression safeguards reproduction in the mouse. Endocrinology 154, 2784-2794.

28   Garcia-Galiano, D., Pinilla, L. & Tena-Sempere, M. (2012) Sex steroids and the control of the Kiss1 system: developmental roles and major regulatory actions. J Neuroendocrinol 24, 22-33.

29   Smith, J.T., Cunningham, M.J., Rissman, E.F., Clifton, D.K. & Steiner, R.A. (2005) Regulation of Kiss1 gene expression in the brain of the female mouse. Endocrinology 146, 3686-3692.

30   Lehman, M.N., Coolen, L.M. & Goodman, R.L. (2010) Minireview: kisspeptin/neurokinin B/dynorphin (KNDy) cells of the arcuate nucleus: a central node in the control of gonadotropin-releasing hormone secretion. Endocrinology 151, 3479-3489.

31   Navarro, V.M., Gottsch, M.L., Chavkin, C., Okamura, H., Clifton, D.K. & Steiner, R.A. (2009) Regulation of gonadotropin-releasing hormone secretion by kisspeptin/dynorphin/neurokinin B neurons in the arcuate nucleus of the mouse. J Neurosci 29, 11859-11866.

32   Hrabovszky, E., Sipos, M.T., Molnar, C.S., Ciofi, P., Borsay, B.A., Gergely, P., Herczeg, L., Bloom, S.R., Ghatei, M.A., Dhillo, W.S. & Liposits, Z. (2012) Low degree of overlap between kisspeptin, neurokinin B, and dynorphin immunoreactivities in the infundibular nucleus of young male human subjects challenges the KNDy neuron concept. Endocrinology 153, 4978-4989.

33   Overgaard, A., Ruiz-Pino, F., Castellano, J.M., Tena-Sempere, M. & Mikkelsen, J.D. (2014) Disparate changes in kisspeptin and neurokinin B expression in the arcuate nucleus after sex steroid manipulation reveal differential regulation of the two KNDy peptides in rats. Endocrinology 155, 3945-3955.

34   Navarro, V.M., Ruiz-Pino, F., Sanchez-Garrido, M.A., Garcia-Galiano, D., Hobbs, S.J., Manfredi-Lozano, M., Leon, S., Sangiao-Alvarellos, S., Castellano, J.M., Clifton, D.K., Pinilla, L., Steiner, R.A. & Tena-Sempere, M. (2012) Role of neurokinin B in the control of female puberty and its modulation by metabolic status. J Neurosci 32, 2388-2397.

35   Gill, J.C., Navarro, V.M., Kwong, C., Noel, S.D., Martin, C., Xu, S., Clifton, D.K., Carroll, R.S., Steiner, R.A. & Kaiser, U.B. (2012) Increased neurokinin B (Tac2) expression in the mouse arcuate nucleus is an early marker of pubertal onset with differential sensitivity to sex steroid-negative feedback than Kiss1. Endocrinology 153, 4883-4893.

36   Nakahara, T., Uenoyama, Y., Iwase, A., Oishi, S., Nakamura, S., Minabe, S., Watanabe, Y., Deura, C., Noguchi, T., Fujii, N., Kikkawa, F., Maeda, K. & Tsukamura, H. (2013) Chronic peripheral administration of kappa-opioid receptor antagonist advances puberty onset associated with acceleration of pulsatile luteinizing hormone secretion in female rats. J Reprod Dev 59, 479-484.

37   Uenoyama, Y., Tsukamura, H. & Maeda, K. (2014) KNDy neuron as a gatekeeper of puberty onset. J Obstet Gynaecol Res 40, 1518-1526.

38   Vazquez, M.J., Romero-Ruiz, A. & Tena-Sempere, M. (2015) Roles of leptin in reproduction, pregnancy and polycystic ovary syndrome: consensus knowledge and recent developments. Metabolism 64, 79-91.

39   Castellano, J.M. & Tena-Sempere, M. (2016) Metabolic control of female puberty: potential therapeutic targets. Expert Opin Ther Targets 20, 1181-1193.

40   Manfredi-Lozano, M., Roa, J. & Tena-Sempere, M. (2017) Connecting metabolism and gonadal function: Novel central neuropeptide pathways involved in the metabolic control of puberty and fertility. Front Neuroendocrinol.

41   Castellano, J.M., Navarro, V.M., Fernandez-Fernandez, R., Roa, J., Vigo, E., Pineda, R., Dieguez, C., Aguilar, E., Pinilla, L. &Tena-Sempere, M. (2006) Expression of hypothalamic KiSS-1 system and rescue of defective gonadotropic responses by kisspeptin in streptozotocin-induced diabetic male rats. Diabetes 55, 2602-2610.

42   Smith, J.T., Acohido, B.V., Clifton, D.K. & Steiner, R.A. (2006) KiSS-1 neurones are direct targets for leptin in the ob/ob mouse. J Neuroendocrinol 18, 298-303.

43   Backholer, K., Smith, J.T., Rao, A., Pereira, A., Iqbal, J., Ogawa, S., Li, Q. & Clarke, I.J. (2010) Kisspeptin cells in the ewe brain respond to leptin and communicate with neuropeptide Y and proopiomelanocortin cells. Endocrinology 151, 2233-2243.

44   Qiu, J., Fang, Y., Bosch, M.A., Ronnekleiv, O.K. & Kelly, M.J. (2011) Guinea pig kisspeptin neurons are depolarized by leptin via activation of TRPC channels. Endocrinology 152, 1503-1514.

45   Luque, R.M., Kineman, R.D. & Tena-Sempere, M. (2007) Regulation of hypothalamic expression of KiSS-1 and GPR54 genes by metabolic factors: analyses using mouse models and a cell line. Endocrinology 148, 4601-4611.

46   Morelli, A., Marini, M., Mancina, R., Luconi, M., Vignozzi, L., Fibbi, B., Filippi, S., Pezzatini, A., Forti, G., Vannelli, G.B. & Maggi, M. (2008) Sex steroids and leptin regulate the "first Kiss" (KiSS 1/G-protein-coupled receptor 54 system) in human gonadotropin-releasing-hormone-secreting neuroblasts. J Sex Med 5, 1097-1113.

47   Quennell, J.H., Howell, C.S., Roa, J., Augustine, R.A., Grattan, D.R. & Anderson, G.M. (2011) Leptin deficiency and diet-induced obesity reduce hypothalamic kisspeptin expression in mice. Endocrinology 152, 1541-1550.

48   Cravo, R.M., Margatho, L.O., Osborne-Lawrence, S., Donato, J., Jr., Atkin, S., Bookout, A.L., Rovinsky, S., Frazao, R., Lee, C.E., Gautron, L., Zigman, J.M. & Elias, C.F. (2011) Characterization of Kiss1 neurons using transgenic mouse models. Neuroscience 173, 37-56.

49   Cravo, R.M., Frazao, R., Perello, M., Osborne-Lawrence, S., Williams, K.W., Zigman, J.M., Vianna, C. & Elias, C.F. (2013) Leptin signaling in kiss1 neurons arises after pubertal development. PLoS One 8, e58698.

50   Qiu, X., Dao, H., Wang, M., Heston, A., Garcia, K.M., Sangal, A., Dowling, A.R., Faulkner, L.D., Molitor, S.C., Elias, C.F. & Hill, J.W. (2015) Insulin and Leptin Signaling Interact in the Mouse Kiss1 Neuron during the Peripubertal Period. PLoS One 10, e0121974.

51   Donato, J., Jr., Cravo, R.M., Frazao, R., Gautron, L., Scott, M.M., Lachey, J., Castro, I.A., Margatho, L.O., Lee, S., Lee, C., Richardson, J.A., Friedman, J., Chua, S., Jr., Coppari, R., Zigman, J.M., Elmquist, J.K. & Elias, C.F. (2011) Leptin's effect on puberty in mice is relayed by the ventral premammillary nucleus and does not require signaling in Kiss1 neurons. J Clin Invest 121, 355-368.

52   Bellefontaine, N., Chachlaki, K., Parkash, J., Vanacker, C., Colledge, W., d'Anglemont de Tassigny, X., Garthwaite, J., Bouret, S.G. & Prevot, V. (2014) Leptin-dependent neuronal NO signaling in the preoptic hypothalamus facilitates reproduction. J Clin Invest 124, 2550-2559.

53   Messina, A., Langlet, F., Chachlaki, K., Roa, J., Rasika, S., Jouy, N., Gallet, S., Gaytan, F., Parkash, J., Tena-Sempere, M., Giacobini, P. & Prevot, V. (2016) A microRNA switch regulates the rise in hypothalamic GnRH production before puberty. Nat Neurosci 19, 835-844.

54   Sangiao-Alvarellos, S., Manfredi-Lozano, M., Ruiz-Pino, F., Navarro, V.M., Sanchez-Garrido, M.A., Leon, S., Dieguez, C., Cordido, F., Matagne, V., Dissen, G.A., Ojeda, S.R., Pinilla, L. & Tena-Sempere, M. (2013) Changes in hypothalamic expression of the Lin28/let-7 system and related microRNAs during postnatal maturation and after experimental manipulations of puberty. Endocrinology 154, 942-955.

55   Lomniczi, A., Wright, H. & Ojeda, S.R. (2015) Epigenetic regulation of female puberty. Front Neuroendocrinol 36, 90-107.



Comments
Name*: Surname*:
E-mail*:
Hospital*:
Address:
C.P.: Country:
Comments*:
(450 Words)
Security code*:
* Required Fields
Send
Send Send Send
Send