You are on page 1of 7

Accepted: 11 December 2017

DOI: 10.1111/and.12965

INVITED REVIEW

Review of the role of leptin in the regulation of male


reproductive function

J. Zhang  | M. Gong

Department of Urology, Shanghai Pudong


Hospital, Fudan University, Shanghai, China Summary
Since discovered in 1994, leptin has been thought to be a pleiotropic hormone that
Correspondence
Min Gong, Department of Urology, Shanghai regulates food intake, controls energy balance in the body and influences multiple tis-
Pudong Hospital, Fudan University, Shanghai, sues in the body. Leptin plays an important mediating role in the regulation of neu-
China.
Email: mg813@hotmail.com roendocrine and can transmit the nutritional status signals to the reproductive-­related

Funding information
central nervous system. Many studies have shown that leptin may play an important
This work was supported by Research role in the control of reproductive function. Leptin can act on all levels of the hypo-
Grant for Health Science and Technology of
Shanghai Municipal Commission of Health
thalamus–pituitary–gonadal (HPG) axis and may have local effects on the function of
and family Planning (201440455) and Key testis and spermatogenesis. Leptin is critical for puberty initiation and can also modu-
Discipline Construction Project of Pudong
Health and Family Planning Commission of
late testosterone synthesis by downregulating cAMP-­dependent activation of steroi-
Shanghai (PWZxk2017-­21). dogenic genes expressions. Leptin is found to be higher in infertile men than in normal
subjects. Yet, the exact role of leptin in the regulation of male reproductive function
remains incomplete. The purpose of this review was to summarise the recent research
about the biological effects of leptin on male reproductive system. In-­depth study of
leptin in reproductive system will help to reveal the pathogenesis of infertility and
provide new treatment ideas for human assisted reproductive technology.

KEYWORDS
infertility, leptin, leptin receptor, male reproductive system

1 |  INTRODUCTION reproductive system regulation and demonstrate that leptin partic-
ipates in reproductive regulation (Gonzalez et al., 2000; Henson &
In 1994,Zhang et al. (1994) obtained the obese gene using the posi- Castracane, 2006; Messinis & Milingos, 1999). However, there are
tional cloning technique of mutant genes and initiated the study of still many problems to be explored in the regulation mechanism of
leptin. Leptin, secreted by adipose tissue, is the hormonal product leptin on male reproductive system. In this review, we highlight
of the obese gene. It is found that leptin is a metabolic signal that the regulation of leptin and leptin receptor on male reproductive
connects nutrition and other physiological functions (Barash et al., system.
1996). Leptin not only plays an important role in energy metabolism
but also participates in a series of important physiological activi-
ties such as immune regulation, angiogenesis, inflammatory reac- 2 | STRUCTURE AND EXPRESSION OF
tion and bone formation (Budak et al., 2006; Huang & Li, 2000). LEPTIN AND LEPTIN RECEPTOR
Obese gene mutations in mice (ob/ob) show obesity and infertility
(Caprio et al., 1999), and leptin administration to leptin-­deficient Leptin is a 16-­kDa protein hormone composed of 167 amino acids.
ob/ob mice can promote their sexual maturity (Cravo et al., 2013), The N-­terminal of leptin has a signal peptide sequences, consisting of
suggesting that leptin may play an important role in the reproduc- 21 amino acid residues, and the C-­terminal has a ring structure with
tive system. Emerging studies are mainly concentrated in the female intramolecular disulphide bond, consisting of two cysteine residues

Andrologia. 2018;e12965. wileyonlinelibrary.com/journal/and © 2018 Blackwell Verlag GmbH  |  1 of 7


https://doi.org/10.1111/and.12965
|
2 of 7       ZHANG and GONG

F I G U R E   1   Six different isoforms of


leptin receptor (OB-­R). The six isoforms
of OB-­R are divided into three categories:
short (OB-­Ra, OB-­Rc, OB-­Rd, OB-­Rf), long
(OB-­Rb) and soluble (OB-Re) forms. Apart
from OB-Re, every form includes three
domains: extracellular, transmembrane and
intracellular. All leptin receptor isoforms
share the same ligand-­binding extracellular
domain. The short type receptor contains
an intracellular fragment with 30~40
amino acids while the long isoform has
an extended intracellular domain of 301
amino acids with binding sites for signal
transduction

(Imagawa et al., 1998). After entering the blood circulation, the N-­ 3 | REGULATION OF LEPTIN ON MA LE
terminal signal peptide is removed to form a biologically active lep- REPRODUCTIVE SYSTEM
tin. Leptin is free in the blood or binds to the leptin binding protein.
3.1 | Puberty
It reaches the central or peripheral tissue and plays a biological role
in combination with a variety of leptin receptors (Margetic, Gazzola, The development of puberty is mainly controlled by genes and in-
Pegg, & Hill, 2002; Sainz, Barrenetxe, Moreno-­Aliaga, & Martinez, fluenced by a multihormonal effect (Budak et al., 2006). Puberty is
2015). Leptin is predominantly expressed in adipose tissue and also characterised by an increase in testosterone driven by increasing
found in other tissues or cells such as hypothalamus, pituitary, gas- concentrations of pituitary gonadotrophins and gonadotrophin-­
tric epithelium, embryo, skeletal muscle, testis and sperm (Gale, releasing hormone (GnRH) in boys (El-­Eshmawy, Aal, & El Hawary,
Castracane, & Mantzoros, 2004; Ishikawa, Fujioka, Ishimura, Takenaka, 2010). Both animal experiments and human studies have confirmed
& Fujisawa, 2007). that leptin plays an important role in the initiation of puberty. Mice
In 1995, Tartaglia et al. (1995) identified and cloned the leptin lacking leptin (ob/ob) or the leptin receptor (db/db) exhibit hypo-
receptor (OB-­R) gene in mice and humans. The human OB-­R is com- gonadism and stay prepubertal (Strobel, Issad, Camoin, Ozata, &
posed of 1165 amino acids and is divided into three domains: ex- Strosberg, 1998). Leptin administration to ob/ob mice leads to pu-
tracellular, transmembrane and intracellular. OB-­R belongs to class bertal development, and the same situation does not occur in db/db
I cytokine superfamily, and there are at least six different isoforms, mice (Donato et al., 2011). In addition, ob/ob mice demonstrated a
named OB-­Ra, OB-­Rb, OB-­Rc, OB-­Rd, OB-­Re, OB-­Rf. The six isoforms marked sensitivity to negative feedback of gonadotropins because
are divided into three categories: short (OB-Ra, OB-Rc, OB-­Rd, OB-­ of persistent immaturity of the hypothalamic–pituitary axis, con-
Rf), long (OB-­Rb) and soluble (OB-­Re) forms (Catteau et al., 2016; sistent with a prepubertal state of high negative feedback restraint
Wauman, Zabeau, & Tavernier, 2017). (Figure 1) Different OB-­R ac- (Elias & Purohit, 2013; Swerdloff, Batt, & Bray, 1976). In parallel
tivation conveys different biological activities of leptin. Short forms with mice, those who have mutation in leptin or leptin receptor
of OB-­R are mainly distributed in the brain, lung, cerebral choroid gene show failure of pubertal growth spurt and lack of secondary
plexus and the brain microvascular plexus (Banks, Kastin, Huang, sexual characteristics (Farooqi & O’Rahilly, 2006). Leptin treatment
Jaspan, & Maness, 1996). OB-­Ra and OB-­Rc are responsible for the can lead to elevated gonadotropin and sex hormone levels, puber-
translocation of leptin, which helps leptin penetrate the cell barrier, tal development and gonadal development (Farooqi et al., 2002).
such as the blood–brain barrier and the placenta (Dardeno et al., Several studies found that adolescent boys with constitutional
2010; Smith & Waddell, 2003). OB-­Re is a soluble receptor lack- delay of growth and puberty had significantly lower leptin than
ing intracellular and transmembrane segment. OB-­Re is the major the normal (Bideci, Cinaz, Hasanoglu, & Tumer, 2002; El-­Eshmawy
leptin binding protein in circulation and modulates leptin bioavail- et al., 2010; Gill, Hall, Tillmann, & Clayton, 1999). The delayed pu-
ability (Ge, Huang, Pourbahrami, & Li, 2002; Schaab & Kratzsch, berty onset is the result of low leptin production (Elias & Purohit,
2015). OB-­Rb, which is highly expressed in the hypothalamus, is a 2013). However, the exact role of leptin in the initiation of puberty
functional receptor that contains three highly conserved tyrosine is unclear. Several studies have shown that leptin levels increase
residues binding to the Janus kinase/signal transducer and activator prepubertally and then decline to baseline levels after the initiation
of transcription (JAK/STAT) for signal transduction (Wauman et al., of puberty (Mantzoros, Flier, & Rogol, 1997; Maqsood et al., 2007),
2017). suggesting that leptin plays an important role in the initiation of
ZHANG and GONG |
      3 of 7

puberty. However, Ozata, Ozdemir, and Licinio (1999) believed that et al., 2003). In vivo, experiments confirm that kisspeptins can pro-
leptin was not the primary factor of puberty initiation because they mote the release of GnRH and increase serum FSH, LH and testos-
found a delay of over 20 years in the onset of regular menstruation terone (Thompson et al., 2004). Smith, Acohido, Clifton, and Steiner
in a leptin-­deficient woman. Instead, it might only play a permis- (2006) found approximately 40% of the cells expressing Kiss1 in
sive role, and other metabolic and endocrine factors contributed the ARC coexpressed the leptin receptor. The mRNA of kisspeptins
to the maturation of the reproductive system. Rutters et al. (2009) in the ARC of ob/ob mice significantly reduced compared to the
found that the leptin concentrations increased significantly from wild-­type control (Quennell et al., 2011) and increased when leptin
Tanner stage 1 to 2 and decreased significantly from Tanner stage was administered to ob/ob mice (Luque, Kineman, & Tenasempere,
2 to 5, suggesting that leptin played a different role at different 2007). Furthermore, the Kiss1 cells in the ARC contain leptin-­
times. Exogenous leptin accelerates puberty initiation without af- activated signal transduction pathways (Quennell et al., 2011).
fecting body weight, suggesting that leptin links puberty initiation Leptin can promote pulsed secretion of GnRH in arcuate nucleus
with weight and energy storage (Farooqi, 2002). Many nuclei in the neurons of hypothalamus in a dose-­dependent manner (Lebrethon,
hypothalamus are involved in the regulation of leptin on puberty ini- Vandersmissen, Gerard, Parent, & Bourguignon, 2000). In addition,
tiation. Donato et al. (2011) found that the development of puberty leptin can promote the release of LH and FSH by activation of nitric
regulated by leptin was achieved by the expression of the excitatory oxide synthase and regulate the growth and differentiation of pitui-
neurotransmitter glutamate in ventral premammillary nucleus(PMV) tary cells (Iqbal, Pompolo, Considine, & Clarke, 2000; Tezuka et al.,
instead of Kiss1 neurons. Leptin directly activates PMV, which in 2002). Studies have shown that leptin is also expressed in the tes-
turn activates its downstream GnRH neurons (Leshan et al., 2009). tis, and leptin is involved in the regulation of testis (Soyupek et al.,
α-­melanocyte-­stimulating hormone (α-­MSH) is the major product 2005; Zamorano et al., 1997). Leptin seems to be a direct factor
of proopiomelanocortin (POMC) gene in the arcuate nucleus (ARC) in inhibiting testicular steroidogenesis (Landry, Cloutier, & Martin,
(Yong, Faulkner, & Hill, 2011). Manfredilozano et al. (2016) unveiled 2013). Tena-­
Sempere found that leptin could inhibit basal and
a novel leptin-­α-­MSH-­Kiss1-­GnRH signalling pathway which regu- stimulated testicular testosterone secretion in vitro, and several de-
lated puberty onset and he believed thatα-­MSH was an important creased mRNAs of upstream elements in the steroidogenic pathway
downstream regulator of leptin’s effect on pubertal awakening of (steroidogenic factor 1, steroidogenic acute regulatory protein and
the HPG axis. Venancio found that short-­term high-­fat diet could in- cytochrome P450 cholesterol sidechain cleavage enzyme) might
crease kisspeptin expression and induce earlier puberty, which was contribute to leptin-­induced inhibition of rat testicular steroido-
associated with an increase in cocaine-­and amphetamine-­regulated genesis (Tena-­Sempere et al., 1999, 2001). Fombonne (Fombonne,
transcript (CART) neurons expression in the ARC (Venancio et al., Charrier, Goddard, Moyse, & Krantic, 2007) found that leptin could
2017). Mammalian target of rapamycin (mTOR) is also involved in inhibit division of prepubertal Leydig cells via a cyclin D1. Leptin
puberty initiation by regulating the expression of Kiss1 and blocking inhibited testosterone secretion by interfering with the cAMP sig-
the mTOR pathway can disrupt normal puberty initiation time (Roa nalling and downregulating the expression of steroidogenic acute
& Tena-­Sempere, 2010; Roa et al., 2010). regulatory protein (Lin et al., 2009). Landry found that high levels
of leptin downregulated STAT transcriptional activity and reduced
cAMP-­dependent steroidogenic genes. (Landry, Sormany, Haché,
3.2 | Regulation on hypothalamus–pituitary–
Roumaud, & Martin, 2017). In addition to abundant expressions of
testis axis
OB-­Rb, other isoforms such as OB-Ra, Ob-­Rf, OB-Re and OB-Rc
A large number of studies have shown that the brain is a key posi- were also detected in prepubertal and adult testes, suggesting a
tion for leptin to play a role. Leptin plays a role in multiple parts of complex pattern of signalling processing (Tena-­Sempere & Barreiro,
the brain, including arcuate nucleus (ARC), ventral medial nucleus 2002). In contrast, testosterone can also inhibit leptin secretion by
(VMN), paraventricular nucleus (PVN)(Van Vugt et al., 2006). Both in acting on white fat, resulting in inverse correlation between leptin
vitro and in vivo, experiments have shown that leptin can stimulate and testosterone levels (Luukkaa et al., 1998). Overall, the mecha-
LH secretion (Yu, Kimura, Walczewska, Karanth, & McCann, 1997). nism of leptin regulating the hypothalamus–pituitary–testis axis is
However, GnRH cells do not express leptin receptors (Skorupskaite, complex, involving not only stimulatory but also inhibitory effects
George, & Anderson, 2014), suggesting that leptin may regulate the (Figure 2).
reproductive system indirectly. It has been proposed that GnRH
neurons are affected by leptin mediated via other neuropeptides,
3.3 | Leptin and male infertility
such as neuropeptide Y (NPY), agouti-­regulated peptide (AgRP),
proopiomelanocortin (POMC) and gama-­aminobutyric acid (GABA) Both leptin-­deficient genes in humans and rats exhibit a range of
(Iqbal et al., 2001; Sullivan & Moenter, 2004). In addition, more and reproductive system abnormalities, including decreased weights of
more studies have shown that leptin mediates the reproductive ventral prostate and testes, fewer sperm in the seminiferous tubules
system by regulating kisspeptins produced by the Kiss1 gene. Both and smaller size of Leydig cells (Elias & Purohit, 2013; Mounzih, Lu, &
mice and human experiments show that the lack of Kiss1 receptors Chehab, 1997). Studies have shown that leptin levels in patients with
caused by genetic mutations can lead to hypogonadism (Seminara azoospermia are higher than those with normal semen, and plasma
|
4 of 7       ZHANG and GONG

F I G U R E   2   Interactions between leptin and the hypothalamic–pituitary–testicle axis. Leptin interacts with neuroendocrine axis and transmits
body metabolic signals to the reproductive system. Leptin can stimulate POMC/CART and inhibit NPY/AgRP neurons of the arcuate nucleus
which elicit anorexic signals to increase the feeling of satiety. Meanwhile, leptin can act on the hypothalamus–pituitary–testicle axis to regulate
gonadotropin secretion and spermatogenesis. POMC, proopiomelanocortin; CART, amphetamine-­regulated transcript; NPY, neuropeptide Y;
AgRP, agouti-­related peptide; ME, median eminence; GnRH, gonadotropin-­releasing hormone; LH, luteinizing hormone; FSH, follicle-­stimulating
hormone; T: testosterone; DHT, dihydrotestosterone

leptin levels are not significantly correlated with FSH and LH, suggest- of sperm cell membrane. Aquila et al. (2005) found leptin secreted by
ing that leptin can act directly on the testis (Ma, Chen, Wang, Hu, & sperm modulated its metabolism independently by systemic leptin
Huang, 2011; Steinman et al., 2001; Zorn, Osredkar, Meden-­Vrtovec, and was closely related to sperm capacitation. Lampiao and du Plessis
& Majdic, 2007). Ishikawa et al. (2007) investigated the relationships (2008) found that leptin could trigger STAT3 signal transduction path-
between the expression of leptin in the testis and spermatogenesis way and anti-­apoptotic protein BCL-­2 to enhance sperm capacitation
and found that leptin was expressed on germ cells and was mainly index and acrosin activity. Exogenous leptin administration adversely
expressed in spermatocytes, whereas OB-­R was expressed predomi- affects sperm count and morphology (Almabhouh et al., 2015; Haron,
nantly on Leydig cells. He also found that the expression rate of OB-­R D’Souza, Jaafar, Zakaria, & Singh, 2010), which can be prevented by
in Leydig cells was negatively correlated with the serum testosterone melatonin (Almabhouh & Singh, 2017; Almabhouh et al., 2017). These
concentration. Chen et al. (2009) found that the expression of leptin findings indicate a pathophysiological relevance of seminal leptin in
and its receptor in the testis of experimental varicocele rats increased sperm. However, Hatami-­Baroogh et al. (2010) evaluated leptin re-
and was negatively correlated with testicular weight, Johnsen score, ceptor expression and found that the long isoform of leptin recep-
thickness of seminiferous epithelium and diameter of the seminifer- tor was not expressed in sperm. The expression of leptin receptor at
ous tubules. Many studies have found that human seminal plasma mRNA level may be related to the contamination of other cells. In
contains leptin, which is mainly secreted by the seminal vesicles or addition, Li, Chiu, Cheung, and Yeung (2009) believed that leptin did
prostate tissues and exists in free form (Aquila et al., 2005; Camiña not exert any effects on sperm motility and capacitation/acrosome
et al., 2002). Glander, Lammert, Paasch, Glasow, and Kratzsch (2002) reaction. Further study is needed to elucidate these controversial is-
found that leptin level in seminal plasma was significantly higher in in- sues regarding the presence, location and function of leptin receptor
fertility patients and showed a significantly negative correlation with in human spermatozoa.
percentage and velocity of motile spermatozoa, whereas Zorn et al.
(2007) found no correlation between leptin and sperm characteris-
tics. Ma et al. (2011) thought that leptin was an important marker for 4 | CONCLUSION
the detection of nonobstructive azoospermia. In addition to affecting
sperm motility, leptin may also be involved in regulating the physio- In recent years, with the progress of genetic modified mouse mod-
logical function of spermatozoa. Jope, Lammert, Kratzsch, Paasch, and els and molecular biology techniques, we have a better understand-
Glander (2003) found leptin and leptin receptor expression in human ing of the role of leptin in reproductive neuroendocrine axis. Leptin
spermatozoa, and leptin receptor was closely related to the integrity may play an important role in regulating the reproductive system
ZHANG and GONG |
      5 of 7

through different levels of expression and through acting on differ- Chen, B., Guo, J. H., Lu, Y. N., Ying, X. L., Hu, K., Xiang, Z. Q., … Huang, Y.
ent parts of the hypothalamus–pituitary–testis axis. Leptin interacts R. (2009). Leptin and varicocele-­related spermatogenesis dysfunction:
Animal experiment and clinical study. International Journal of Andrology,
with neuroendocrine axis and transmits body metabolic signals to
32(5), 532–541.
the reproductive system. Many mechanisms such as the effects of Cravo, R. M., Frazao, R., Perello, M., Osborne-Lawrence, S., Williams, K. W.,
leptin on testicular physiology and spermatogenesis are not yet fully Zigman, J. M., … Elias, C. F. (2013). Leptin signaling in Kiss1 neurons
understood. An in-­depth study of leptin on male reproductive regu- arises after pubertal development. PLoS ONE, 8(3), e58698.
Dardeno, T. A., Chou, S. H., Moon, H.-S., Chamberland, J. P., Fiorenza, C. G.,
lation will help to reveal some of the mechanisms of male infertil-
& Mantzoros, C. S. (2010). Leptin in human physiology and therapeu-
ity, which is expected to provide new ideas for the diagnosis and tics. Frontiers in Neuroendocrinology, 31(3), 377–393.
treatment. Donato, J. Jr, Cravo, R. M., Frazao, R., Gautron, L., Scott, M. M., Lachey, J.,
… 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
CO NFLI CT OF I NTE RE ST neurons. The Journal of Clinical Investigation, 121(1), 355–368.
El-Eshmawy, M. M., Aal, I. A. A., & El Hawary, A. K. (2010). Association
The authors declare that there are no conflict of interests regarding of ghrelin and leptin with reproductive hormones in constitutional
the publication of this paper. delay of growth and puberty. Reproductive Biology and Endocrinology,
8, 153.
Elias, C. F., & Purohit, D. (2013). Leptin signaling and circuits in puberty and
fertility. Cellular and Molecular Life Sciences, 70(5), 841–862.
O RCI D Farooqi, I. S. (2002). Leptin and the onset of puberty: Insights from ro-
dent and human genetics. Seminars in Reproductive Medicine, 20(2),
J. Zhang  http://orcid.org/0000-0001-8831-7464
139–144.
Farooqi, I. S., Matarese, G., Lord, G. M., Keogh, J. M., Lawrence, E., Agwu,
C., & O’Rahilly, S. (2002). Beneficial effects of leptin on obesity, T cell
hyporesponsiveness, and neuroendocrine/metabolic dysfunction of
REFERENCES
human congenital leptin deficiency. The Journal of Clinical Investigation,
Almabhouh, F. A., Osman, K., Ibrahim, S. F., Gupalo, S., Gnanou, J., Ibrahim, 110(8), 1093–1103.
E., & Singh, H. J. (2017). Melatonin ameliorates the adverse effects of Farooqi, S., & O’Rahilly, S. (2006). Genetics of obesity in humans. Endocrine
leptin on sperm. Asian Journal of Andrology, 19(6), 647–654. https://doi. Reviews, 27(7), 710–718.
org/10.1111/and.12325 Fombonne, J., Charrier, C., Goddard, I., Moyse, E., & Krantic, S. (2007).
Almabhouh, F. A., Osman, K., Siti Fatimah, I., Sergey, G., Gnanou, J., & Leptin-­mediated decrease of cyclin A2 and increase of cyclin D1 ex-
Singh, H. J. (2015). Effects of leptin on sperm count and morphology in pression: Relevance for the control of prepubertal rat Leydig cell divi-
Sprague-­Dawley rats and their reversibility following a 6-­week recov- sion and differentiation. Endocrinology, 148(5), 2126–2137.
ery period. Andrologia, 47(7), 751–758. Gale, S. M., Castracane, V. D., & Mantzoros, C. S. (2004). Energy homeo-
Almabhouh, F. A., & Singh, H. J. (2017). Adverse effects of leptin on histone-­ stasis, obesity and eating disorders: Recent advances in endocrinology.
to-­protamine transition during spermatogenesis are prevented by mel- Journal of Nutrition, 134(2), 295–298.
atonin in Sprague-­Dawley rats. Andrologia, 50(1). http://onlinelibrary. Ge, H., Huang, L., Pourbahrami, T., & Li, C. (2002). Generation of soluble
wiley.com/doi/10.1111/and.12814/full. leptin receptor by ectodomain shedding of membrane-­spanning re-
Aquila, S., Gentile, M., Middea, E., Catalano, S., Morelli, C., Pezzi, V., & Ando, ceptors in vitro and in vivo. Journal of Biological Chemistry, 277(48),
S. (2005). Leptin secretion by human ejaculated spermatozoa. Journal of 45898–45903.
Clinical Endocrinology and Metabolism, 90(8), 4753–4761. Gill, M. S., Hall, C. M., Tillmann, V., & Clayton, P. E. (1999). Constitutional
Banks, W. A., Kastin, A. J., Huang, W., Jaspan, J. B., & Maness, L. M. (1996). delay in growth and puberty (CDGP) is associated with hypoleptinae-
Leptin enters the brain by a saturable system independent of insulin. mia. Clinical Endocrinology -­Oxford, 50(6), 721–726.
Peptides, 17(2), 305–311. Glander, H. J., Lammert, A., Paasch, U., Glasow, A., & Kratzsch, J. (2002).
Barash, I. A., Cheung, C. C., Weigle, D. S., Ren, H., Kabigting, E. B., Kuijper, J. Leptin exists in tubuli seminiferi and in seminal plasma. Andrologia,
L., … Steiner, R. A. (1996). Leptin is a metabolic signal to the reproduc- 34(4), 227–233.
tive system. Endocrinology, 137(7), 3144–3147. Gonzalez, R. R., Simon, C., Caballero-Campo, P., Norman, R., Chardonnens,
Bideci, A., Cinaz, P., Hasanoglu, A., & Tumer, L. (2002). Leptin, insulin-­like D., Devoto, L., & Bischof, P. (2000). Leptin and reproduction. Human
growth factor (IGF)-­I and IGF binding protein-­3 levels in children with Reproduction Update, 6(3), 290–300.
constitutional delay of growth. Journal of Pediatric Endocrinology and Haron, M. N., D’Souza, U. J. A., Jaafar, H., Zakaria, R., & Singh, H. J. (2010).
Metabolism, 15(1), 41–46. Exogenous leptin administration decreases sperm count and increases
Budak, E., Fernandezsanchez, M., Bellver, J., Cervero, A., Simon, C., & the fraction of abnormal sperm in adult rats. Fertility and Sterility, 93(1),
Pellicer, A. (2006). Interactions of the hormones leptin, ghrelin, adi- 322–324.
ponectin, resistin, and PYY3-­36 with the reproductive system. Fertility Hatami-Baroogh, L., Razavi, S., Zarkesh-Esfahani, H., Tavalaee, M., Tanhaei,
and Sterility, 85(6), 1563–1581. S., Ghaedi, K., … Nasr-Esfahani, M. H. (2010). Evaluation of the leptin re-
Camiña, J. P., Lage, M., Menendez, C., Graña, M., García-Devesa, J., Dieguez, ceptor in human spermatozoa. Reproductive Biology and Endocrinology,
C., & Casanueva, F. F. (2002). Evidence of free leptin in human seminal 8, 17.
plasma. Endocrine, 17(3), 169–174. Henson, M. C., & Castracane, V. D. (2006). Leptin in pregnancy: An update.
Caprio, M., Isidori, A. M., Carta, A. R., Moretti, C., Dufau, M. L., & Fabbri, A. Biology of Reproduction, 74(2), 218–229.
(1999). Expression of functional leptin receptors in rodent Leydig cells. Huang, L., & Li, C. (2000). Leptin: A multifunctional hormone. Cell Research,
Endocrinology, 140(11), 4939–4947. 10(2), 81–92.
Catteau, A., Caillon, H., Barrière, P., Denis, M. G., Masson, D., & Fréour, T. Imagawa, K., Numata, Y., Katsuura, G., Sakaguchi, I., Morita, A., Kikuoka, S.,
(2016). Leptin and its potential interest in assisted reproduction cycles. … Nakao, K. (1998). Structure-­function studies of human leptin. Journal
Human Reproduction Update, 22(3), 320–341. of Biological Chemistry, 273(52), 35245–35249.
|
6 of 7       ZHANG and GONG

Iqbal, J., Pompolo, S., Considine, R. V., & Clarke, I. J. (2000). Localization of Margetic, S., Gazzola, C., Pegg, G. G., & Hill, R. A. (2002). Leptin: A review of
leptin receptor-­like immunoreactivity in the corticotropes, somatotro- its peripheral actions and interactions. International Journal of Obesity
pes, and gonadotropes in the ovine anterior pituitary. Endocrinology, and Related Metabolic Disorders, 26(11), 1407–1433.
141(4), 1515–1520. Messinis, I. E., & Milingos, S. D. (1999). Leptin in human reproduction.
Iqbal, J., Pompolo, S., Murakami, T., Grouzmann, E., Sakurai, T., Meister, Human Reproduction Update, 5(1), 52–63.
B., & Clarke, I. J. (2001). Immunohistochemical characterization of Mounzih, K., Lu, R., & Chehab, F. F. (1997). Leptin treatment rescues the
localization of long-­form leptin receptor (OB-­Rb) in neurochemically sterility of genetically obese ob/ob Males. Endocrinology, 138(3),
defined cells in the ovine hypothalamus. Brain Research, 920(1–2), 1190–1193.
55–64. Ozata, M., Ozdemir, I. C., & Licinio, J. (1999). Human leptin deficiency caused
Ishikawa, T., Fujioka, H., Ishimura, T., Takenaka, A., & Fujisawa, M. (2007). by a missense mutation: Multiple endocrine defects, decreased sympa-
Expression of leptin and leptin receptor in the testis of fertile and infer- thetic tone, and immune system dysfunction indicate new targets for
tile patients. Andrologia, 39(1), 22–27. leptin action, greater central than peripheral resistance to the effects of
Jope, T., Lammert, A., Kratzsch, J., Paasch, U., & Glander, H. J. (2003). Leptin leptin, and spontaneous correction of leptin-­mediated defects. Journal
and leptin receptor in human seminal plasma and in human spermato- of Clinical Endocrinology and Metabolism, 84(10), 3686–3695.
zoa. International Journal of Andrology, 26(6), 335–341. Quennell, J. H., Howell, C. S., Roa, J., Augustine, R. A., Grattan, D. R., &
Lampiao, F., & du Plessis, S. S. (2008). Insulin and leptin enhance human Anderson, G. M. (2011). Leptin deficiency and diet-­induced obesity reduce
sperm motility, acrosome reaction and nitric oxide production. Asian hypothalamic kisspeptin expression in mice. Endocrinology, 152(4), 1541.
Journal of Andrology, 10(5), 799–807. Roa, J., Garcíagaliano, D., Castellano, J. M., Gaytan, F., Pinilla, L., &
Landry, D., Cloutier, F., & Martin, L. J. (2013). Implications of leptin in neuro- Tenasempere, M. (2010). Metabolic control of puberty onset: New play-
endocrine regulation of male reproduction. Reproductive Biology, 13(1), ers, new mechanisms. Molecular & Cellular Endocrinology, 324(1–2), 87.
1–14. Roa, J., & Tena-Sempere, M. (2010). Energy balance and puberty onset:
Landry, D. A., Sormany, F., Haché, J., Roumaud, P., & Martin, L. J. (2017). Emerging role of central mTOR signaling. Trends in Endocrinology &
Steroidogenic genes expressions are repressed by high levels of leptin Metabolism, 21(9), 519–528.
and the JAK/STAT signaling pathway in MA-­10 Leydig cells. Molecular Rutters, F., Nieuwenhuizen, A. G., Verhoef, S. P. M., Lemmens, S. G. T.,
& Cellular Biochemistry, 433(1–2), 79–95. Vogels, N., & Westerterp-Plantenga, M. S. (2009). The relationship be-
Lebrethon, M. C., Vandersmissen, E., Gerard, A., Parent, A. S., & tween leptin, gonadotropic hormones, and body composition during
Bourguignon, J. P. (2000). Cocaine and amphetamine-­ regulated-­ puberty in a Dutch children cohort. European Journal of Endocrinology,
transcript peptide mediation of leptin stimulatory effect on the rat 160(6), 973–978.
gonadotropin-­releasing hormone pulse generator in vitro. Journal of Sainz, N., Barrenetxe, J., Moreno-Aliaga, M. J., & Martinez, J. A. (2015).
Neuroendocrinology, 12(5), 383–385. Leptin resistance and diet-­induced obesity: Central and peripheral ac-
Leshan, R. L., Louis, G. W., Jo, Y. H., Rhodes, C. J., Munzberg, H., & Myers, tions of leptin. Metabolism, 64(1), 35–46.
M. G. Jr (2009). Direct innervation of GnRH neurons by metabolic-­and Schaab, M., & Kratzsch, J. (2015). The soluble leptin receptor. Best Practice
sexual odorant-­sensing leptin receptor neurons in the hypothalamic ven- & Research Clinical Endocrinology & Metabolism, 29(5), 661–670.
tral premammillary nucleus. Journal of Neuroscience, 29(10), 3138–3147. Seminara, S. B., Messager, S., Chatzidaki, E. E., Thresher, R. R., Acierno, J. S.
Li, H. W. R., Chiu, P. C. N., Cheung, M. P. L., & Yeung, W. S. B. (2009). Effect Jr, Shagoury, J. K., … Colledge, W. H. (2003). The GPR54 gene as a regu-
of leptin on motility, capacitation and acrosome reaction of human lator of puberty. New England Journal of Medicine, 349(17), 1614–1627.
spermatozoa. International Journal of Andrology, 32(6), 687–694. Skorupskaite, K., George, Jyothis. T., & Anderson, Richard. A. (2014). The
Lin, Q., Poon, S. L., Chen, J., Cheng, L., Hoyuen, B., & Leung, P. C. (2009). kisspeptin-­GnRH pathway in human reproductive health and disease.
Leptin interferes with 3′,5′-­cyclic adenosine monophosphate Human Reproduction Update, 20(4), 485.
(cAMP) signaling to inhibit steroidogenesis in human granulosa cells. Smith, J. T., Acohido, B. V., Clifton, D. K., & Steiner, R. A. (2006). KiSS-­1
Reproductive Biology & Endocrinology, 7(1), 115. neurones are direct targets for leptin in the ob/ob mouse. Journal of
Luque, R. M., Kineman, R. D., & Tenasempere, M. (2007). Regulation of Neuroendocrinology, 18(4), 298–303.
Hypothalamic Expression of KiSS-­1 and GPR54 Genes by Metabolic Smith, J. T., & Waddell, B. J. (2003). Developmental changes in plasma leptin
Factors: Analyses Using Mouse Models and a Cell Line. Endocrinology, and hypothalamic leptin receptor expression in the rat: Peripubertal
148(10), 4601–4611. changes and the emergence of sex differences. Journal of Endocrinology,
Luukkaa, V., Pesonen, U., Huhtaniemi, I., Lehtonen, A., Tilvis, R., Tuomilehto, 176(3), 313–319.
J., … Huupponen, R. (1998). Inverse correlation between serum testos- Soyupek, S., Armagan, A., Serel, T. A., Hoscan, M. B., Perk, H., Karaoz, E.,
terone and leptin in men. Journal of Clinical Endocrinology & Metabolism, & Candir, O. (2005). Leptin expression in the testicular tissue of fertile
83(9), 3243. and infertile men. Archives of Andrology, 51(3), 239–246.
Ma, Y., Chen, B., Wang, H., Hu, K., & Huang, Y. (2011). Prediction of sperm Steinman, N., Gamzu, R., Yogev, L., Botchan, A., Schreiber, L., & Yavetz, H.
retrieval in men with non-­ obstructive azoospermia using artificial (2001). Serum leptin concentrations are higher in azoospermic than in
neural networks: Leptin is a good assistant diagnostic marker. Human normozoospermic men. Fertility and Sterility, 75(4), 821–822.
Reproduction, 26(2), 294–298. Strobel, A., Issad, T., Camoin, L., Ozata, M., & Strosberg, A. D. (1998). A
Manfredilozano, M., Roa, J., Ruizpino, F., Piet, R., Garciagaliano, D., Pineda, leptin missense mutation associated with hypogonadism and morbid
R., … Romeroruiz, A. (2016). Defining a novel leptin–melanocortin–kiss- obesity. Nature Genetics, 18(3), 213–215.
peptin pathway involved in the metabolic control of puberty. Molecular Sullivan, S. D., & Moenter, S. M. (2004). γ-­Aminobutyric acid neurons in-
Metabolism, 5(10), 844–857. tegrate and rapidly transmit permissive and inhibitory metabolic cues
Mantzoros, C. S., Flier, J. S., & Rogol, A. D. (1997). A longitudinal assessment to gonadotropin-­ releasing hormone neurons. Endocrinology, 145(3),
of hormonal and physical alterations during normal puberty in boys. V. 1194–1202.
Rising leptin levels may signal the onset of puberty. Journal of Clinical Swerdloff, R. S., Batt, R. A., & Bray, G. A. (1976). Reproductive hormonal
Endocrinology and Metabolism, 82(4), 1066–1070. function in the genetically obese (ob/ob) mouse. Endocrinology, 98(6),
Maqsood, A. R., Trueman, J. A., Whatmore, A. J., Westwood, M., Price, D. 1359–1364.
A., Hall, C. M., & Clayton, P. E. (2007). The relationship between noc- Tartaglia, L. A., Dembski, M., Weng, X., Deng, N., Culpepper, J., Devos, R.,
turnal urinary leptin and gonadotrophins as children progress towards … Tepper, R. I. (1995). Identification and expression cloning of a leptin
puberty. Hormone Research, 68(5), 225–230. receptor, OB-­R. Cell, 83(7), 1263–1271.
ZHANG and GONG |
      7 of 7

Tena-Sempere, M., & Barreiro, M. L. (2002). Leptin in male reproduction: Wauman, J., Zabeau, L., & Tavernier, J. (2017). The leptin receptor complex:
The testis paradigm. Molecular and Cellular Endocrinology, 188(1–2), Heavier than expected? Frontiers in Endocrinology, 8, 30.
9–13. Yong, X., Faulkner, L. D., & Hill, J. W. (2011). Cross-­talk between metabo-
Tena-Sempere, M., Manna, P. R., Zhang, F. P., Pinilla, L., Gonzalez, L. C., lism and reproduction: The role of POMC and SF1 neurons. Frontiers in
Dieguez, C., … Aguilar, E. (2001). Molecular mechanisms of leptin ac- Endocrinology, 2, 98.
tion in adult rat testis: Potential targets for leptin-­induced inhibition of Yu, W. H., Kimura, M., Walczewska, A., Karanth, S., & McCann, S. M. (1997).
steroidogenesis and pattern of leptin receptor messenger ribonucleic Role of leptin in hypothalamic-­pituitary function. Proceedings of the
acid expression. Journal of Endocrinology, 170(2), 413–423. National Academy of Sciences, 94(3), 1023–1028.
Tena-Sempere, M., Pinilla, L., Gonzalez, L. C., Dieguez, C., Casanueva, F. F., Zamorano, P. L., Mahesh, V. B., De Sevilla, L. M., Chorich, L. P., Bhat, G. K., &
& Aguilar, E. (1999). Leptin inhibits testosterone secretion from adult Brann, D. W. (1997). Expression and localization of the leptin receptor
rat testis in vitro. Journal of Endocrinology, 161(2), 211–218. in endocrine and neuroendocrine tissues of the rat. Neuroendocrinology,
Tezuka, M., Irahara, M., Ogura, K., Kiyokawa, M., Tamura, T., Matsuzaki, T., 65(3), 223.
… Aono, T. (2002). Effects of leptin on gonadotropin secretion in juve- Zhang, Y., Proenca, R., Maffei, M., Barone, M., Leopold, L., & Friedman, J.
nile female rat pituitary cells. European Journal of Endocrinology, 146(2), M. (1994). Positional cloning of the mouse obese gene and its human
261–266. homologue. Nature, 372(6505), 425–432.
Thompson, E. L., Patterson, M., Murphy, K. G., Smith, K. L., Dhillo, W. S., Zorn, B., Osredkar, J., Meden-Vrtovec, H., & Majdic, G. (2007). Leptin lev-
Todd, J. F., … Bloom, S. R. (2004). Central and peripheral administration els in infertile male patients are correlated with inhibin B, testosterone
of kisspeptin-­10 stimulates the hypothalamic-­pituitary-­gonadal axis. and SHBG but not with sperm characteristics. International Journal of
Journal of Neuroendocrinology, 16(10), 850–858. Andrology, 30(5), 439–444.
Van Vugt, D. A., Lujan, M. E., Froats, M., Krzemien, A., Couceyro, P. R., &
Reid, R. L. (2006). Effect of fasting on cocaine-­amphetamine-­regulated
transcript, neuropeptide Y, and leptin receptor expression in the non-­
How to cite this article: Zhang J, Gong M. Review of the role
human primate hypothalamus. Neuroendocrinology, 84(2), 83–93.
of leptin in the regulation of male reproductive function.
Venancio, J. C., Margatho, L. O., Rorato, R. C., Rosales, R. R. C., Debarba, L.
K., Coletti, R., , & Elias, L. L. K. (2017). Short-­term high-­fat diet ­increases Andrologia. 2018;e12965. https://doi.org/10.1111/and.12965
leptin activation of cart neurons and advances puberty in female mice.
Endocrinology, 158(11), 3929–3942.

You might also like