You are on page 1of 9

YNPEP-01701; No of Pages 9

Neuropeptides xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Neuropeptides

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

Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in
the hypothalamic–pituitary–gonadal (HPG) axis and peripheral organs
(fat, pancreas and liver) in male rats
M. Dudek a, P.A. Kołodziejski b, E. Pruszyńska-Oszmałek b, M. Sassek b, K. Ziarniak a,
K.W. Nowak b, J.H. Sliwowska a,⁎
a
Laboratory of Neurobiology, Institute of Zoology, Poznan University of Life Sciences, Wojska Polskiego 71C, 60-625 Poznan, Poland
b
Department of Animal Physiology and Biochemistry, Poznan University of Life Sciences, Wołynska 33, 60-625 Poznan, Poland

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

Article history: Recent data indicates that kisspeptin, encoded by the KISS1 gene, could play a role in transducing metabolic in-
Received 24 June 2015 formation into the hypothalamic–pituitary–gonadal (HPG) axis, the mechanism that controls reproductive func-
Received in revised form 12 December 2015 tions. Numerous studies have shown that in a state of negative energy balance, the hypothalamic kisspeptin
Accepted 20 January 2016
system is impaired. However, data concerning positive energy balance (e.g. diabetes and obesity) and the role
Available online xxxx
of kisspeptin in the peripheral tissues is scant.
Keywords:
We hypothesized that: 1) in diet-induced obese (DIO) male rats and/or rats with diabetes type 1 (DM1) and type
Obesity 2 (DM2), altered reproductive functions are related to an imbalance in Kiss1 and GPR54 mRNA in the HPG axis;
Diabetes and 2) in DIO and/or DM1 and/or DM2 rats, Kiss1 and GPR 54 expression are altered in the peripheral tissues in-
Hypothalamic–pituitary–gonadal (HPG) axis volved in metabolic functions (fat, pancreas and liver).
Fat Animals were fed a high-fat or control diets and STZ (streptozotocin — toxin, which destroys the pancreas) was
Pancreas injected in high or low doses to induce diabetes type 1 (DM1) or diabetes type 2 (DM2), respectively. RT-PCR and
Liver Western blot techniques were used to assess the expression of Kiss1 and GRP54 in tissues.
Kiss1
At the level of mRNA, we found that diabetic but not obese rats have alterations in Kiss1 and/or GPR54 mRNA
GPR54
levels in the HPG axis as well as in peripheral tissues involved in metabolic functions (fat, pancreas and liver).
The most severe changes were seen in DM1 rats. However, in the case of protein levels in the peripheral tissues
(fat, pancreas and liver), changes in Kiss1/GPR54 expression were noticed in DIO, DM1 and DM2 animals and
were tissue-specific.
Our data support the hypothesis that alterations in Kiss1/GPR54 balance may account for both reproductive and
metabolic abnormalities reported in obese and diabetic rats.
© 2016 Elsevier Ltd. All rights reserved.

1. Introduction Kisspeptins (encoded by the KISS1 gene), acting upstream of GnRH,


are potent stimulators of the HPG axis in a number of species, including
Reproductive function in vertebrates is regulated by a cascade of humans (Caraty et al., 2010; Dhillo et al., 2007; Gottsch et al., 2004;
events organized in the hypothalamic–pituitary–gonadal axis (HPG). Irwig et al., 2004; Navarro et al., 2004; Shahab et al., 2005). Both in
The hypothalamus, situated at the top of the axis, synthetizes and humans and mice, mutations in the KISS1 receptor (GPR54 or KISS1R)
secretes gonadotropin releasing hormone (GnRH), which stimulates were associated with hypogonadotropic hypogonadism, a condition
the pituitary to secrete gonadotropins: luteinizing hormone (LH) and characterized by low sex steroids and gonadotropin levels (de Roux
follicle stimulating hormone (FSH) (Fink, 2000; Schwartz, 2000). et al., 2003; Seminara et al., 2003). Importantly, perturbations in energy
Gonadotropins, in turn, stimulate gonads to produce gametes and balance, including negative energy balance (e.g. undernutrition) or pos-
synthetize and release sex steroid hormones, which act on the brain, an- itive energy balance (e.g. obesity and diabetes) frequently result in im-
terior pituitary and peripheral organs (Hotchkiss and Knobil, 1994). pairments of fertility (Pasquali et al., 2007; Wahab et al., 2015) and
hypothalamic hypogonadotropic hypogonadism was reported in obese
and diabetic patients (Skorupskaite et al., 2014). Moreover, in men,
⁎ Corresponding author. low serum testosterone (T) and lower LH pulse frequency were often
E-mail addresses: monika.dudeekk@gmail.com (M. Dudek), pawelbigi@o2.pl
(P.A. Kołodziejski), ewaprusz@up.poznan.pl (E. Pruszyńska-Oszmałek),
associated with obesity and diabetes mellitus type 2 (DM2) (Al Hayek
macsass@up.poznan.pl (M. Sassek), kamilziarniak@gmail.com (K. Ziarniak), et al., 2013; Costanzo et al., 2014; Ding et al., 2006; Ford and Liu,
kwnowak@up.poznan.pl (K.W. Nowak), joanna.sliwowska@gmail.com (J.H. Sliwowska). 2005; George et al., 2013; Glass et al., 1977; Lakshman et al., 2010;

http://dx.doi.org/10.1016/j.npep.2016.01.005
0143-4179/© 2016 Elsevier Ltd. All rights reserved.

Please cite this article as: Dudek, M., et al., Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the
hypothalamic–pituitary–gonadal (HPG..., Neuropeptides (2016), http://dx.doi.org/10.1016/j.npep.2016.01.005
2 M. Dudek et al. / Neuropeptides xxx (2016) xxx–xxx

Stellato et al., 2000; Wu et al., 2008). As metabolic status is important for insulin secretion in the presence of glucose in mice and humans
reproduction, it would be reasonable to assume that kisspeptin can pro- (Hauge-Evans et al., 2006). The administration of kisspeptin causes
vide a link between metabolism and fertility. Indeed, kisspeptin-10 ad- rapid glucose-induced insulin secretion in rats' islets, with greater re-
ministration restored LH and T secretion in hypotestosteronaemic men sponse to KP-10 compared to KP-13 (Bowe et al., 2012). It is also well
with DM2 (George et al., 2013). documented that insulin has a direct effect on glucagon secretion,
Similarly to studies in humans, in the streptozotocin (STZ)-induced both in control animals and humans with DM1 and DM2 (D'Alessio,
(STZ — toxin, which destroys pancreas cells) rat model of diabetes type 2011; Dumonteil et al., 1998; Kawamori and Kulkarni, 2009;
1 (DM1), reduced T and LH as well as testis and prostate weights were Kawamori et al., 2009; Muller et al., 1973). In vitro studies on mice's pri-
found (Castellano et al., 2006; Szkudelski, 2001). Moreover, in male rats mary hepatocytes have shown that kisspeptin expression was stimulat-
with STZ-induced diabetes, Kiss1 mRNA levels decreased and the ed by glucagon (Song et al., 2014). In addition, in vivo experiments in
postorchidectomy rise in Kiss1 mRNA was blunted. Importantly, repeat- mice, both controls and those lacking glucagon receptors in hepato-
ed administration of kisspeptin rescued the LH and T responses and in- cytes, have revealed that treatment by glucagon stimulates kisspeptin
creased testis and prostate weights (Castellano et al., 2006). Thus, it is expression, but only in control animals (Song et al., 2014). Finally, the
suggested that altered expression and function of the hypothalamic role of the kisspeptin system in metabolic function has been suggested
Kiss1 system is involved in the etiopathogenesis of hypogonadotropism by recent findings from studies in Kiss1r KO mice, which lack functional
(Castellano et al., 2005). kisspeptin signaling. Results from Kiss1r KO mice fed with both stan-
On the other hand, data concerning Kiss1 expression in a state of dard chow diet and HFD indicate that aside from stimulating the HPG
obesity depends on animal models. In high-fat diet (HFD)-induced axis, the kisspeptin system is also an important player in body weight,
obese rats, elevation of hypothalamic Kiss1 mRNA was reported energy balance, locomotion, and glucose regulation (Tolson et al.,
(Brown et al., 2008). In female DBA/2J mice that were fed HFD and 2014). Thus, the presence of the tri-hormonal circuit linking kisspeptin,
that were highly susceptible to obesity-induced infertility, a decrease islets, and liver in the regulation of glucose, glucagon and insulin secre-
in the hypothalamic Kiss1 mRNA and the number of kisspeptin- tion was proposed/see review (Hussain et al., 2015).
immunoreactive (-ir) cells was found (Quennell et al., 2011). By con- In light of the above findings, we tested the hypothesis that alter-
trast, in C57BL/6J mice fed a HFD there was no change in Kiss1 mRNA ations in the HPG axis are related to imbalanced levels of Kiss1 and
in the hypothalamus (Quennell et al., 2011). In genetically obese male GPR54 mRNA in diet-induced obese (DIO) male rats and/or rats with di-
Zucker rats (Brown et al., 2008) and in leptin deficient female ob/ob abetes type 1 (DM1) and diabetes type 2 (DM2) compared to control. As
mice (genetic model of DM2) a decrease in hypothalamic Kiss1 mRNA Kiss1 and GPR54 are also expressed in the peripheral tissues involved in
was found (Quennell et al., 2011; Smith et al., 2006). But there is a metabolic functions, which are deregulated in obesity and diabetes, we
lack of data on Kiss1 and GPR54 expression in animal models of DM2 in- evaluated whether Kiss1 and GPR54 expression in fat, the pancreas and
duced by HFD and STZ injections. Importantly, DM2, which accounts for the liver are altered in DIO, DM1 and DM2 male rats.
90% of diabetes cases reported around the world, is often associated
with obesity (Mendis et al., 2015). As obesity and diabetes rates surge 2. Materials and methods
across the globe, the WHO recognized both obesity and diabetes as
our biggest public health problem. Of particular interest for the current 2.1. Experimental animals
paper, in Eastern Europe and in the United States, about 50% of women
are overweight (Mendis et al., 2015). For this reason, we see a need to Adult male Wistar rats (200 ± 30 g, 10 week old age) were pur-
gain more knowledge about the integration of metabolic and reproduc- chased from Animal Breeding (Brwinów, Poland) and given 1 week of
tive systems both in healthy organisms as well as in cases of obesity and acclimatization. All protocols and experimental procedures were ap-
diabetes. proved by the Local Ethics Commission for Investigation on Animals,
Of particular interest for this paper, it is also suggested that Poznań University of Life Sciences (Poland).
kisspeptin has additional roles outside of reproduction — possibly in-
volving regulation of metabolic functions. Besides the hypothalamus 2.2. Inductions of obesity and diabetes states in rats
(Lehman et al., 2013), Kiss1/GPR54 are expressed in peripheral tissues
including those in the pituitary, gonads, fat, liver, and pancreas Male Wistar rats were housed in standard cages for rats and main-
(Bellingham et al., 2009; Castellano et al., 2006; Cockwell et al., 2013; tained under controlled conditions (room temperature 21 °C ± 1 °C;
Gutierrez-Pascual et al., 2007; Hauge-Evans et al., 2006; Quennell constant humidity with 12/12-h light–dark cycle). After 1 week of accli-
et al., 2010) and metabolic status influences Kiss1 expression peripher- matization, animals were divided into four groups: 1) control group (C;
ally. Kiss1 mRNA in the pituitary was elevated in obese Zucker male rats, n = 8) — fed a regular chow diet (Labofed B, Morawski Kcynia,
but in male rats on HFD it was unchanged (Brown et al., 2008). In HFD- Poland); 2) type 1 diabetes group (DM1; n = 8) — fed regular chow
induced obese female rats, ovarian Kiss1 and kisspeptin were down- (Labofed B, Morawski Kcynia, Poland); 3) diet induced obesity group
regulated (Zhou et al., 2014). But there is no study on Kiss1/GPR54 ex- (DIO; n = 8) — fed a high-fat diet (HFD) consisting of 50% energy from
pression in the pituitary and testes of diabetic rats. fat (Morawski Kcynia, Poland); and 4) type 2 diabetes group (DM2;
Kiss1 mRNA has been detected in rat adipose tissue (Brown et al., n = 8) — fed a high-fat diet (HFD) consisting of 50% energy from fat
2008; Cockwell et al., 2013) and food restriction (18 h) increased (Morawski, Kcynia, Poland). Animals received diets for 6 weeks ad
Kiss1 mRNA in the fat of both male and female rats (Brown et al., libitum and had water access.
2008). In contrast, Kiss1 mRNA was reduced in obese HFD-fed and In order to induce diabetes, rats fed a regular chow diet were
obese Zucker rats. In women, a positive correlation between Kiss1 injected intraperitoneally (i.p.) with streptozotocin — STZ (DM1
mRNA and BMI (body mass index) was reported in the visceral (omen- group). Rats were first injected with STZ (70 mg/kg body weight in citric
tal) adipose tissue (Cockwell et al., 2013). In an experiment in which a buffer; pH = 4.4) followed by a second injection of STZ (30 mg/kg body
rat's arcuate nucleus of the hypothalamus (ARC) was damaged, hypo- weight). Development of the diabetic phenotype was monitored, mea-
thalamic Kiss1 mRNA was reduced, but Kiss1 mRNA in the fat was unaf- suring fasting (12 h) glucose levels in blood taken from the tail tip
fected. This suggests that regulation of Kiss1 mRNA in the adipose tissue (using a Roche AccuCheck Active device, range 10–600 mg/dl). Serum
is independent of the hypothalamus (Brown et al., 2008). levels of insulin after overnight (12 h) fasting were determined.
Kisspeptin and its receptor are also expressed in the pancreas, and After 6 weeks on HFD, a single dose of STZ (25 mg/kg body weight)
pancreatic islet function may be dependent on kisspeptin signaling was administered to induce diabetes (mimicking early stage of DM2).
(Hauge-Evans et al., 2006; Kotani et al., 2001). Kisspeptin can regulate These animal models are well characterized and commonly used

Please cite this article as: Dudek, M., et al., Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the
hypothalamic–pituitary–gonadal (HPG..., Neuropeptides (2016), http://dx.doi.org/10.1016/j.npep.2016.01.005
M. Dudek et al. / Neuropeptides xxx (2016) xxx–xxx 3

(Reed et al., 2000; Szkudelski, 2001; Vater et al., 2013; Zhang et al., Germany) on a Versa Doc scanner (BioRad, USA). β-actin was used as
2008). Fasting (overnight) glucose and insulin levels were assessed. a reference protein (anti β-actin antibody, Sigma-Aldrich, Germany).
Plasma insulin levels were measured using a radioimmunoassay
(RIA) kit (Rat Insulin RIA, RI-13K, Merck, Millipore, Darmstadt, 2.5. Statistics
Germany), with sensitivity of 0.1 ng/ml; inter-assay: 8.5–9.4; and intra-
assay: 1.4–4.6. Body weight, glucose and insulin serum levels were presented as
Additionally, animals from all groups were weighed at termination, mean ± SEM (Graph Pad Prism, GraphPad Software, Inc., USA). RT
two weeks after their last STZ-injections. Animals were decapitated PCR and Western blot data was presented as percentage of control
and hypothalami, pituitaries, testes, gonadal fat, pancreas and livers levels. Statistical analysis was performed using ANOVA on body weight,
were dissected and immediately frozen in liquid nitrogen and stored glucose and insulin on the RT PCR and Western blot results to explore
at −80 °C for further analysis. group effects (groups: DIO — obese, DM1 — diabetes type 1, DM2 — di-
abetes type 2 and C — control). This was followed by a Fisher post hoc
2.3. Semiquantitive real time PCR test (Statistica, StatSoft, Poland). Statistical significance was accepted
at p b 0.05.
Total RNA was extracted using Tripure Reagent Roche according to
the manufacturer's instructions. cDNA was generated from 1 μg using 3. Results
a commercial Transcriptor First Strand cDNA Synthesis Kit (Cat. #
04,597,030,001, Roche). Real-time PCR was performed with gene- 3.1. The impact of a high-fat diet and streptozotocin-induced types 1 and 2
specific intron spanning primers (Table 1). Each real-time PCR reaction diabetes on body weight and metabolic parameters in rats
containing Fast SYBR® Green Master Mix (Cat.# 1,406,113, Applied
Biosystems), 3 μl of cDNA diluted 1:10 and 3 μl of primer's mix was car- Body weight data and metabolic parameters (glucose and insulin
ried out in a QuantStudio 12K Flex™ Real-Time PCR system (Applied serum levels) are presented in Fig. 1. There was a group effect on body
Biosystems). Results were normalized with a glyceraldehyde-3- weight (p = 0.000001). DIO rats were significantly heavier compared to
phosphate dehydrogenase (GADPH) reference gene. The sequence of C rats (means: DIO: 734.3 ± 36.54 vs. C: 597.5 ± 26.6; p b 0.005,
primers and product size have been included in Table 1. Fig. 1A). On the other hand, DM1 rats had a reduction in body weight
compared to C, DIO and DM2 animals (means: DM1: 490.1 ± 19.47 vs.
2.4. Western blots C: 597.5 ± 26.6; p b 0.01; DM1: 490.1 ± 19.47 vs. DIO: 734.3 ± 36.54;
p b 0.00001 and DM1: 490.1 ± 19.47 vs. DM2 743 ± 23.87;
2.4.1. Preparation of samples p b 0.000001). Additionally, DM2 males had a higher body weight com-
Protein isolation was performed by tissue homogenization in a RIPA pared to C (means: DM2: 743 ± 23.87 vs. C: 597.5 ± 26.6; p b 0.001),
buffer (25 mM Tris–HCl pH 7.6, 150 mM NaCl, 5 mM EDTA, 1% NP-40 or Fig. 1A.
1% Triton X-100, 1% sodium deoxycholate, 0.1% SDS) supplemented There was a group effect on fasting glucose serum levels
with a protease inhibitor cocktail (Roche Diagnostics, Penzberg, (p b 0.0000001). As predicted, DM1 animals had higher levels of glucose
Germany). Then samples (n = 4/tissue) were centrifuged for 10 min compared to C, DIO and DM2 males (all p's b 0.0000001, Fig. 1B). There
and 10,000 g of intermediate phase protein was collected and stored was an approximate 40% increase in glucose levels in DM2 compared to
at −80 °C. C rats, (p = 0.18, trend).
Moreover, as predicted, there was a group effect on insulin fasting
2.4.2. Electrophoresis and protein transfer serum levels (p b 0.000005). Serum insulin levels in the DM1 group
20 μg of total protein per lane (for pancreas and liver) and 15 μg of fat were the lowest (mean: 0.6735 ± 0.15 mg/dl) compared to C and
were mixed 1:3 with β-mercaptoethanol supplemented Laemmli Buffer other experimental groups (DM1 vs. C; p b 0.005; DM1 vs. DIO;
(Biorad, USA) and denatured for 5 min at 95 °C. Electrophoresis was p b 0.0005; DM1 vs. DM2; p b 0.0000001, Fig. 1C). On the other hand,
performed in 10% polyacrylamide gel at 150 V. Then proteins were in DM2 males, serum insulin levels were the highest (mean: 3.601 ±
transferred to the nitrocellulose membrane. Non-specific binding to 0.14 mg/dl) compared to C and other experimental groups (all
membranes was blocked using 3% BSA in a TBST solution (20 mM Tris p's N 0.01).
base, 150 mM NaCl, 0.15% Tween20) for 1 h at room temperature.
3.2. Kiss1 and GRP54 mRNA in the HPG axis of control (C), obese (DIO), and
2.4.3. Western blots diabetes type 1 (DM1) type 2 (DM2) rats
Membranes were incubated overnight at 4 °C with primary antibod-
ies, rabbit anti-kisspeptin AC564 (INRA, Physiologie de la Reproduction There was no difference in hypothalamic Kiss1 mRNA levels among
et des Comportements, Nouzilly, France) and rabbit anti-GPR54 (cat No the groups (p N 0.356; Fig. 2A). But there was a group effect on GPR54
254512, ABBIOTEC, San Diego, USA) (1:1000 dilution), in a TBST solu- mRNA (p b 0.0003), with an increase in DM1 and DM2 compared to C
tion supplemented with 1% BSA. The primary kisspeptin and GPR anti- rats (p b 0.05 and p b 0.0001, respectively, Fig. 2B). The highest levels
bodies were previously used (Desroziers et al., 2010; Guimiot et al., of GPR54 mRNA were found in the DM2 group (about 26% over the con-
2012; Zhou et al., 2014). Membranes were washed 3 times for 10 min trol value). In DM2 rats, GPR54 mRNA was also higher compared to
in a TBST buffer and then incubated with anti-rabbit secondary antibody DM1 animals (p b 0.05).
for 1 h at room temperature (1:5000, # OABP 01378, Aviva Systems San In the pituitary, there was no group effect on Kiss1 mRNA and GPR54
Diego, USA). Signals were visualized using Amersham ECL Prime West- mRNA (p = 0.09 and p = 0.172, respectively) (Fig. 2C and D).
ern Blotting Detection Reagent (GE Healthcare Europe, Freiburg, The biggest changes within the HPG axis were found in the testes,
with a group effect on Kiss1 mRNA (p b 0.01). In DM1 rats, Kiss1
mRNA in testes was about 14% higher compared to C (p b 0.005),
Table 1
Sequence of primers and product size used in the study. about 7% higher compared to DIO and about 19% higher compared to
DM2 rats (p b 0.0005). In DM2 animals, Kiss1 mRNA was also about
Gene Left primer (5′˃3′) Right primer (5′˃3′) Product size
12% lower compared to DIO rats (p b 0.05). In the testes there was
KISS1 atgatctcgctggcttcttg ccaggcattaacgagttcct 141 also a group effect on GPR54 mRNA (p = 0.0027). In general, in DM1 an-
GPR54 cttcatgtgcaaattcgtcaa aagtggcacatgtggcttg 62 imals GPR54 mRNA was the lowest of all the groups (about 30% lower
Gapdh ctgcaccaccaactgcttag tgatggcatggactgtgg 92
compared to C, 58% lower compared to DM2 and 42% lower compared

Please cite this article as: Dudek, M., et al., Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the
hypothalamic–pituitary–gonadal (HPG..., Neuropeptides (2016), http://dx.doi.org/10.1016/j.npep.2016.01.005
4 M. Dudek et al. / Neuropeptides xxx (2016) xxx–xxx

Fig. 1. Body weight (A), serum glucose (B) and insulin (C) levels in control (C), obese (DIO) and diabetes type 1 (DM1) and type 2 (DM2) rats. (A): Body weight: a: DIO N C, (p b 0.005); b:
DM1 b C, (p b 0.01); c: DM1 b DIO, (p b 0.00001); d: DM1 b DM2, (p b 0.0000001); e: DM2 N C, (p b 0.001). (B): Glucose levels: a: DM1 N C, (p b 0.01); b: DM1 N DIO, (p b 0.00001); c:
DM1 N DM2, (p b 0.000001); d: DM2 N C, (p = 0.18) (C): Insulin levels: a: DM2 N C, (p b 0.01); b: DM2 N DIO, (p b 0.001); c: DM2 N DM1, (p b 0.0000001); d: DM1 b C, (p b 0.005); e:
DM1 b DIO, (p b 0.0005).

to DIO males; p's: p b 0.01, p b 0.00005, p b 0.001, respectively). On the p b 0.05, respectively); Fig. 3 C. We have also discerned a group effect
other hand, in DM2 males GPR54 mRNA was about 28% higher com- on GPR54 mRNA (p = 0.0413). There was a 450% increase in GPR54
pared to C animals (p b 0.05) (Fig. 2E and F). mRNA in DM1 compared to C (p b 0.05), about 480% compared to DIO
(p b 0.05), and about 530% compared to DM2 rats (p b 0.05); Fig. 3 D.
3.3. Kiss1 and GRP54 mRNA in the peripheral organs (fat, pancreas, liver) of In contrast to the fat and the pancreases, there was no group effect
control (C), obese (DIO), and diabetes type 1 (DM1) and diabetes type 2 on Kiss1 mRNA in the liver (p = 0.239). However, an inspection of
(DM2) rats Fig. 3 indicates a tendency toward an increase in DIO, DM1 and DM2 an-
imals compared to controls (p = 0.066). Additionally, in the liver, there
Kiss1 and GPR54 mRNA have been found in fat, pancreas and liver tis- was a group effect on GPR54 mRNA (p = 0.0084), with the levels of the
sues. There was a group effect on Kiss1 mRNA in the fat, with a dramatic receptor being highest in DM1 animals compared to controls and all
increase in Kiss1 mRNA in DM1 males (p = 0.00097) compared to con- other experimental groups. There was an approximate 860% increase
trols and all other experimental groups (Fig. 3A). In DM1, Kiss1 mRNA in GPR54 mRNA compared to C (p b 0.005), about 770% increase com-
was 1000% higher compared to C, DIO and DM2 rats (p b 0.0005, pared to DIO (p b 0.01), and an approximate 790% increase compared
p b 0.001 and p b 0.0005, respectively). Similarly to Kiss1 mRNA, there to DM2 males (p b 0.01); Fig. 3E and F.
was also a group effect on GPR54 mRNA in the fat (p = 0.014), with
the highest levels reported in DM1 males. These had an approximate 3.4. Kisspeptin and GRP54 levels in the peripheral organs (fat, pancreas,
400% increase compared to C (p b 0.005), about 200% compared to DIO liver) of control (C), obese (DIO), and diabetes type 1 (DM1) and diabetes
(p b 0.05) and about 400% compared to DM2 rats (p b 0.01); Fig. 3B. type 2 (DM2) rats
In the pancreas, there was a group effect on Kiss1 mRNA (p =
0.0273), with Kiss1 mRNA being about 190% higher in DM1 and about Kisspeptin and GPR54 were expressed in fat, the pancreas and liver,
130% higher in DM2 compared to control animals (p b 0.005 and with the lowest levels observed in the fat. There was a group effect on

Fig. 2. Effects of diet-induced obesity (DIO) and STZ-induced diabetes type 1 and type 2 in male rats on Kiss1 mRNA (A, C, E) and GPR54 mRNA (B, D, F) levels in the hypothalamus (A, B), the
pituitary (C, D) and the testes (E, F). mRNA values were determined by semi quantitative real-time RT-PCR and are expressed as percentage changes compared to controls fed ad libitum
(mean ± S.E.M.). Letters represent the difference among groups. C — control; DIO — obese rats; DM1 — rats with diabetes type 1; DM2 — rats with diabetes type 2. (A–B): Hypothalamus:
(A) (p N 0.356); (B) a: DM1 N C (p b 0.05), b: DM2 N C (p b 0.0001); c: DM2 N DM1 (p b 0.05). (C–D): Pituitary: (C) (p = 0.09) and (D) (p = 0.172); (E–F) Testes: (E) a: DM1 N C,
(p b 0.01); b: DM1 N DM2, (p b 0.005); c: DM2 b DIO, (p b 0.05); (F): a: DM1 b C, (p b 0.01); b: DM1 b DIO, (p b 0.001); c: DM2 N C, (p b 0.05); d: DM1 b DM2, (p b 0.00005).

Please cite this article as: Dudek, M., et al., Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the
hypothalamic–pituitary–gonadal (HPG..., Neuropeptides (2016), http://dx.doi.org/10.1016/j.npep.2016.01.005
M. Dudek et al. / Neuropeptides xxx (2016) xxx–xxx 5

Fig. 3. Effects in male rats of diet-induced obesity (DIO) and STZ-induced type 1 and type 2 diabetes on Kiss1 mRNA 1 (A, C, E) and GPR54 (B, D, F) mRNA levels in adipose tissue (fat) (A, B),
the pancreas (C, D) and the liver (E, F). mRNA values were determined by semi quantitative real-time RT-PCR and are expressed as percentage changes compared to controls fed ad libitum
(mean ± S.E.M.). Letters represent the difference amongt groups. C — control; DIO — obese rats; DM1 — rats with diabetes type 1; and DM2 — rats with diabetes type 2. (A-B): Fat: (A): a:
DM1 N C, (p b 0.0005), b: DM1 N DIO (p b 0.001); c: DM1 N DM2 (p b 0.0005). (B): a: DM1 N C, (p b 0.005); b: DM1 N DOI, (p b 0.05); c: DM1 N DM2, (p b 0.01). (C–D): Pancreas: (C): a:
DM1 N C, (p b 0.005); b: DM2 N C, (p b 0.05); (D): a: DM1 N C (p b 0.05), b: DM1 N DIO (p b 0.05), c: DM1 N DM2 (p b 0.05). (E–F) Liver: (E):(p = 0.239) (F): a: DM1 N C, (p b 0.005); b:
DM1 N DIO, (p b 0.01); c: DM1 N DM2, (p b 0.01).

kisspeptin levels in the fat (p b 0.0000001), and we noted that in the DIO and liver) in diabetic but not in obese male rats. The most severe alter-
and DM2 groups, kisspeptin was beyond detectible levels (Fig. 4A). Ad- ations were present in STZ-induced DM1 male rats. Importantly, the
ditionally, in the fat we reported a 58% decrease in kisspeptin levels in analysis of proteins for kisspeptin and GPR54 in fat, the pancreas and
DM1 rats compared to controls (p b 0.00005). Similarly to kisspeptin, the liver has shown that differences were group- and tissue-specific.
we were unable to detect GPR54 in DIO and DM2 animals in the fat Thus, we proposed that: 1) alterations in Kiss1/GPR54 balance in the hy-
and there was no difference between levels of this peptide in DM1 com- pothalamus and testes may contribute to reproductive impairments ob-
pared to C groups (p = 0.100, Fig. 4B). served in these animals; and 2) alterations in kisspeptin/GPR54 balance
In the pancreas, there was a group effect on kisspeptin levels (p = in peripheral tissues involved in metabolic functions (fat, liver and pan-
0.000015), with kisspeptin being about 30% lower both in DIO and creas) may contribute to impaired regulation of glucose-stimulated in-
DM2 compared to C groups (p's b 0.05, Fig. 4C). In DM1, levels of sulin secretion observed in obese and diabetic rats.
kisspeptin were undetectable. There was also a group effect on GPR54 The lack of changes we noted in Kiss1 mRNA in the hypothalamus of
(p = 0.000356), with the levels of the receptor being lower in DIO com- male rats with diabetes (two weeks after STZ injections) compared to
pared to C rats (p b 0.05). Moreover, in the DM1 group, GPR54 levels controls is in agreement with data from short-term diabetic male rats
were below detectable values (Fig. 4D). (one week after STZ injection) (Castellano et al., 2009). Moreover,
Finally, in the liver, there was a group effect on kisspeptin levels using immunocytochemistry in the above-mentioned model, we also ob-
(p = 0.036633), with an increase (about 50%) in levels of kisspeptin served no differences among the groups in the number of kisspeptin-ir
in DM2 compared to C rats (p b 0.05) (Fig. 4E). A similar trend was cells in the ARC (in preparation). However, both in long-term diabetic
also observed in DIO animals (p = 0.053). Additionally, kisspeptin males and females (four weeks after STZ injections), a decrease in
levels were lower in DM1 compared to DIO and DM2 rats (p's b 0.05). Kiss1 mRNA in the hypothalamus was found (Castellano et al., 2006,
However, in the liver, there was no statistically significant difference 2009). Thus, it seems that the difference between results may be related
in GPR54 levels between the groups (p = 0.453820; Fig. 4F). to the animal model of diabetes (e.g. use of different doses of STZ, timing
after injections, severity of disease). Castellano et al. (2006) found that
4. Discussion diabetic rats (DM1) had glucose levels of approximately 600 mg/dl,
and in our study, glucose levels were about 393 mg/dl. It is also worth
Based on our understanding, this is the first comprehensive study of mentioning that in both Castellano's and our study, the RT PCR technique
the expression of Kiss1 and GPR54 in HPG axis, which control reproduc- was used on entire hypothalamic fragments, which made it impossible
tive functions and peripheral tissues (fat, pancreas, liver), which control to distinguish between the two different nuclei where Kiss1 is
metabolism in obese and diabetic (DM1 and DM2) rats. We have con- expressed: the anteroventral periventricular nucleus (AVPV) and the ar-
firmed the hypothesis that diabetic but not obese male rats have alter- cuate nucleus (ARC).
ations in Kiss1 and/or GPR54 mRNA in the HPG axis. Within the HPG Similarly to Luque et al. (2007), in their study of obese male C57BL/6J
axis, the biggest changes were revealed in the testes and small but sig- mice fed a HFD for 16 weeks, and to the Quennell study of C57BL/6J
nificant alterations in the hypothalamus. However, in agreement with HFD-fed female mice (2011), we did not find differences in hypotha-
previous findings in rats (Brown et al., 2008), we did not find changes lamic Kiss1 mRNA in DIO compared to C rats. Moreover, neither Luque
at the level of the pituitary. Additionally, we have observed changes in et al. (2007) nor our study revealed changes in GPR54 mRNA in DIO an-
Kiss1 and/or GPR54 mRNA levels in the peripheral tissues (fat, pancreas imals. However, we found an increase in GPR54 mRNA in both DM1 and

Please cite this article as: Dudek, M., et al., Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the
hypothalamic–pituitary–gonadal (HPG..., Neuropeptides (2016), http://dx.doi.org/10.1016/j.npep.2016.01.005
6 M. Dudek et al. / Neuropeptides xxx (2016) xxx–xxx

DM2 compared to C rats. On the other hand, experiments with short- in terms of LH discharge to exogenous KP injections is conserved
time (72-h) food deprivation showed a decrease in Kiss1 mRNA in pre- (Castellano et al., 2006; George et al., 2013). Unfortunately, we did not
pubertal male rats (Castellano et al., 2005) and adult female rats (Brown have enough tissue in this study to examine the protein levels in the
et al., 2008) as well as an increase in GPR54 mRNA in the hypothalamus pituitary.
of pubertal rats (Castellano et al., 2005). Importantly, a number of stud- A novel finding is that within the HPG axis, the strongest alterations
ies in diabetic human patients and experimental diabetic animals have in Kiss1 and GPR54 mRNA were found in the rats' testes. Kiss1 and
reported preserved GnRH content and secretory capacity in the hypo- GPR54 mRNA was detected in mouse testes and immunocytochemistry
thalamus, suggesting that the central cause of reproductive impairment confirmed the presence of this peptide in the Leydig cells (Hsu et al.,
lies upstream of GnRH neurons (Howland and Zebrowski, 1976; Sexton 2014; Salehi et al., 2015). We have also reported that in DM1 animals,
and Jarow, 1997; Spindler-Vomachka and Johnson, 1985). Kiss1 mRNA expression was the highest and was accompanied by a de-
We did not find changes in pituitary Kiss1 mRNA and GPR54 mRNA crease in GPR54 mRNA. In contrast, in DM2 animals, a significant in-
in DM1 and DM2 groups. This is in agreement with studies in human di- crease in GPR54 mRNA levels was reported. Thus, the results suggest
abetic patients and diabetic rats, which showed that pituitary response that metabolic status influences the expression of Kiss1 and GPR54

Fig. 4. Effects in male rats of diet-induced obesity (DIO) and STZ-induced diabetes type 1 and type 2 on kisspeptin (A, C, E) and GPR54 (B, D, F) levels in the adipose tissue (fat) (A, B), the
pancreas (C, D) and the liver (E, F). Values were determined by Western blot and were expressed as percentage changes compared to controls fed ad libitum (mean ± S.E.M.). Letters
represent the difference between groups. C — control; DIO — obese rats; DM1 — rats with diabetes type 1; DM2 — rats with diabetes type 2. (A–B): Fat: (A): a: DM1 N C, (p b 0.00005),
nd — non detectable. (B): nd — non detectable; (C–D): Pancreas: (C): a: DIO b C, (p b 0.05); b: DM2 b C, (p b 0.05); nd — non detectable; (D): a: DIO b C (p b 0.05); nd — non
detectable. (E–F) Liver: (E): a: DM2 N C, (p b 0.05); b: DIO N C, (trend p = 0.053); c: DM1 b DIO, (p b 0.05), d: DM1 b DM2 (p b 0.05).

Please cite this article as: Dudek, M., et al., Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the
hypothalamic–pituitary–gonadal (HPG..., Neuropeptides (2016), http://dx.doi.org/10.1016/j.npep.2016.01.005
M. Dudek et al. / Neuropeptides xxx (2016) xxx–xxx 7

Fig. 4 (continued).

mRNA in the testes and has differential effects in DM1 and DM2 male Our study confirmed previous data showing Kiss1 and GPR54 ex-
rats. Our unpublished data also indicated that DM1 and DM2 rats had pression in the pancreas, which may affect the functioning of the pan-
lower levels of T and LH compared to C animals. Thus, it seems of partic- creatic islets (Dahri et al., 1995; Hauge-Evans et al., 2006; Kotani et al.,
ular interest to further explore alterations in testes in this animal model. 2001; Rizza et al., 1980). Moreover, we have shown that proteins for
Moreover, we have added to the literature that Kiss1 mRNA in rats is kisspeptin and GPR54 are also present in this tissue. It was found that
present not only in visceral (epididymal) adipose tissue (Brown et al., kisspeptin-ir and GPR54-ir cells were co-localized in beta and alpha
2008) but also in gonadal fat (current data). Of relevance, studies in cells within the islets (Hauge-Evans et al., 2006). Recently, several stud-
women have shown a significant positive correlation between Kiss1 ies suggested a role for kisspeptin in the regulation of B-cell secretion.
mRNA and BMI in omental adipose tissue. However, no correlation Exposure of mouse and human islets to kisspeptin caused the stimula-
was found in subcutaneous fat (Cockwell et al., 2013). tion of glucose-induced insulin (Hauge-Evans et al., 2006; Vikman and
Results of previous experiments suggested that Kiss1 expression in Ahrén, 2009). However, kisspeptin did not influence the basal rate of se-
fat is independent of the hypothalamus (Brown et al., 2008). Moreover, cretion at a sub-stimulatory concentration of glucose; rather, it regulat-
nutritional status differentially alters Kiss1 mRNA in fat, pituitary and ed insulin secretion at physiological concentrations of glucose in
hypothalamus tissues. While food restriction (18 h) increased Kiss1 isolated mouse islets (Hauge-Evans et al., 2006). Since pancreatic beta
mRNA in fat in both male and female rats, it decreased Kiss1 mRNA in cells have an important role in fuel homeostasis, and kisspeptin and
males' pituitary and females' hypothalamus (Brown et al., 2008). In con- GPR54 are present there, it is speculated that the kisspeptin system
trast, a reduction in the fat expression of Kiss1 mRNA was reported in may be involved in the magnitude of insulin secretion in response to cir-
response to HFD treatment and in obese Zucker rats (Brown et al., culation nutrients (Hauge-Evans et al., 2006). Our current study con-
2008). In our study, we did not find a significant increase in Kiss1 firms this scenario, as Kiss1 and GPR54 expression was altered in the
mRNA in DIO and DM2 animals, but we reported a robust increase in pancreas in DIO, DM1 and DM2 animals. We have observed a downreg-
DM1 rats compared to other groups. Thus, our data confirmed the pre- ulation of kisspeptin in DIO and DM2 animals and its levels were below
vious findings that changes in Kiss1 and GPR54 mRNA in fat were inde- detectible values in DM1, likely due to destruction of the pancreas by
pendent of the hypothalamus. Moreover, we have shown that the most STZ injections. Similarly, we were unable to detect GPR54 in this tissue
severe alterations were present in DM1 rats. However, levels of both in DM1 rats. Thus, we have shown that in the case of increased glucose
Kiss1 and GPR54 mRNA and respective proteins were relatively low in concentrations (DM2 and DM1 animals), the kisspeptin/GPR54 system
fat compared to other examined tissues, and kisspeptin and GPR54 may not function properly and is unable to control insulin secretion.
were undetectable in DIO and DM2 animals using Western blots. We This is in agreement with the proposed tri-hormonal endocrine circuit
have reported a decrease in DM1 animals compared to C animals. between islets and the liver implied in the control of glucose regulation
Thus, in spite of higher Kiss1 mRNA in DM1, protein levels were lower (Hussain et al., 2015). Importantly, our data in DM2 animals confirms
compared to control rats. We did not explore the mechanism responsi- the results obtained from humans with diabetes type 2, where an in-
ble for alterations in protein levels in these animals. However, processes crease in liver kisspeptin levels was observed (Song et al., 2014).
like alternative splicing, folding, assembly into multimers, transport to In general, early in the pathogenesis of DM2, deregulated glucagon
an appropriate subcellular location, or posttranslational protein modifi- secretion from pancreatic alpha cells occurs prior to impaired glucose-
cation can occur, leading to lower levels of kisspeptin in DM1 rats stimulated insulin secretion (GSIS) from beta cells (Song et al., 2014).
(Anderson and Seilhamer, 1997; Wang et al., 2008). Interestingly, we Hyperglucagonemia, which occurs early during development of DM2,
also demonstrated that both DIO and DM2 rats fed a high-fat diet had upregulates Kiss1 production by the liver. Kiss1 in turn functions as a
undetectable levels of kisspeptin and GPR54 in their fat. Further exper- hormone to suppress GSIS. Thus, in DM2, the beta cell is exposed to
iments are needed to explore the mechanisms of kisspeptin/GRP54 ac- two counteracting stimuli elicited by glucagon action on the liver. It
tions in fat, examining different types of adipose tissue in this animal was proposed that glucagon-induced hepatic glucose production
model. (HGP) and hyperglycemia stimulate, whereas kisspeptin production

Please cite this article as: Dudek, M., et al., Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the
hypothalamic–pituitary–gonadal (HPG..., Neuropeptides (2016), http://dx.doi.org/10.1016/j.npep.2016.01.005
8 M. Dudek et al. / Neuropeptides xxx (2016) xxx–xxx

inhibits GSIS. It is suggested that a pathogenic mechanism, in the case of D'Alessio, D., 2011. The role of dysregulated glucagon secretion in type 2 diabetes. Diabe-
tes Obes. Metab. 13 (Suppl. 1), 126–132.
DM2, involves hyperglucagonemia via hepatic kisspeptin to impair in- Dahri, S., Reusens, B., Remacle, C., Hoet, J., 1995. Nutritional influences on pancreatic de-
sulin secretion (Song et al., 2014). These findings could also point to- velopment and potential links with non-insulin-dependent diabetes. Proc. Nutr.
ward the therapeutic potential for kisspeptin antagonism to improve Soc. 54, 345–356.
de Roux, N., Genin, E., Carel, J.C., Matsuda, F., Chaussain, J.L., Milgrom, E., 2003.
beta cell function in diabetes (Song et al., 2014). It was also reported Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived pep-
that mice selectively lacking pancreas GPR54 receptors and fed with tide receptor GPR54. Proc. Natl. Acad. Sci. 10972–10976.
HFD showed improved glucose tolerance due to increased glucose stim- Desroziers, E., Mikkelsen, J., Simonneaux, V., Keller, M., Tillet, Y., Caraty, A., Franceschini, I.,
2010. Mapping of kisspeptin fibres in the brain of the pro-oestrous rat.
ulating insulin secretion (GSIS) (Song et al., 2014). Moreover, our data
J. Neuroendocrinol. 22, 1101–1112.
indicates that impairment in the kisspeptin system exists in DM1 rats, Dhillo, W.S., Chaudhri, O.B., Thompson, E.L., Murphy, K.G., Patterson, M., Ramachandran, R.,
both in the levels of kisspeptin and GPR54; and in the pancreas, levels Nijher, G.K., Amber, V., Kokkinos, A., Donaldson, M., Ghatei, M.A., Bloom, S.R., 2007.
Kisspeptin-54 stimulates gonadotropin release most potently during the preovulatory
of the proteins were undetectable, likely due to damage of the tissue
phase of the menstrual cycle in women. J. Clin. Endocrinol. Metab. 92, 3958–3966.
via STZ. Thus, we proposed that, in the case of DM1 animals, very low Ding, E.L., Song, Y., Malik, V.S., Liu, S., 2006. Sex differences of endogenous sex hormones and
(liver and fat) or completely absent kisspeptin/GPR54 signaling (the risk of type 2 diabetes: a systematic review and meta-analysis. JAMA 295, 1288–1299.
case of the pancreas) may contribute to impaired regulation of Dumonteil, E., Magnan, C., Ritz-Laser, B., Meda, P., Dussoix, P., Gilbert, M., Ktorza, A.,
Philippe, J., 1998. Insulin, but not glucose lowering corrects the hyperglucagonemia
glucose-stimulated insulin secretion. and increased proglucagon messenger ribonucleic acid levels observed in
Importantly, recent findings of impaired glucose tolerance in Kiss1R insulinopenic diabetes 1. Endocrinology 139, 4540–4546.
KO female mice could be related to alterations in kisspeptin signaling, Fink, G., 2000. Neuroendocrine Regulation of Pituitary Function, Neuroendocrinology in
Physiology and Medicine. Springer, pp. 107–133.
not only in the brain but also peripherally. Both in vitro and in vivo stud- Ford, E.S., Liu, S., 2005. Invited commentary: acne in adolescence—protecting the heart
ies showed that kisspeptin treatment augments glucose-induced insulin but damaging the prostate later in life? Am. J. Epidemiol. 161, 1102–1106.
secretion supporting a possible role in beta cell functions, and George, J.T., Veldhuis, J.D., Tena-Sempere, M., Millar, R.P., Anderson, R.A., 2013. Exploring
the pathophysiology of hypogonadism in men with type 2 diabetes: kisspeptin-10
underlining impairments observed in Kiss1R KO (Hauge-Evans et al., stimulates serum testosterone and LH secretion in men with type 2 diabetes and
2006; Pallais et al., 2006). mild biochemical hypogonadism. Clin. Endocrinol. 79, 100–104.
Our study confirms above findings and added to the literature that Glass, A.R., Swerdloff, R.S., Bray, G.A., Dahms, W.T., Atkinson, R.L., 1977. Low serum testos-
terone and sex-hormone-binding-globulin in massively obese men. J. Clin.
positive energy balance has profound effects on the HPG axis and pe- Endocrinol. Metab. 45, 1211–1219.
ripheral organs involved in metabolic functions activing via kisspeptin. Gottsch, M.L., Cunningham, M.J., Smith, J.T., Popa, S.M., Acohido, B.V., Crowley, W.F.,
Seminara, S., Clifton, D.K., Steiner, R.A., 2004. A role for kisspeptins in the regulation
of gonadotropin secretion in the mouse. Endocrinology 145, 4073–4077.
Source of funding Guimiot, F., Chevrier, L., Dreux, S., Chevenne, D., Caraty, A., Delezoide, A.L., de Roux, N.,
2012. Negative fetal FSH/LH regulation in late pregnancy is associated with declined
kisspeptin/KISS1R expression in the tuberal hypothalamus. J. Clin. Endocrinol. Metab.
This study was supported by the OPUS grant NCN 2011/01/B/NZ4/ 97, 2221–2229.
04992 to J.H.S. Gutierrez-Pascual, E., Martínez-Fuentes, A., Pinilla, L., Tena-Sempere, M., Malagon, M.,
Castano, J., 2007. Direct pituitary effects of kisspeptin: activation of gonadotrophs
and somatotrophs and stimulation of luteinising hormone and growth hormone se-
Conflicts of interest cretion. J. Neuroendocrinol. 19, 521–530.
Hauge-Evans, A., Richardson, C., Milne, H., Christie, M., Persaud, S., Jones, P., 2006. A role
for kisspeptin in islet function. Diabetologia 49, 2131–2135.
None. Hotchkiss, J., Knobil, E., 1994. The menstrual cycle and its neuroendocrine control. Physiol.
Reprod. 2, 711–749.
Howland, B., Zebrowski, E., 1976. Some effects of experimentally-induced diabetes on pi-
References tuitary–testicular relationships in rats. Horm. Metab. Res. 8, 465–469.
Hsu, M.-C., Wang, J.-Y., Lee, Y.-J., Jong, D.-S., Tsui, K.-H., Chiu, C.-H., 2014. Kisspeptin mod-
Al Hayek, A.A., Khader, Y.S., Jafal, S., Khawaja, N., Robert, A.A., Ajlouni, K., 2013. Prevalence ulates fertilization capacity of mouse spermatozoa. Reproduction 147, 835–845.
of low testosterone levels in men with type 2 diabetes mellitus: a cross-sectional Hussain, M.A., Song, W.-J., Wolfe, A., 2015. There is kisspeptin—and then there is
study. J. Fam. Commun. Med. 20, 179–186. kisspeptin. Trends Endocrinol. Metab. 26, 564–572.
Anderson, L., Seilhamer, J., 1997. A comparison of selected mRNA and protein abundances Irwig, M.S., Fraley, G.S., Smith, J.T., Acohido, B.V., Popa, S.M., Cunningham, M.J., Gottsch,
in human liver. Electrophoresis 18, 533–537. M.L., Clifton, D.K., Steiner, R.A., 2004. Kisspeptin activation of gonadotropin releasing
Bellingham, M., Fowler, P.A., Amezaga, M.R., Rhind, S.M., Cotinot, C., Mandon-Pepin, B., hormone neurons and regulation of KiSS-1 mRNA in the male rat. Neuroendocrinol-
Sharpe, R.M., Evans, N.P., 2009. Exposure to a complex cocktail of environmental ogy 80, 264–272.
endocrine-disrupting compounds disturbs the kisspeptin/GPR54 system in ovine hy- Kawamori, D., Kulkarni, R.N., 2009. Insulin modulation of glucagon secretion: the role
pothalamus and pituitary gland. Environ. Health Perspect. 117, 1556–1562. of insulin and other factors in the regulation of glucagon secretion. Islets 1,
Bowe, J.E., Foot, V.L., Amiel, S.A., Huang, G.C., Lamb, M.W., Lakey, J., Jones, P.M., Persaud, 276–279.
S.J., 2012. GPR54 peptide agonists stimulate insulin secretion from murine, porcine Kawamori, D., Kurpad, A.J., Hu, J., Liew, C.W., Shih, J.L., Ford, E.L., Herrera, P.L., Polonsky,
and human islets. Islets 4, 20–23. K.S., McGuinness, O.P., Kulkarni, R.N., 2009. Insulin signaling in alpha cells modulates
Brown, R., Imran, S., Ur, E., Wilkinson, M., 2008. KiSS-1 mRNA in adipose tissue is regulat- glucagon secretion in vivo. Cell Metab. 9, 350–361.
ed by sex hormones and food intake. Mol. Cell. Endocrinol. 281, 64–72. Kotani, M., Detheux, M., Vandenbogaerde, A., Communi, D., Vanderwinden, J.-M., Le Poul,
Caraty, A., Franceschini, I., Hoffman, G.E., 2010. Kisspeptin and the preovulatory E., Brézillon, S., Tyldesley, R., Suarez-Huerta, N., Vandeput, F., 2001. The metastasis
gonadotrophin-releasing hormone/luteinising hormone surge in the ewe: basic as- suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G
pects and potential applications in the control of ovulation. J. Neuroendocrinol. 22, protein-coupled receptor GPR54. J. Biol. Chem. 276, 34631–34636.
710–715. Lakshman, K.M., Bhasin, S., Araujo, A.B., 2010. Sex hormone-binding globulin as an inde-
Castellano, J., Navarro, V., Fernandez-Fernandez, R., Nogueiras, R., Tovar, S., Roa, J., pendent predictor of incident type 2 diabetes mellitus in men. J. Gerontol. A Biol. Sci.
Vazquez, M., Vigo, E., Casanueva, F., Aguilar, E., 2005. Changes in hypothalamic Med. Sci. 65, 503–509.
KiSS-1 system and restoration of pubertal activation of the reproductive axis by Lehman, M.N., Hileman, S.M., Goodman, R.L., 2013. Neuroanatomy of the kisspeptin sig-
kisspeptin in undernutrition. Endocrinology 146, 3917–3925. naling system in mammals: comparative and developmental aspects. Adv. Exp.
Castellano, J.M., Navarro, V.M., Fernández-Fernández, R., Roa, J., Vigo, E., Pineda, R., Med. Biol. 784, 27–62.
Dieguez, C., Aguilar, E., Pinilla, L., Tena-Sempere, M., 2006. Expression of hypothalam- Luque, R.M., Kineman, R.D., Tena-Sempere, M., 2007. Regulation of hypothalamic expres-
ic KiSS-1 system and rescue of defective gonadotropic responses by kisspeptin in sion of KiSS-1 and GPR54 genes by metabolic factors: analyses using mouse models
streptozotocin-induced diabetic male rats. Diabetes 55, 2602–2610. and a cell line. Endocrinology 148, 4601–4611.
Castellano, J.M., Navarro, V.M., Roa, J., Pineda, R., Sanchez-Garrido, M.A., Garcia-Galiano, Mendis, S., Davis, S., Norrving, B., 2015. Organizational update: the world health organiza-
D., Vigo, E., Dieguez, C., Aguilar, E., Pinilla, L., Tena-Sempere, M., 2009. Alterations in tion global status report on noncommunicable diseases 2014; one more landmark
hypothalamic KiSS-1 system in experimental diabetes: early changes and functional step in the combat against stroke and vascular disease. Stroke 46, 121–122.
consequences. Endocrinology 150, 784–794. Muller, W.A., Faloona, G.R., Unger, R.H., 1973. Hyperglucagonemia in diabetic
Cockwell, H., Wilkinson, D.A., Bouzayen, R., Imran, S.A., Brown, R., Wilkinson, M., 2013. ketoacidosis. Its prevalence and significance. Am. J. Med. 54, 52–57.
KISS1 expression in human female adipose tissue. Arch. Gynecol. Obstet. 287, Navarro, V.M., Castellano, J.M., Fernandez-Fernandez, R., Barreiro, M.L., Roa, J., Sanchez-
143–147. Criado, J.E., Aguilar, E., Dieguez, C., Pinilla, L., Tena-Sempere, M., 2004. Developmental
Costanzo, P.R., Suarez, S.M., Scaglia, H.E., Zylbersztein, C., Litwak, L.E., Knoblovits, P., 2014. and hormonally regulated messenger ribonucleic acid expression of KiSS-1 and its
Evaluation of the hypothalamic–pituitary–gonadal axis in eugonadal men with type 2 putative receptor, GPR54, in rat hypothalamus and potent luteinizing hormone-
diabetes mellitus. Andrology 2, 117–124. releasing activity of KiSS-1 peptide. Endocrinology 145, 4565–4574.

Please cite this article as: Dudek, M., et al., Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the
hypothalamic–pituitary–gonadal (HPG..., Neuropeptides (2016), http://dx.doi.org/10.1016/j.npep.2016.01.005
M. Dudek et al. / Neuropeptides xxx (2016) xxx–xxx 9

Pallais, J.C., Bo-Abbas, Y., Pitteloud, N., Crowley Jr., W.F., Seminara, S.B., 2006. Neuroendo- Song, W.-J., Mondal, P., Wolfe, A., Alonso, L.C., Stamateris, R., Ong, B.W., Lim, O.C., Yang,
crine, gonadal, placental, and obstetric phenotypes in patients with IHH and muta- K.S., Radovick, S., Novaira, H.J., 2014. Glucagon regulates hepatic kisspeptin to impair
tions in the G-protein coupled receptor, GPR54. Mol. Cell. Endocrinol. 254-255, insulin secretion. Cell Metab. 19, 667–681.
70–77. Spindler-Vomachka, M., Johnson, D., 1985. Altered hypothalamic–pituitary function in the
Pasquali, R., Patton, L., Gambineri, A., 2007. Obesity and infertility. Curr. Opin. Endocrinol. adult female rat with streptozotocin-induced diabetes. Diabetologia 28, 38–44.
Diabetes Obes. 14, 482–487. Stellato, R.K., Feldman, H.A., Hamdy, O., Horton, E.S., McKinlay, J.B., 2000. Testosterone,
Quennell, J., Rizwan, M., Relf, H.L., Anderson, G., 2010. Developmental and steroidogenic sex hormone-binding globulin, and the development of type 2 diabetes in middle-
effects on the gene expression of RFamide related peptides and their receptor in aged men: prospective results from the Massachusetts male aging study. Diabetes
the rat brain and pituitary gland. J. Neuroendocrinol. 22, 309–316. Care 23, 490–494.
Quennell, J.H., Howell, C.S., Roa, J., Augustine, R.A., Grattan, D.R., Anderson, G.M., 2011. Szkudelski, T., 2001. The mechanism of alloxan and streptozotocin action in B cells of the
Leptin deficiency and diet-induced obesity reduce hypothalamic kisspeptin expres- rat pancreas. Physiol. Res. 50, 537–546.
sion in mice. Endocrinology 152, 1541–1550. Tolson, K.P., Garcia, C., Yen, S., Simonds, S., Stefanidis, A., Lawrence, A., Smith, J.T.,
Reed, M.J., Meszaros, K., Entes, L.J., Claypool, M.D., Pinkett, J.G., Gadbois, T.M., Reaven, Kauffman, A.S., 2014. Impaired kisspeptin signaling decreases metabolism and pro-
G.M., 2000. A new rat model of type 2 diabetes: the fat-fed, streptozotocin-treated motes glucose intolerance and obesity. J. Clin. Invest. 124, 3075–3079.
rat. Metabolism 49, 1390–1394. Vater, A., Sell, S., Kaczmarek, P., Maasch, C., Buchner, K., Pruszynska-Oszmalek, E.,
Rizza, R.A., Gerich, J.E., Haymond, M.W., Westland, R.E., Hall, L.D., Clemens, A.H., Service, Kolodziejski, P., Purschke, W.G., Nowak, K.W., Strowski, M.Z., Klussmann, S., 2013. A
F.J., 1980. Control of blood sugar in insulin-dependent diabetes: comparison of an ar- mixed mirror-image DNA/RNA aptamer inhibits glucagon and acutely improves glu-
tificial endocrine pancreas, continuous subcutaneous insulin infusion, and intensified cose tolerance in models of type 1 and type 2 diabetes. J. Biol. Chem. 288, 21136–21147.
conventional insulin therapy. N. Engl. J. Med. 303, 1313–1318. Vikman, J., Ahrén, B., 2009. Inhibitory effect of kisspeptins on insulin secretion from isolat-
Salehi, S., Adeshina, I., Chen, H., Zirkin, B.R., Hussain, M.A., Wondisford, F., Wolfe, A., ed mouse islets. Diabetes Obes. Metab. 11, 197–201.
Radovick, S., 2015. Developmental and endocrine regulation of kisspeptin expression Wahab, F., Shahab, M., Behr, R., 2015. The involvement of gonadotropin inhibitory hor-
in mouse leydig cells. Endocrinology 156, 1514–1522. mone and kisspeptin in the metabolic regulation of reproduction. J. Endocrinol.
Schwartz, N.B., 2000. Neuroendocrine Regulation of Reproductive Cyclicity, Neuroendo- 225, 49–66.
crinology in Physiology and Medicine. Springer, pp. 135–145. Wang, E.T., Sandberg, R., Luo, S., Khrebtukova, I., Zhang, L., Mayr, C., Kingsmore, S.F.,
Seminara, S.B., Messager, S., Chatzidaki, E.E., Thresher, R.R., Acierno Jr., J.S., Shagoury, J.K., Schroth, G.P., Burge, C.B., 2008. Alternative isoform regulation in human tissue
Bo-Abbas, Y., Kuohung, W., Schwinof, K.M., Hendrick, A.G., Zahn, D., Dixon, J., Kaiser, transcriptomes. Nature 456, 470–476.
U.B., Slaugenhaupt, S.A., Gusella, J.F., O'Rahilly, S., Carlton, M.B., Crowley Jr., W.F., Wu, F.C., Tajar, A., Pye, S.R., Silman, A.J., Finn, J.D., O'Neill, T.W., Bartfai, G., Casanueva, F., Forti,
Aparicio, S.A., Colledge, W.H., 2003. The GPR54 gene as a regulator of puberty. N. G., Giwercman, A., Huhtaniemi, I.T., Kula, K., Punab, M., Boonen, S., Vanderschueren, D.,
Engl. J. Med. 349, 1614–1627. European Male Aging Study, G., 2008. Hypothalamic–pituitary–testicular axis disrup-
Sexton, W.J., Jarow, J.P., 1997. Effect of diabetes mellitus upon male reproductive function. tions in older men are differentially linked to age and modifiable risk factors: the
Urology 49, 508–513. European Male Aging Study. J. Clin. Endocrinol. Metab. 93, 2737–2745.
Shahab, M., Mastronardi, C., Seminara, S.B., Crowley, W.F., Ojeda, S.R., Plant, T.M., 2005. In- Zhang, M., Lv, X.Y., Li, J., Xu, Z.G., Chen, L., 2008. The characterization of high-fat diet and
creased hypothalamic GPR54 signaling: a potential mechanism for initiation of pu- multiple low-dose streptozotocin induced type 2 diabetes rat model. Exp. Diabetes
berty in primates. Proc. Natl. Acad. Sci. 102, 2129–2134. Res. 2008, 704045.
Skorupskaite, K., George, J.T., Anderson, R.A., 2014. The kisspeptin-GnRH pathway in Zhou, Q., Chen, H., Yang, S., Li, Y., Wang, B., Chen, Y., Wu, X., 2014. High-fat diet decreases
human reproductive health and disease. Hum. Reprod. 20, 485–500. the expression of Kiss1 mRNA and kisspeptin in the ovary, and increases ovulatory
Smith, J.T., Acohido, B.V., Clifton, D.K., Steiner, R.A., 2006. KiSS-1 neurones are direct tar- dysfunction in postpubertal female rats. Reprod. Biol. Endocrinol. 12, 127 http://
gets for leptin in the ob/ob mouse. J. Neuroendocrinol. 18, 298–303. www.who.int/mediacentre/factsheets/fs311/en/.

Please cite this article as: Dudek, M., et al., Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the
hypothalamic–pituitary–gonadal (HPG..., Neuropeptides (2016), http://dx.doi.org/10.1016/j.npep.2016.01.005

You might also like