You are on page 1of 14

Am J Physiol Endocrinol Metab 322: E494–E507, 2022.

First published April 11, 2022; doi:10.1152/ajpendo.00432.2021

RESEARCH ARTICLE

Deciphering the Contribution of the Gastrointestinal Tract on Glucose, Lipid, and Energy
Metabolism

Ghrelin deficiency sex-dependently affects food intake, locomotor activity, and


adipose and hepatic gene expression in a binge-eating mouse model
Karina Prins, Martin Huisman, Anke McLuskey, Rosinda Mies, Bas Karels,† Patric J. D. Delhanty, and
Jenny A. Visser
Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands

Abstract
Binge-eating disorder is the most prevalent eating disorder diagnosed, affecting three times more women than men. Ghrelin
stimulates appetite and reward signaling, and loss of its receptor reduces binge-eating behavior in male mice. Here, we exam-
ined the influence of ghrelin itself on binge-eating behavior in both male and female mice. Five-wk-old wild-type (WT) and ghre-
lin-deficient (Ghrl / ) mice were housed individually in indirect calorimetry cages for 9 wks. Binge-like eating was induced by
giving mice ad libitum chow, but time-restricted access to a Western-style diet (WD; 2 h access, 3 days/wk) in the light phase
(BE); control groups received ad libitum chow (CO), or ad libitum access to both diets (CW). All groups of BE mice showed
binge-eating behavior, eating up to 60% of their 24-h intake during the WD access period. Subsequent dark phase chow intake
was decreased in Ghrl / mice and remained decreased in Ghrl / females on nonbinge days. Also, nonbinge day locomotor
activity was lower in Ghrl / than in WT BE females. Upon euthanasia, Ghrl / BE mice weighed less and had a lower lean body
mass percentage than WT BE mice. In BE and CW groups, ghrelin and sex altered the expression of genes involved in lipid
processing, thermogenesis, and aging in white adipose tissue and livers. We conclude that, although ghrelin deficiency does not
hamper the development of binge-like eating, it sex-dependently alters food intake timing, locomotor activity, and metabolism.
These results add to the growing body of evidence that ghrelin signaling is sexually dimorphic.
NEW & NOTEWORTHY Ghrelin, a peptide hormone secreted from the gut, is involved in hunger and reward signaling, which
are altered in binge-eating disorder. Although sex differences have been described in both binge-eating and ghrelin signaling,
this interaction has not been fully elucidated. Here, we show that ghrelin deficiency affects the behavior and metabolism of mice
in a binge-like eating paradigm, and that the sex of the mice impacts the magnitude and direction of these effects.

binge eating; food intake; ghrelin; locomotor activity; sex differences

INTRODUCTION times more likely to be diagnosed with BED in their life-


time (1, 6).
Binge-eating disorder (BED) is currently the most diag- These sex differences in BED have also been researched
nosed eating disorder (1, 2). It is characterized by binge- using laboratory animals. Several models of binge-like eating
eating episodes: excessive food intake in a short time have been developed for rodents (7–9). The model of limited
span, in which patients experience a loss of control over access to a highly palatable, energy-rich food without food
their eating (3). During these episodes, patients often deprivation, developed for rats and adapted for mice, has
consume palatable, high-sugar/high-fat foods. Besides been well characterized and used in many studies. In this
the emotional distress this causes, patients do not com- model, animals are provided sporadic, time-limited access to
pensate for the surplus calories they eat during episodes. a high-fat diet or to a Western-style diet (WD) and eat more
This often leads to weight gain and resulting metabolic calories on access days than on nonaccess days. Although
comorbidities, such as obesity and metabolic syndrome this model does not cause overt obesity in the short term,
(4, 5). Like in other eating disorders, the prevalence of metabolic effects have been observed, such as impaired glu-
BED is greater in women than in men: women are three cose homeostasis and increased fat mass (9). Importantly,

P. J. D. Delhanty and J. A. Visser contributed equally to this work.


†Deceased 15 December 2021.
Correspondence: J. A. Visser (j.visser@erasmusmc.nl).
Submitted 14 December 2021 / Revised 25 March 2022 / Accepted 4 April 2022

E494 0193-1849/22 Copyright © 2022 the American Physiological Society. http://www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

this model emulates aspects of the disturbed eating behav- found that male rats are more sensitive to ghrelin treatment,
iors observed in humans with BED, particularly those related assessed by acute food intake, than female rats (37). However
to hedonic rather than homeostatic control of eating. In this in mice, when ghrelin is administered just before the dark pe-
model, female rats escalated their intake of palatable food riod, females are not only more sensitive in terms of its orexi-
faster than males (10) and were more likely to be classified as genic effects, but also the effects of treatment lasted longer
binge-eating prone or high responders (11, 12). The increased than in males (38). Likewise, the GH secretagogue activity of
sensitivity of females to food-related reward signaling has ghrelin appears to be more potent in females than in males
been implicated to play a role in this process (13). Similarly, (38), and it has been shown that there is estrogen dependency
in humans, it appears that altered food cues and reward on ghrelin levels and the orexigenic efficacy of ghrelin. For
processing, and sex differences herein, play a role in the eti- example, the orexigenic effects of ghrelin in male and ovariec-
ology of BED (14, 15). tomized female rats are reduced after estrogen treatment.
Ghrelin is a peptide hormone that is secreted by the Furthermore, the inductive effects of ovariectomy on food
stomach. It is present in the blood in two forms: acylated intake and body weight are lost in GHSR-null mice (37). This
ghrelin (AG) and unacylated ghrelin (UAG). Acylated ghre- could be related to the direct modulation of neuronal sensitiv-
lin is octanoylated by ghrelin-O-acyltransferase (GOAT), ity to ghrelin by estrogens (39, 40). However, these studies
and is the endogenous ligand of the growth hormone se- only give an idea of the acute physiological response to the
cretagogue receptor 1a (GHSR1a) (16). Through this recep- administration of pharmacological doses of ghrelin.
tor, AG stimulates growth hormone secretion, enhances Given the sex differences in BED prevalence and in ghre-
appetite, and plays a role in stress and reward signaling lin signaling, it is surprising that most of the ghrelin-related
(17, 18). UAG, however, appears to be antidiabetogenic and studies on BED have only been performed in male mice.
its levels correlate with fasting insulin, BMI, waist circum- This study aims to find out whether sex impacts the effects
ference, and weight loss, although its mechanism of action of ghrelin on the development and metabolic consequences
is not known (19, 20). Altogether, these functions make of binge-eating behavior. We hypothesized that ghrelin defi-
ghrelin an interesting potential modulator of binge-like ciency would attenuate the binge-like pattern of eating that
eating behavior and resulting metabolic comorbidities. occurs in WT mice, using an intermittent access model, like
Data on both postprandial and fasted plasma ghrelin lev- that of King et al. (34) in their study in Ghsr / mice. We also
els in patients with BED are conflicting, however. Although hypothesized that the sex of the mice would modulate this
most studies show that plasma ghrelin concentrations are binge-like eating behavior, perhaps in interaction with ghre-
lower in patients with BED than in weight-matched controls lin deficiency. Therefore, to assess these hypotheses, we
(21–24), others find no significant differences between these exposed male and female mice, with or without ghrelin defi-
groups (25). A recent meta-analysis of genome-wide associa- ciency, to a limited access model (2 h access to WD/day 3
tion studies in BED identified ghrelin as a potential gene of times a week, relative to mice given either free access to both
interest: Leu72Met ghrelin is more commonly expressed in WD and regular chow or chow-only controls). We ran this
patients with BED than in healthy controls (9, 26). Moreover, model for 9 wk, a relatively long time, with the aim of accen-
fasting plasma AG levels were observed to correlate posi- tuating any metabolic effects. In addition to food intake,
tively with the risk of developing eating disorders in women using a metabolic cage system, we have also assessed the sex
with obesity (27). Interestingly, total plasma ghrelin levels and genotype effects of the binge-like eating protocol on
positively correlate with both estrogens in women and tes- energy expenditure, activity, and substrate use. Finally, the
tosterone in men (28–30). In contrast, high androgen levels expression of genes relevant to glucose homeostasis and
in women decrease circulating total ghrelin (31), and anti- lipid metabolism in livers and inguinal white adipose tissue
androgen treatment in women increases plasma total ghrelin (WAT), and plasma metabolite levels have been determined.
(32). Altogether, these studies imply that ghrelin levels are
altered in BED and that ghrelin levels are sex dependent.
In laboratory animals, components of the ghrelin signal-
MATERIALS AND METHODS
ing pathway have proven an interesting target in BED Animals and Housing
research. Results in Ghsr knockout (Ghsr / ) mice, however,
strongly depend on the frequency of access to the palatable WT and Ghrl / male and female mice with a C57BL/6
diet that is used to evoke binge-eating behavior. That is, background (41, 42) from the same breeding colony were
when mice are given daily time-limited access to a palatable bred in-house. The mice (48 males, 48 females) were 5 to 6
diet, Ghsr / mice develop binge-like eating behavior simi- wk old at the start of the experiment and selected from litters
larly to wild-type (WT) mice (33, 34). In contrast, when the of 7–10 pups to reduce variability in body weight. The age of
access to the palatable food is only given once every 2 days, the mice corresponds to adolescence, which is the most com-
Ghsr / mice eat less of the palatable diet than WT mice mon age of onset of eating disorders in humans (43).
(34). In a shorter experiment, Valdivia et al. (35) showed that Housing conditions provided a 12-h light:dark cycle with
Ghsr / mice did not escalate the intake of a high-fat diet lights on at 7:00 AM [zeitgeber time (ZT) 0]. Ambient temper-
over 4 days of restricted access to it, whereas WT mice did. ature was maintained at 23 C. Throughout the experiment,
Despite these findings, Ghrl / and Goat / mice have not all mice had ad libitum access to water. All experimental pro-
been investigated using these models. cedures were approved as part of license AVD1010020174317
Furthermore, there are very few basic studies so far that (Central Animal Testing Committee, The Netherlands).
have examined sex-dependent differences in the physiologi- Mice were individually housed in a Promethion metabolic
cal response to ghrelin (36). Intriguingly, one of these studies cage system (Sable Systems, Las Vegas, NV) for 9 wk,

AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org E495


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

including a 5-day habituation period. The cages (Greenline, Hormone Assessment


Tecniplast, Italy) with woodchip bedding (Lignocel BK 8/15,
J. Rettenmaier & So € hne GmbH, Germany) were fitted with a The blood aliquot for measuring AG and UAG was stabi-
water bottle, a nest, and two food containers. Horizontal and lized with 4-(2-aminoethyl)benzenesulfonyl fluoride hydro-
vertical location, food intake, and body mass were detected chloride (AEBSF, Roche, Germany; final concentration 0.2
at 1 Hz by an infrared beam grid and load cells from which mM) before centrifugation. Plasma AG and UAG were meas-
the food containers and nests were suspended, respectively. ured by mouse/rat-specific ELISA (RRID:AB_2619624, Bertin
The cage air influx was set to 2 L/min, and respiratory gases Pharma, France; RRID:AB_2619623, Bertin Pharma, France)
were collected and analyzed at 5-min intervals. Volumes of (45). Plasma levels of leptin were determined by mouse/rat
consumed O2 (V_ O2), produced CO2 (V_ CO2), and water vapor leptin ELISA (RRID:AB_2801467, Alpco Diagnostics, NH),
(V_ H2O) were measured. The cage system and data acquisi- and total adiponectin concentrations were measured with a
tion were controlled with MetaScreen (v. 2.3.15.11) software. mouse/rat HMW and total adiponectin ELISA kit (RRID:
AB_2801466, Alpco Diagnostics, NH) (46). Corticosterone
Diets was extracted from feces from the cage bedding of weeks 5–7
of the experiment as previously described (47), and meas-
Throughout the experiment, mice in the chow-only
ured by ELISA (RRID:AB_2307314, Enzo Life Sciences,
(CO) group had ad libitum access to pelleted rodent chow,
Switzerland).
containing 3.59 kcal/g [22% from protein, 69% from car-
bohydrate (of which 10% sucrose), 9% from fat; 801722
CRM-P, Special Diets Services, UK]. In the binge-eating
Metabolite Analysis
(BE) group, binge-like eating was induced by limiting the Plasma triglycerides (A11A01640, Horiba Group, France),
access of the mice to a palatable WD [4.69 kcal/g, 17% non-esterified fatty acids (NEFA) (3055, Instruchemie, The
from protein, 43% from carbohydrate (of which 70% su- Netherlands), and cholesterol concentrations (A11A01634,
crose), 41% from fat; D12079B, Research Diets, New Horiba Group, France) were measured colorimetrically,
Brunswick, NJ] (7). After a 1-wk habituation period, in according to the manufacturer’s protocol. Liver glycerol
which mice had free access to both chow and WD, BE was determined by colorimetric measurement, according
mice could only access WD for 2 h during the light phase to the manufacturer’s protocol (2913, Instruchemie, The
(ZT4-6) on Monday, Wednesday, and Friday. WD access Netherlands). Hepatic glycogen content was determined
was automatically regulated using software-controlled as previously described by subtracting free glucose from
shutters. To control for the effects of WD consumption, a glycogen, measured using a colorimetric assay (2319,
final group of mice (CW) had free access to both chow and Instruchemie, The Netherlands) (48, 49).
WD throughout the experiment.
Gene Expression Analysis
Metabolic Assessment
RNA was isolated from snap-frozen livers and inguinal
In week 7, glucose tolerance was determined by an intra- WAT (TriPure Isolation Reagent, Roche Diagnostics), and
peritoneal glucose tolerance test (IPGTT), as previously genomic DNA contamination was removed (RQ1 RNase-
described (44). Briefly, mice were transferred to new cages Free DNase, Promega Corporation, WI). cDNA was synthe-
where they were fasted for 5 h (ZT2-7). Next, the mice were sized (Transcriptor high-fidelity cDNA synthesis kit,
weighed, and fasting blood glucose was measured in tail cut Roche Diagnostics), and quantitative PCR was performed
blood. A 2 g/kg glucose bolus was injected intraperitoneally on a QuantSTudio 7 flex real-time PCR system (Applied
and blood glucose levels were determined with a glucometer Biosystems, Life Technologies, CA), using FastStart Universal
at 15, 30, 60, and 120 min after injection. SYBR Green Master (Rox) (Roche Diagnostics). Target gene
expression was normalized to the mean of two housekeeping
Euthanasia and Tissue Collection genes (b-actin and 18S for liver, b2-microglobulin and 18S for
Mice were euthanized in week 9 between ZT2-5 on inguinal WAT) by the 2 DDCT method, as described previously
Monday, Wednesday, or Friday (access time/days for BE (46). Amplicon specificity was assessed by melting curve anal-
mice). Mice were weighed manually, and nonfasting ysis. Primer sequences are listed in Supplemental Table S1
blood glucose concentrations were measured. In addi- (https://doi.org/10.6084/m9.figshare.17197274).
tion, body composition was determined by magnetic res-
Data Processing
onance imaging (EchoMRI-100V, EchoMRI LLC, TX). Fat
and lean mass are presented as percentages of total body Data from Monday to Saturday in week 8, the last full week
weight. Next, mice were deeply anesthetized with isoflur- of the experiment, were processed with MacroInterpreter
ane (Pharmachemie BV, The Netherlands) and eutha- (v2.40, Sable Systems, TX) using One-Click Macro 1 (v2.35.0) to
nized by exsanguination. Cardiac blood was collected obtain body mass, energy expenditure, respiratory exchange
using EDTA-coated syringes (Na3EDTA, Sigma Aldrich, ratio (RER), and locomotor activity variables. Energy expendi-
The Netherlands), aliquoted immediately, and centri- ture was calculated using the Weir equation (50), and the RER
fuged for 5 min at 1,300 g to obtain plasma. The resulting was calculated as V_ CO2/ V_ O2.
plasma samples were snap-frozen in liquid nitrogen and Food intake data were analyzed in R (v4.0.2, R Core
stored at 80 C until further use. Liver and brown (BAT) Team). Briefly, food container mass was imported using the
and white adipose tissues (WAT) were dissected, weighed, SableBase package (51) and median filtered to reduce sensor
and snap-frozen in liquid nitrogen for later RNA isolation. noise. The start and end of feeding events were defined as

E496 AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

differential mass values outside the means ± 2SD range. In the CW group, the 2-h binned food intake data showed
Events were defined as lasting more than 10 s and less than a significantly higher food intake in females than in males
15 min, with two events being separated by at least 2 min. (PS = 0.002; Fig. 1D). This sex effect originated from the WD
The position of the mouse in the cage was determined using intake (PS < 0.001; Supplemental Fig. 1D), which made up
the XY infrared beam breaks, and an event was recorded over 90% of the total food intake. Notably, as in the BE
if the mouse was within reach of the food containers for at group, Ghrl / mice in the CW group showed an even more
least 10% of the event duration. Intake was determined by decreased preference for chow, consuming 3.5 ± 1.1% of their
the difference in container mass before and after an event, calories from chow, compared with 8.8 ± 1.6% in WT mice
and events with an intake over 1.2 g were considered unreal- (PGT = 0.029).
istic and excluded.
The resulting data were binned in 2 h blocks to inspect Energy Expenditure
daily profiles, in which the light phase and subsequent dark During the transition from light to dark phase, EE profiles
phase were considered for one experimental day. Cumulative in the CO group showed a genotype-sex interaction effect
data were binned for the light phase, dark phase, and full 24-h (P < 0.05; Fig. 1E), which affected total light phase energy ex-
period. To reduce between-day variation, data for each mouse penditure (PGT:S = 0.019; Supplemental Fig. S1E). However,
were averaged over all days (CO and CW mice), or for access this effect did not last into the dark phase, thus not signifi-
and nonaccess days separately (BE mice). cantly changing the 24-h EE.
In BE mice, EE was increased during the access period on
Statistical Analysis
access days (PZT < 0.001, Fig. 1F). During the light phase,
Statistical analyses were performed in R using the RStatix Ghrl / mice had a higher EE than WT mice (PGT = 0.003,
package (52). Data were analyzed by two-way repeated-meas- Supplemental Fig. S1F). In contrast, on nonaccess days, light
ures ANOVA (2-h binned data) or by two-way ANOVA and phase EE was lower in Ghrl / mice than in WT mice (PGT =
considered significant at P < 0.05 (abbreviations for P values: 0.001, Fig. 1G and Supplemental Fig. S1G).
PGT for genotype effect, PS for sex effect, PT for time effect). Regardless of access, female BE mice expended less
When a significant interaction between factors (genotype and energy than male BE mice (PS < 0.05). Similarly, EE was
sex) was detected, the Tukey HSD post hoc test was used to increased with ghrelin deficiency and female sex in the CW
compare groups. If no significant interaction effect was group (PGT = 0.027, PS = 0.002, Fig. 1H). Ghrl / females, in
observed, t tests with Bonferroni multiple comparison correc- particular, had a higher EE, resulting in a significantly
tions were applied. For the IPGTT data, the area under the increased 24-h EE compared with WT females (P = 0.027)
curve (AUC) was normalized to the fasting glucose level (53). and Ghrl / males (P = 0.014).
Data are visualized in GraphPad Prism (v. 8, GraphPad
Software, La Jolla, CA). All data are represented as means ± SE. Respiratory Exchange Ratio
In the CO group, RER was low during the light phase
RESULTS and increased upon the transition to the dark phase, corre-
sponding with fat oxidation during the inactive (light)
Food Intake phase, and increased carbohydrate use during the active
In the CO group, 2-h binned food intake patterns largely (dark) phase (Fig. 1I). Ghrelin deficiency altered RER in a
overlapped between the groups (Fig. 1A). Although the geno- sex-dependent way in this group (PGT:S < 0.001). That is,
type effect reached significance (PGT = 0.02), the magnitude ghrelin deficiency increased RER in males, whereas the op-
of the difference between WT and Ghrl / was small. posite occurred in females. In males, this effect occurred
Likewise, over the full 24-h period, we only observed a trend mainly during the dark phase, whereas the RER of Ghrl /
toward an interaction effect of the ghrelin genotype and sex females also remained lower during the light phase, albeit
(PGT:S = 0.059; Supplemental Fig. S1A, https://doi.org/10. not significantly.
6084/m9.figshare.19384001). On access days, the RER of BE mice increased upon WD
On access days, BE mice consumed a large proportion of access and remained elevated throughout the remainder of
their total caloric intake from WD during the 2-h access pe- the light phase and subsequent dark phase (Fig. 1J). In the
riod (Fig. 1B). Interestingly, in the subsequent dark phase, early dark phase, Ghrl / mice had a lower RER (PGT:ZT =
Ghrl / mice, and especially Ghrl / males, ate less 0.024), corresponding to the lower food intake around that
chow than WT mice (PGT = 0.005, Supplemental Fig. S1B). time. In the late dark phase, females had a lower RER than
Consequently, on access days, Ghrl / mice consumed a sig- males (PS:ZT = 0.011). On nonaccess days, RER is lower in
nificantly lower percentage of their total food intake from Ghrl / mice (PGT = 0.027, Fig. 1K). Moreover, females have a
chow (47.8 ± 3.7%) than WT mice (60.3 ± 3.8%, PGT = 0.020). lower RER than males (PS = 0.049).
On nonaccess days, Ghrl / mice continued to have a lower The RER of CW mice also showed a circadian pattern, but
chow intake than WT mice (PGT = 0.009; Fig. 1C). Moreover, its amplitude was lower than that of the other groups (PZT <
particularly during the dark phase, females had a lower 0.001, Fig. 1L). This indicates reduced variation in the meta-
chow intake than males (PS:ZT = 0.019). The resulting total bolic fuel that these animals use (predominantly fat).
food intake on the nonaccess day trended toward a geno-
Locomotor Activity
type-sex effect (PGT:S = 0.065; Supplemental Fig. S1C), which
appeared to be mainly driven by the Ghrl / females (P = In CO mice, females moved a greater distance than
0.016 vs. Ghrl / male; P = 0.020 vs. WT female). males, especially during the dark phase (PS:ZT < 0.001,

AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org E497


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

A B C D

E F G H

I J K L

Figure 1. Food intake, energy expenditure, and respiratory exchange ratio. Food intake for CO mice (A), BE mice on access days (B) and nonaccess
days (C), and CW mice (D). Dotted lines represent chow intake, and solid lines represent WD intake. Energy expenditure in kcal of CO mice (E), BE mice
on access days (F) and nonaccess days (G), and CW mice (H). Respiratory exchange ratio of CO mice (I), BE mice on access days (J) and nonaccess days
(K), and CW mice (L). Experimental groups: WT M, wild-type male; Ghrl / M, ghrelin-deficient male; WT F, wild-type female; Ghrl / F, ghrelin-deficient
female. Group size n = 6–8 mice. The access period is marked in gray. Data are presented as means ± SE, and 2-h binned total food intake, energy ex-
penditure, and respiratory exchange ratio were analyzed by two-way repeated-measures ANOVA. Main effects were considered significant if P < 0.05.
g
P < 0.05, (g)P < 0.10 for genotype differences; sP < 0.05, (s)P < 0.10 for sex differences, and P < 0.05, ()P < 0.10 for interaction effects, determined
by post hoc analysis. BE, binge eating; CO, chow only; CW, chow and WD; WD, Western-style diet.

Fig. 2A). Moreover, in both sexes, Ghrl / mice had lower Body Weight and Composition
locomotor activity than WT mice (PGT = 0.029, Supple-
mental Fig. 2F). Although this effect was larger in From week 2 of the experiment, CO mice gained weight
females, the interaction did not reach statistical signifi- significantly, increasing to 23.1 ± 1.0% over baseline at the 8-
cance (PGT:S = 0.080). wk timepoint (PWK < 0.001, Fig. 3A). The resulting body
During the access period, BE mice increased their loco- mass upon euthanasia was lower in females than in males
motor activity, although there were no significant differ- (PS < 0.001, Fig. 3B). Interestingly, ghrelin deficiency
ences between the sexes or genotypes (Fig. 2B). Like CO sex-dependently altered the body composition of the mice
mice, BE females walked more than males, especially dur- (PGT:S < 0.005, Fig. 3C): Ghrl / females had a higher fat per-
ing the dark phase (PS:ZT = 0.045). Interestingly, this sex centage (P = 0.031) and lower lean mass percentage (P =
effect was ablated in Ghrl / females on nonaccess days 0.034) than WT females. Notably, the effect of ghrelin defi-
(PGT:S:ZT = 0.027): the activity of Ghrl / females was simi- ciency was the opposite in males: Ghrl / males trended to-
lar to that of males during the dark phase. ward a higher lean body mass percentage than WT males
In contrast to the other groups, ghrelin deficiency has no (P = 0.067). Differences in body-weight-corrected WAT mass
effect on locomotor activity in CW mice (Fig. 2D). However, corresponded with the body fat percentage (PGT:S < 0.05),
like in CO mice, females walk more than males, especially whereas the body-weight-corrected BAT mass was similar
during the dark phase (PS:ZT < 0.001). across all groups (Fig. 4A).

E498 AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

A B C D

Figure 2. Directed locomotor activity. Distance walked by CO mice (A), BE mice on access days (B) and non-access days (C), and CW mice (D).
Experimental groups: WT M, wild-type male; Ghrl / M, ghrelin-deficient male; WT F, wild-type female; Ghrl / F, ghrelin-deficient female. Group size n =
6–8 mice. The access period is marked in gray. Data are presented as means ± SE and were analyzed by two-way repeated-measures ANOVA. Main
effects were considered significant if P < 0.05. sP < 0.05, (s)P < 0.10 for sex differences, and P < 0.05, ()P < 0.10 for interaction effects, determined
by post hoc analysis. BE, binge eating; CO, chow only; CW, chow and WD; WD, Western-style diet.

Like the CO mice, body weights of BE mice increased sig- both genotypes, being significantly lower than that of WT
nificantly from week 2 of the experiment, up to 16 ± 1.03% in males (P = 0.035).
week 8 (PWK < 0.001, Fig. 3D). Upon euthanasia, females in
this group also weighed less than males (PS < 0.001, Fig. 3E). Metabolic Gene Expression in Livers and Inguinal WAT
Body weights were also lower in Ghrl / mice versus WT As the inguinal WAT mass was affected by genotype and/
mice in both sexes (PGT = 0.001). Although body fat percent- or sex in all diets, we decided to analyze gene expression pro-
age was not significantly affected, lean body mass percent- files in this depot. In CO mice, both sex and genotype
age was lower in Ghrl / mice and in females (PGT = 0.014, affected gene expression in the inguinal WAT depot (Fig. 6A;
PS = 0.002, Fig. 3F). Consistent with the fat percentages, the Supplemental Table S2). Mainly genes involved in lipid stor-
relative weights of most adipose tissues in BE mice were not age and mobilization were expressed less in females (Plin1,
affected by sex or genotype (Fig. 4B). Only inguinal WAT rel- Hsl). However, macrophage markers (Cd206, Cd11c) and
ative weight was increased in Ghrl / mice compared with browning-related genes (Fgf21, Pgc1a) were expressed differ-
WT mice (PGT = 0.022). entially in Ghrl / and WT animals. In livers, gene expres-
Remarkably, female CW mice gained 15% more weight sion was mainly affected by sex (Fig. 6B; Supplemental Table
over baseline than male CW mice (PS:WK < 0.001, Fig. 3G). S3): females showed higher expression of genes involved in
Although body weight upon euthanasia remained lower in fatty acid uptake and metabolism (Cd36, Ppara, Fasn) and of
females than in males (PS = 0.047, Fig. 3H), females of both secreted proteins (Lcn2, Leap2).
genotypes had a higher fat percentage (PS < 0.001), at the Likewise, in BE mice, the expression of genes involved in
expense of a lower lean body mass percentage (PS < 0.001, lipid storage and mobilization in inguinal WAT was lower in
Fig. 3I). Accordingly, relative subcutaneous WAT and ingui- females (Plin1, Atgl, Hsl). Expression of uncoupling proteins,
nal WAT weights were higher in females (PS < 0.05, Fig. 4C). however, was affected by genotype: Ghrl / females
In addition, inguinal WAT was relatively heavier in Ghrl / expressed significantly more Ucp1 than any other group,
animals (PGT = 0.036). Interestingly, ghrelin deficiency pre- whereas Ghrl / mice of both sexes expressed less Ucp2. In
vented the increase in relative retroperitoneal WAT weight addition, there was a suppressive effect of ghrelin deficiency
in CW females (PGT:S = 0.018). Moreover, relative BAT weight on the expression of the proinflammatory M1 macrophage
was increased in CW Ghrl / males compared with WT marker Cd11c (Fig. 6A; Supplemental Table S2). In contrast to
males (PGT:S = 0.035). CO mice, hepatic gene expression in the BE group was
mainly affected by genotype. Ghrl / mice showed a lower
Glucose Tolerance
expression of genes involved in triglyceride and fatty acid
In the IPGTT, CO females cleared the glucose bolus faster metabolism (Dgat1, Gpat1, Ppara, Elovl3, Fasn, Scd1,
than CO males (PS:T = .015, Fig. 5A). Ghrl / animals were Srebp1c), and in the storage and transport of glucose (Gck1,
more glucose tolerant than WT mice (PGT:T = .025), especially Gys2, Slc2a2) (Fig. 6B; Supplemental Table S3).
in males (P = 0.037, Fig. 5B). Finally, in CW mice, inguinal WAT gene expression only
In contrast, all BE groups cleared glucose at a similar rate, showed minor effects of sex (Plin1, Ucp1), whereas genotype
not showing any effects of genotype or sex (Fig. 5, C and D). affected the expression of macrophage polarization and
Conversely, the genotype and sex-dependent effects uncoupling proteins. As in the BE dietary group, but more
seen in the CO group were exaggerated in the CW diet pronounced, inguinal WAT of Ghrl / mice of both sexes
(PGT:T = 0.007, PS:T = 0.002, Fig. 5, E and F). Although WT had lower expression of Cd11c, as well as a trend to lower
females cleared glucose faster than WT males (P = 0.014), expression of Cd68, a general macrophage marker. Ucp1
this sex effect was lost in Ghrl / animals. Instead, the expression was increased, but Ucp2 expression was decreased
AUC of Ghrl / males was similar to that of the females of in iWAT of Ghrl / mice compared with their WT

AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org E499


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

A B C

Figure 3. Body weight and composition.


Weight gain over baseline for CO mice (A),
BE mice (D), and CW mice (G). Body
weight upon euthanasia for CO mice (B),
BE mice (E), and CW mice (H). Percentage
fat and lean body mass of CO mice (C), BE
mice (F), and CW mice (I). Experimental
D E F
groups: WT M, wild-type male; Ghrl / M,
ghrelin-deficient male; WT F, wild-type
female; Ghrl / F, ghrelin-deficient female.
Group size n = 6–8 mice. Data are pre-
sented as means ± SE and were analyzed
by two-way repeated-measures ANOVA
for weight gain over baseline and two-
way ANOVA for the other panels. Main
effects were considered significant if P <
0.05. (g)P < 0.10 for genotype differences;
s
P < 0.05, (s)P < 0.10 for sex differences,
and ()P < 0.10 for interaction effects,
determined by post hoc analysis. BE,
binge eating; CO, chow only; CW, chow
and WD; WD, Western-style diet. G H I

counterparts (Fig. 6A; Supplemental Table S2). In the livers of tended to have lower levels than WT males, this sex differ-
CW mice, gene expression effects were more pronounced ence was reversed in Ghrl / animals. In addition, plasma
compared with those of BE mice, being mainly affected by ge- cholesterol levels were lower in females than in males (PS <
notype and not sex (Fig. 6B; Supplemental Table S3). 0.001). Liver glycerol content was higher in females (PS =
0.003), and liver glycogen content trended toward being
Circulating and Hepatic Metabolites lower in Ghrl / mice (PGT = 0.060).
Upon euthanasia, Ghrl / CO mice had lower nonfasted In contrast to the other diets, plasma metabolite concen-
blood glucose concentrations (PGT = 0.003), but higher trations were neither affected by genotype nor affected by
NEFA (PGT = 0.006) and cholesterol plasma levels (PGT = sex in the CW group. However, like in the BE group, liver
0.021, Table 1). Both glucose (PS = 0.004) and cholesterol glycerol content was higher in females (PS < 0.001), and liver
concentrations were lower in females (PS = 0.002). glycogen content showed a trend toward being lower in
Triglyceride and free glycerol plasma levels, and liver glyc- Ghrl / mice than in WT mice (PGT = 0.092) and higher in
erol content were comparable across groups, similar to females than in males (PS = 0.077).
previous reports (54). Liver glycogen content, however,
Circulating and Fecal Hormones
showed a trend toward a genotype-sex interaction effect
(PGT:S = 0.054), with ghrelin deficiency increasing glyco- Although plasma ghrelin and leptin levels were affected
gen content in males, but decreasing it in females. by the dietary interventions (P < 0.001), for none of the diets
In BE animals, NEFA (PGT:S = 0.030), triglyceride (PGT:S = these levels were affected by genotype or sex (Table 2). In
0.020), and free glycerol levels (PGT:S = 0.044) showed inter- contrast, fecal corticosterone levels were higher in females
action effects of genotype and sex: although WT females than in males in the CO (PS = 0.002) and CW (PS = 0.027)

E500 AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

were able to develop binge-like eating similar to WTs in lon-


ger-term daily access paradigms (33, 34). It was based on
A these previous findings that we chose an intermittent model
to assess the longer-term effects of bingeing in our mice. The
reason for the difference in response to daily as opposed to
intermittent access models may be related to entrainment
effects, although the mechanisms for this remain to be

Figure 4. Fat distribution. Fat depot weights as a percentage of body


weight for CO mice (A), BE mice (B), and CW mice (C). Experimental
groups: WT M, wild-type male; Ghrl / M, ghrelin-deficient male; WT F,
wild-type female; Ghrl / F, ghrelin-deficient female. Group size n = 6–8
mice. Data are presented as means ± SE and were analyzed by two-way
ANOVA. Main effects were considered significant if P < 0.05. gP < 0.05,
(g)
P < 0.10 for genotype differences; sP < 0.05, (s)P < 0.10 for sex differen-
ces, determined by post hoc analysis. BE, binge eating; CO, chow only;
CW, chow and WD; WD, Western-style diet.

groups. In the BE group, however, only a trend toward a ge-


notype-sex interaction was observed (PGT:S = 0.086): Ghrl /
males had lower fecal corticosterone levels than all other
groups, albeit not significantly.

DISCUSSION
The aim of this study was to characterize the effects of sex
and ghrelin and their interaction on the development of
BED and its metabolic consequences in a model of binge-like
eating in mice. Using an intermittent access to a palatable
diet model, we successfully induced binge-like eating behav-
Figure 5. Glucose tolerance, as assessed by intraperitoneal glucose toler-
ior in mice. Unexpectedly, Ghrl / mice of both sexes devel- ance test. Blood glucose concentrations after glucose injection at 0 min
oped binge-like eating behavior to a similar extent as their and area under the curve of blood glucose levels in CO mice (A, B), BE
WT counterparts. Our observations are different from find- mice (C, D), and CW mice (E, F). Group size n = 5–8 mice. Data are pre-
ings in male Ghsr / mice, which were less prone to develop sented as means ± SE and were analyzed by two-way ANOVA. Main
effects were considered significant if P < 0.05. gP < 0.05 for genotype
binge-like eating in both short-term daily access (35) and differences; sP < 0.05, (s)P < 0.10 for sex differences, determined by post
long-term intermittent access BED mouse models (34). hoc analysis. BE, binge eating; CO, chow only; CW, chow and WD; WD,
However, it is important to note that male Ghsr / mice Western-style diet.

AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org E501


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

However, in the study by Egecioglu et al. (55), it was found


that this preference for rewarding food was lost in female
A Ghsr / mice. In contrast, the Ghrl / mice of both sexes in
our study maintained their preference for the rewarding
WD, and even lowered their chow intake, in both the BE and
CW dietary groups. The latter finding is thus more in line
with the unexpected effects of acute AG infusions described
in a binge-like eating rat model: acute ghrelin administration
increased preference for less rewarding, but healthier, chow
(33). It could be speculated that this indicates that ghrelin, as
a hormone, enhances healthy eating, perhaps even inde-
pendent of GHSR.
The differential effects of GHSR deficiency in these
paradigms, as well as our finding that binge-like eating is
not impaired in Ghrl / mice, may point to alternative
functions of GHSR, that are independent of peripheral
ghrelin, and vice versa. Indeed, in vitro evidence has
shown that GHSR has constitutive activity, which may
have physiological relevance (56, 57), although this is pres-
ently poorly understood (58). It is present in brain struc-
tures that are involved in reward signaling and memory
formation. Here, it is thought to form heteromers with
B other receptors, such as dopaminergic receptors (59, 60),
modulating their downstream signaling pathways. It is
still debated, however, whether ghrelin can access and
bind GHSR in these brain regions (61). For example,
although peripheral ghrelin administration has been
shown to strongly affect neuronal activity and connectiv-
ity in areas such as the ventral tegmental area (62), it needs
an intact arcuate nucleus to do so (63). However, others
have suggested that ghrelin might be able to penetrate
deeper into the brain (64), for example, by crossing into
the cerebrospinal fluid (65). Considering the difference
between our results and those obtained in Ghsr / mice, it
is pivotal to gain more clarity on how accessible different
regions of the brain are to ghrelin. Finally, differential
actions between AG and UAG might also contribute to the
differences observed between Ghrl / and Ghsr / mice.
In previous work by our group, we demonstrated that UAG
infusion affects metabolism in mice (45). This form of
ghrelin does not bind GHSR, in contrast to AG. Effects that
are observed in our Ghrl / mice, that express neither AG
nor UAG, might thus be attributable to decreased UAG
action.
An interesting observation in this study was the timing of
periods of reduced chow intake in Ghrl / relative to WT
mice following the binge period. This developed very early
in the study, as it was already observed during the second
week in Ghrl / females (Supplemental Fig. S2). Although
Figure 6. Gene expression profiles of inguinal WAT and liver. mRNA Ghrl / males mainly reduced their chow intake during
expression in inguinal WAT (A) and liver (B), normalized to WT CO males. the dark phase following WD access, Ghrl / females also
Group size n = 6–8 mice. Data are presented as means ± SE and were an-
showed a lower chow intake during the following nonaccess
alyzed by two-way ANOVA. gP < 0.05, (g)P < 0.10 for genotype differen-
ces; sP < 0.05, (s)P < 0.10 for sex differences, determined by post hoc day dark phase. This decreased food intake at the start of the
analysis. WAT: white adipose tissue; WT CO, wild-type chow only. dark phase in Ghrl / mice after a large meal has, to the best
of our knowledge, not been described before. Perhaps the
homeostatic drive to eat, particularly at the start of the
elucidated. Another notable difference between Ghrl / and active—dark—phase, is diminished in Ghrl / mice: either
Ghsr / mice occurred in the mice given access to both WD the balance between orexigenic and anorexigenic signaling
and chow. When given free access to both rewarding/high- could be disturbed, or ghrelin deficiency could worsen the
energy food and standard chow, WT mice spontaneously disruption of the circadian rhythms of the mice. Ghrelin
consumed most of their calories from the rewarding food. counteracts the effects of other gut hormones that regulate

E502 AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

/
Table 1. WT and ghrelin-deficient (Ghrl ) male (M) and female (F) mice
CO
Metabolite Sig. WT M (n = 5–8) Ghrl /
M (n = 8) WT F (n = 6–8) Ghrl /
F (n = 8)
Plasma, all mmol/L
Glucose GT,S 6.94 ± 0.16 6.13 ± 0.22 6.16 ± 0.27 5.30 ± 0.33
NEFA GT 0.66 ± 0.04 0.74 ± 0.03 0.60 ± 0.04 0.77 ± 0.06(g)
Triglycerides 0.69 ± 0.18 1.34 ± 0.24 1.31 ± 0.28 1.14 ± 0.29
Free glycerol 0.49 ± 0.12 0.83 ± 0.15 0.82 ± 0.15 0.80 ± 0.10
Cholesterol GT,S 2.30 ± 0.11 2.63 ± 0.09 1.72 ± 0.11 2.15 ± 0.25
Liver
Glycogen, mmol glucose/g liver () 85.4 ± 21.1 119.1 ± 29.8 165.6 ± 25.8 99.2 ± 22.0
Glycerol, mmol/g liver 17.7 ± 2.8 14.0 ± 1.1 13.9 ± 2.2 16.9 ± 2.0
BE
Metabolite Sig. WT M (n = 7–8) Ghrl /
M (n = 6) WT F (n = 7–8) Ghrl /
F (n = 8)
Plasma, all mmol/L
Glucose 6.21 ± 0.29 6.00 ± 0.26 6.87 ± 0.58 6.10 ± 0.30
NEFA  0.69 ± 0.04 0.63 ± 0.02 0.56 ± 0.04 0.71 ± 0.07
Triglycerides  1.17 ± 0.18 0.71 ± 0.11 0.52 ± 0.07 0.98 ± 0.28(g)
Free glycerol  0.70 ± 0.08 0.63 ± 0.06 0.44 ± 0.08 0.90 ± 0.22(g)
Cholesterol (GT),S 2.80 ± 0.10 2.87 ± 0.15 1.91 ± 0.08 2.39 ± 0.17
Liver
Glycogen, mmol glucose/g liver (GT) 191.1 ± 28.4 164.5 ± 30.0 183.3 ± 34.9 96.4 ± 14.5
Glycerol, mmol/g liver S 18.3 ± 1.4 16.1 ± 1.2 32.2 ± 9.2 34.4 ± 5.1(s)
CW
Metabolite Sig. WT M (n = 4–8) Ghrl /
M (n = 7) WT F (n = 5–6) Ghrl /
F (n = 7)
Plasma, all mmol/L
Glucose 6.93 ± 0.38 6.48 ± 0.12 6.86 ± 0.57 6.49 ± 0.43
NEFA 0.85 ± 0.09 0.84 ± 0.09 0.88 ± 0.13 0.82 ± 0.05
Triglycerides 2.31 ± 0.72 2.24 ± 0.61 1.91 ± 0.63 1.82 ± 0.37
Free glycerol 1.32 ± 0.32 1.25 ± 0.32 1.31 ± 0.39 1.15 ± 0.18
Cholesterol 3.50 ± 0.42 4.14 ± 0.33 3.34 ± 0.15 3.62 ± 0.18
Liver
Glycogen, mmol glucose/g liver (GT),(S) 137.8 ± 29.5 92.2 ± 31.4 194.3 ± 32.0 140.3 ± 20.1
Glycerol, mmol/g liver S 49.4 ± 5.9 54.0 ± 5.4 81.4 ± 8.1s 82.8 ± 5.3s
Data were analyzed by two-way ANOVA with a Tukey HSD post hoc analysis, or Bonferroni analysis. Sig. lists significant main effects
(P < 0.05): GT, main effect for genotype; S, main effect for sex; , interaction effect. Parenthesized symbols indicate a trend toward sig-
nificance (P < 0.10). gP < 0.05, or (g)P < 0.10 vs. WT mice of the same sex; sP < 0.05, or (s)P < 0.10 vs. males of the same genotype. WT M,
wild-type male; Ghrl / M, ghrelin-deficient male; WT F, wild-type female; Ghrl / F, ghrelin-deficient female. BE, binge eating; CO,
chow only; CW, chow and WD; WD, Western-style diet.

satiety (66, 67). These satiety hormones are released not only in females. The modulation of physical activity by ghrelin
during meals, but also in the subsequent hours (68). Thus signaling has been described by several authors (41, 72–74).
ghrelin deficiency may have resulted in an inadequate orexi- In male mice, acute AG administration was shown to
genic drive to counteract the actions of these hormones fol- enhance locomotor activity (73). Consistently, Mifune et al.
lowing the large meal during the light phase. In addition, the (72) demonstrated that male Ghrl / mice have lower wheel-
disruption of ghrelin signaling affects circadian rhythmicity running activity than WT mice under time-restricted feed-
in mice (69, 70), conceivably decreasing the drive to move ing. However, other authors reported little to no effects of
and eat in the early dark phase. However, we did not observe ghrelin or GHSR deficiency in nonrearing locomotor activity
reductions in locomotor activity at this time, which were in male or female mice (41, 74). Combined with these previ-
observed in Ghsr / mice (71). The mechanism of the effects ous findings, our results suggest that the impact of ghrelin
of ghrelin deficiency on postbinge food intake thus remains signaling on physical activity is not only dependent on die-
to be elucidated. tary intervention, but also on the sex of the mice.
Besides the differentially timed reduction in food intake, This sex difference in locomotor activity has been well
we also observed a strong interaction effect of sex and geno- described for C57BL/6J mice: especially young females have
type on locomotor activity in the binge paradigm. Namely, increased locomotor activity compared with males of a similar
on nonaccess days, Ghrl / females walked less than WT age (75). It is important to note, though, that locomotor activ-
females, whereas ghrelin deficiency did not affect locomotor ity can be affected by social isolation. Although some authors
activity in males. Although in the other interventions, no report increased locomotor activity in socially isolated males
such genotype-sex interaction was observed, in CO mice, compared with group-housed males (76), others report sex
females of both genotypes had increased locomotor activity effects in locomotor activity upon isolation: females were
compared with males and mice of both sexes with ghrelin shown to have higher locomotor activity than males when
deficiency had lower locomotor activity than wild types. In housed individually (77). Moreover, Yamada et al. (77)
CW mice, locomotor activity was only significantly increased reported sexually dimorphic alterations in ghrelin signaling

AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org E503


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

/
Table 2. WT and ghrelin-deficient (Ghrl ) male (M) and female (F) mice
CO
Hormone Sig. WT M (n = 5–8) Ghrl /
M (n = 8) WT F (n = 6-8) Ghrl /
F (n = 8)
Plasma ghrelin
AG, pg/mL 134.4 ± 22.9 108.9 ± 0.142
UAG, pg/mL 863.0 ± 125.4 778.7 ± 103.5
AG:UAG 0.153 ± 0.032 0.142 ± 0.050
Plasma leptin, ng/mL 1.68 ± 0.29 1.16 ± 0.20 1.21 ± 0.29 1.29 ± 0.26
Plasma adiponectin, mg/mL (GT),S 14.5 ± 0.5 15.9 ± 0.3 20.9 ± 1.1s 23.5 ± 1.6s
Fecal corticosterone (nmol/day) S 0.060 ± 0.007 0.059 ± 0.010 0.098 ± 0.011 0.150 ± 0.045(s)
BE
Hormone Sig. WT M (n = 4–8) Ghrl /
M (n = 7) WT F (n = 5–6) Ghrl /
F (n = 7)
Plasma ghrelin
AG, pg/mL 63.4 ± 6.4 71.2 ± 8.7
UAG, pg/mL 513.2 ± 129.9 743.1 ± 81.5
AG:UAG 0.138 ± 0.033 0.098 ± 0.008
Plasma leptin, ng/mL 2.12 ± 0.51 1.28 ± 0.29 1.30 ± 0.30 0.77 ± 0.07
Plasma adiponectin, mg/mL S 12.3 ± 0.9 12.8 ± 0.7 18.9 ± 1.8s 22.3 ± 1.4s
Fecal corticosterone, nmol/day () 0.222 ± 0.022 0.134 ± 0.019 0.215 ± 0.025 0.235 ± 0.047
CW
Hormone Sig. WT M (n = 7–8) Ghrl /
M (n = 6) WT F (n = 7–8) Ghrl /
F (n = 8)
Plasma ghrelin
AG, pg/mL) 42.9 ± 9.4 49.9 ± 7.0
UAG, pg/mL 408.5 ± 55.1 410.2 ± 53.0
AG:UAG 0.123 ± 0.037 0.122 ± 0.008
Plasma leptin, ng/mL 4.64 ± 0.89 5.02 ± 0.77 4.38 ± 0.82 5.67 ± 0.87
Plasma adiponectin, mg/mL S 15.2 ± 0.9 17.7 ± 1.3 23.6 ± 1.4s 22.2 ± 2.2s
Fecal corticosterone (nmol/day) S 0.021 ± 0.003 0.035 ± 0.020 0.057 ± 0.010 0.045 ± 0.047
Ghrelin data were analyzed by one-way ANOVA, and corticosterone data by two-way ANOVA with a Tukey HSD post hoc analysis, or
Bonferroni analysis. Sig. lists significant main effects (P < 0.05): GT, main effect for genotype; S, main effect for sex; , interaction effect.
Parenthesized symbols indicate a trend toward significance (P < 0.10). sP < 0.05, or (s)P < 0.10 vs. males of the same genotype. WT M,
wild-type male; Ghrl / M, ghrelin-deficient male; WT F, wild-type female; Ghrl / F, ghrelin-deficient female. BE, binge eating; CO,
chow only; CW, chow and WD; WD, Western-style diet.

upon social isolation. Altogether, future research into the sex- females. It is well known that female rodents are more glu-
specific effects of ghrelin deficiency on the locomotor activity cose tolerant than males (80). Moreover, the improvement in
should take the social environment of the mice into account. glucose tolerance upon deletion of genes in the ghrelin sig-
Another striking sex-dependent effect of ghrelin defi- naling pathway has been extensively reported in male mice
ciency was observed in the CW group: Ghrl / females had a (81–85). However, Ghsr deficiency does not improve glucose
higher energy expenditure than any of the other groups on tolerance in female mice, which, generally, already have bet-
the same diet. Energy expenditure is affected by body size, ter glucose tolerance than males (74). Consistently, we show
and especially fat-free mass, by food intake, and by physical that the AUC of Ghrl / males in the IPGTT remains similar
activity (78). Compared with CW males, food intake and to that of the female mice, regardless of diet. Our findings on
physical activity were higher, but the fat-free mass was lower gene expression profiles in WAT, in particular, may provide
in CW females, regardless of genotype. These factors partly one of the mechanisms by which glucose homeostasis is
explain the difference between male and female mice, but improved in Ghrl / mice. Cd68 and Cd11c, markers for mac-
not between Ghrl / and WT female mice. Another explana- rophage abundance and proinflammatory status, respec-
tion could lie in a difference in another component of basal tively, were suppressed in Ghrl / mice. This effect occurred
metabolic rate, such as thermogenesis. In Ghsr / mice, particularly in the CW, and to a lesser extent in the BE group.
thermogenesis has been shown to be enhanced in brown and Adipose macrophages play a major role in modulating insu-
white adipose tissues upon aging (79). Indeed, microscopic lin sensitivity and like our findings in Ghrl-deficient mice,
analysis of the inguinal WAT, a browning-prone depot, macrophage-specific deletion of Ghsr leads to an improved
revealed more brown-like areas in Ghrl / than in WT adipose inflammatory profile with aging (86). In addition to
females (data not shown). Consistently, we showed an up- direct markers of macrophage infiltration and phenotype,
regulation of Ucp1 expression in this depot. This upregula- the gene expression of Fgf21 was also increased in the ingui-
tion did not reach significance in male Ghrl / mice, nal WAT of the Ghrl / mice relative to WTs. FGF21 has been
suggesting that ghrelin deficiency not only sex-dependently shown to promote anti-inflammatory M2 macrophage polar-
alters behaviors as discussed above, but also energy metabo- ization, as well as the insulin sensitivity of subcutaneous fat
lism through enhanced thermogenesis in mice. (87). Further investigation of the possible modulation of adi-
Furthermore, in both the CO and CW groups, we demon- pose tissue macrophage recruitment and phenotype by ghre-
strated that ghrelin deficiency improved glucose tolerance in lin deficiency under conditions of high fat-induced obesity
males, but it did not significantly affect glucose tolerance in is thus warranted.

E504 AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

As mentioned earlier, a possible drawback of the current 3. American Psychiatric Association. Eating and feeding disorders. In:
experiment was the prolonged individual housing of the Diagnostic and Statistical Manual of Mental Disorders (5th ed.).
mice. Individual housing is known to affect several meta- Washington, DC: American Psychiatric Association, 2013.
4. Van Alsten SC, Duncan AE. Lifetime patterns of comorbidity in eat-
bolic parameters, such as food intake and visceral fat mass, ing disorders: An approach using sequence analysis. Eur Eat Disord
when comparing socially housed mice to individually Rev 28: 709–723, 2020. doi:10.1002/erv.2767.
housed mice (88). The sex of the mice did not appear to mod- 5. Udo T, Grilo CM. Psychiatric and medical correlates of DSM-5 eating
ulate these effects (88). In our study, all groups were exposed disorders in a nationally representative sample of adults in the United
to the same social isolation conditions. Fecal corticosterone States. Int J Eat Disord 52: 42–50, 2019. doi:10.1002/eat.23004.
6. Erskine HE, Whiteford HA. Epidemiology of binge eating disor-
levels, as an assessment of stress, in these mice were lower
der. Curr Opin Psychiatry 31: 462–470, 2018. doi:10.1097/YCO.
than or comparable to levels observed in group-housed ani- 0000000000000449.
mals from our previous study (46). Importantly, corticoster- 7. Czyzyk TA, Sahr AE, Statnick MA. A model of binge-like eating
one levels were comparable between WT and Ghrl / behavior in mice that does not require food deprivation or stress.
females on the same diet, whereas most of the described Obesity (Silver Spring) 18: 1710–1717, 2010. doi:10.1038/oby.2010.46.
effects of ghrelin deficiency were observed in female mice. 8. Corwin RL, Wojnicki FH, Fisher JO, Dimitriou SG, Rice HB, Young
MA. Limited access to a dietary fat option affects ingestive behavior
Based on these findings, we conclude that social isolation but not body composition in male rats. Physiol Behav 63: 545–553,
stress did not have a major impact on the observed effects. 1998. doi:10.1016/s0031-9384(98)00201-7.
In conclusion, we found that binge-like eating can be 9. Bake T, Murphy M, Morgan DG, Mercer JG. Large, binge-type
induced successfully in both male and female ghrelin-defi- meals of high fat diet change feeding behaviour and entrain food an-
cient mice, as measured by WD intake. We showed that par- ticipatory activity in mice. Appetite 77: 60–71, 2014. doi:10.1016/j.
appet.2014.02.020.
ticularly females were affected by ghrelin deficiency, both in
10. Babbs RK, Wojnicki FH, Corwin RL. Effect of 2-hydroxyestradiol on
the BE and CW groups. Overall, this study provides extensive binge intake in rats. Physiol Behav 103: 508–512, 2011. doi:10.1016/j.
evidence that both diet and sex should be taken into account physbeh.2011.03.029.
when researching ghrelin signaling. 11. Klump KL, Racine S, Hildebrandt B, Sisk CL. Sex differences in
binge eating patterns in male and female adult rats. Int J Eat Disord
46: 729–736, 2013. doi:10.1002/eat.22139.
SUPPLEMENTAL DATA 12. Spierling SR, Kreisler AD, Williams CA, Fang SY, Pucci SN, Kines
KT, Zorrilla EP. Intermittent, extended access to preferred food
Supplemental Tables S1–S3: https://doi.org/10.6084/m9.figshare. leads to escalated food reinforcement and cyclic whole-body me-
17197274 and Figures S1 and S2: https://doi.org/10.6084/m9. tabolism in rats: s.ex differences and individual vulnerability. Physiol
figshare.19384001. Behav 192: 3–16, 2018. doi:10.1016/j.physbeh.2018.04.001.
13. Sinclair EB, Hildebrandt BA, Culbert KM, Klump KL, Sisk CL.
ACKNOWLEDGMENTS Preliminary evidence of sex differences in behavioral and neural
responses to palatable food reward in rats. Physiol Behav 176: 165–
We gratefully acknowledge Prof. Paul Pfluger for the gift of the 173, 2017. doi:10.1016/j.physbeh.2017.03.042.
Ghrl / mice. 14. Novelle MG, Dieguez C. Updating gender differences in the control
of homeostatic and hedonic food intake: implications for binge eat-
ing disorder. Mol Cell Endocrinol 497: 110508, 2019. doi:10.1016/j.
GRANTS mce.2019.110508.
15. Balodis IM, Grilo CM, Potenza MN. Neurobiological features of
This work was supported by departmental laboratory funds (to binge eating disorder. CNS Spectr 20: 557–565, 2015. doi:10.1017/
K. Prins, M. Huisman, A. McLuskey, R. Mies, B. Karels, P. J. D. S1092852915000814.
Delhanty, and J. A. Visser). 16. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K.
Ghrelin is a growth-hormone-releasing acylated peptide from stom-
DISCLOSURES ach. Nature 402: 656–660, 1999. doi:10.1038/45230.
17. Tscho € p MH, Smiley DL, Heiman ML. Ghrelin induces adiposity in
No conflicts of interest, financial or otherwise, are declared by rodents. Nature 407: 908–913, 2000. doi:10.1038/35038090.
the authors. 18. Wren AM, Small CJ, Ward HL, Murphy KG, Dakin CL, Taheri S,
Kennedy AR, Roberts GH, Morgan DGA, Ghatei MA, Bloom SR.
The novel hypothalamic peptide ghrelin stimulates food intake and
AUTHOR CONTRIBUTIONS growth hormone secretion. Endocrinology 141: 4325–4328, 2000.
K.P., P.J.D.D., and J.A.V. conceived and designed research; doi:10.1210/endo.141.11.7873.
K.P., M.H., A.M., R.M., B.K., and P.J.D.D. performed experiments; 19. Delhanty PJ, Neggers SJ, Van der Lely AJ. Should we consider
des-acyl ghrelin as a separate hormone and if so, what does it do?
K.P. and M.H. analyzed data; K.P., P.J.D.D., and J.A.V. interpreted
Front Horm Res 42: 163–174, 2014. doi:10.1159/000358345.
results of experiments; K.P. prepared figures; K.P., P.J.D.D., and 20. Williams RL, Wood LG, Collins CE, Morgan PJ, Callister R. Energy
J.A.V. drafted manuscript; K.P., P.J.D.D., and J.A.V. edited and re- homeostasis and appetite regulating hormones as predictors of
vised manuscript; K.P., M.H., A.M., R.M., B.K., P.J.D.D., and J.A.V. weight loss in men and women. Appetite 101: 1–7, 2016. doi:10.1016/j.
approved final version of manuscript. appet.2016.02.153.
21. Monteleone P, Fabrazzo M, Tortorella A, Martiadis V, Serritella
REFERENCES C, Maj M. Circulating ghrelin is decreased in non-obese and
obese women with binge eating disorder as well as in obese
1. Santomauro DF, Melen S, Mitchison D, Vos T, Whiteford H, Ferrari non-binge eating women, but not in patients with bulimia nerv-
AJ. The hidden burden of eating disorders: an extension of esti- osa. Psychoneuroendocrinology 30: 243–250, 2005. doi:10.
mates from the Global Burden of Disease Study 2019. Lancet 1016/j.psyneuen.2004.07.004.
Psychiatry 8: 320–328, 2021. doi:10.1016/S2215-0366(21)00040-7. 22. Geliebter A, Hashim SA, Gluck ME. Appetite-related gut peptides,
2. Udo T, Grilo CM. Prevalence and correlates of DSM-5-defined eating ghrelin, PYY, and GLP-1 in obese women with and without binge eat-
disorders in a nationally representative sample of U.S. adults. Biol ing disorder (BED). Physiol Behav 94: 696–699, 2008. doi:10.1016/j.
Psychiatry 84: 345–354, 2018. doi:10.1016/j.biopsych.2018.03.014. physbeh.2008.04.013.

AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org E505


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

23. Geliebter A, Gluck ME, Hashim SA. Plasma ghrelin concentrations 41. Pfluger PT, Kirchner H, Gunnel S, Schrott B, Perez-Tilve D, Fu S,
are lower in binge-eating disorder. J Nutr 135: 1326–1330, 2005. Benoit SC, Horvath T, Joost HG, Wortley KE, Sleeman MW, Tschop
doi:10.1093/jn/135.5.1326. MH. Simultaneous deletion of ghrelin and its receptor increases
24. Hernandez D, Mehta N, Geliebter A. Meal-related acyl and des-acyl motor activity and energy expenditure. Am J Physiol Gastrointest
ghrelin and other appetite-related hormones in people with obesity Liver Physiol 294: G610–G618, 2008. doi:10.1152/ajpgi.00321.2007.
and binge eating. Obesity (Silver Spring) 27: 629–635, 2019. doi:10. 42. Wortley KE, Anderson KD, Garcia K, Murray JD, Malinova L, Liu R,
1002/oby.22431. Moncrieffe M, Thabet K, Cox HJ, Yancopoulos GD, Wiegand SJ,
25. Munsch S, Biedert E, Meyer AH, Herpertz S, Beglinger C. CCK, Sleeman MW. Genetic deletion of ghrelin does not decrease food
ghrelin, and PYY responses in individuals with binge eating disorder intake but influences metabolic fuel preference. Proc Natl Acad Sci
before and after a cognitive behavioral treatment (CBT). Physiol USA 101: 8227–8232, 2004. doi:10.1073/pnas.0402763101.
Behav 97: 14–20, 2009. doi:10.1016/j.physbeh.2009.01.015. 43. Solmi M, Radua J, Olivola M, Croce E, Soardo L, Salazar de Pablo
26. Manfredi L, Accoto A, Couyoumdjian A, Conversi D. A systematic G, Il Shin J, Kirkbride JB, Jones P, Kim JH, Kim JY, Carvalho AF,
review of genetic polymorphisms associated with binge eating dis- Seeman MV, Correll CU, Fusar-Poli P. Age at onset of mental disor-
order. Nutrients 13: 848, 2021. doi:10.3390/nu13030848. ders worldwide: large-scale meta-analysis of 192 epidemiological
27. Iceta S, Julien B, Seyssel K, Lambert-Porcheron S, Segrestin B, studies. Mol Psychiatry 27: 281–295, 2022. doi:10.1038/s41380-021-
Blond E, Cristini P, Laville M, Disse E. Ghrelin concentration as an 01161-7.
indicator of eating-disorder risk in obese women. Diabetes Metab 44. van Houten EL, Kramer P, McLuskey A, Karels B, Themmen AP,
45: 160–166, 2019. doi:10.1016/j.diabet.2018.01.006. Visser JA. Reproductive and metabolic phenotype of a mouse
28. Pagotto U, Gambineri A, Pelusi C, Genghini S, Cacciari M, Otto model of PCOS. Endocrinology 153: 2861–2869, 2012. doi:10.1210/
B, Castaneda T, Tschop M, Pasquali R. Testosterone replace- en.2011-1754.
ment therapy restores normal ghrelin in hypogonadal men. J 45. Delhanty PJ, Huisman M, Baldeon-Rojas LY, van den Berge I,
Clin Endocrinol Metab 88: 4139–4143, 2003. doi:10.1210/ Grefhorst A, Abribat T, Leenen PJ, Themmen AP, van der Lely AJ.
jc.2003-030554. Des-acyl ghrelin analogs prevent high-fat-diet-induced dysregula-
29. Paulo RC, Brundage R, Cosma M, Mielke KL, Bowers CY, Veldhuis tion of glucose homeostasis. FASEB J 27: 1690–1700, 2013.
JD. Estrogen elevates the peak overnight production rate of acy- doi:10.1096/fj.12-221143.
lated ghrelin. J Clin Endocrinol Metab 93: 4440–4447, 2008. 46. Kaikaew K, Steenbergen J, van Dijk TH, Grefhorst A, Visser JA.
doi:10.1210/jc.2008-0783. Sex difference in corticosterone-induced insulin resistance in mice.
30. Kellokoski E, Poykko SM, Karjalainen AH, Ukkola O, Heikkinen J, Endocrinology 160: 2367–2387, 2019. doi:10.1210/en.2019-00194.
Kesaniemi YA, Horkko S. Estrogen replacement therapy increases 47. van den Beukel JC, Grefhorst A, Quarta C, Steenbergen J,
plasma ghrelin levels. J Clin Endocrinol Metab 90: 2954–2963, Mastroberardino PG, Lombes M, Delhanty PJ, Mazza R, Pagotto U,
2005. doi:10.1210/jc.2004-2016. van der Lely AJ, Themmen AP. Direct activating effects of adreno-
31. Moran LJ, Noakes M, Clifton PM, Wittert GA, Tomlinson L, Galletly corticotropic hormone (ACTH) on brown adipose tissue are attenu-
C, Luscombe ND, Norman RJ. Ghrelin and measures of satiety are ated by corticosterone. FASEB J 28: 4857–4867, 2014. doi:10.1096/
altered in polycystic ovary syndrome but not differentially affected fj.14-254839.
by diet composition. J Clin Endocrinol Metab 89: 3337–3344, 2004. 48. Grefhorst A, van den Beukel JC, Dijk W, Steenbergen J, Voortman
doi:10.1210/jc.2003-031583. GJ, Leeuwenburgh S, Visser TJ, Kersten S, Friesema ECH,
32. Gambineri A, Pagotto U, Tscho € p M, Vicennati V, Manicardi E, Themmen APN, Visser JA. Multiple effects of cold exposure on liv-
Carcello A, Cacciari M, De Iaso R, Pasquali R. Anti-androgen ers of male mice. J Endocrinol 238: 91–106, 2018. doi:10.1530/JOE-
treatment increases circulating ghrelin levels in obese women 18-0076.
with polycystic ovary syndrome. J Endocrinol 26: 629–634, 2003. 49. Feillet C, Guerin S, Lonchampt M, Dacquet C, Gustafsson JA,
doi:10.1007/BF03347020. Delaunay F, Teboul M. Sexual dimorphism in circadian physiology is
33. Bake T, Hellgren KT, Dickson SL. Acute ghrelin changes food pref- altered in LXRalpha deficient mice. PLoS One 11: e0150665, 2016.
erence from a high-fat diet to chow during binge-like eating in doi:10.1371/journal.pone.0150665.
rodents. J Neuroendocrinol 29, 2017. doi:10.1111/jne.12463. 50. Weir JBdV. New methods for calculating metabolic rate with special
34. King SJ, Rodrigues T, Watts A, Murray E, Wilson A, Abizaid A. reference to protein metabolism. J Physiol 109: 1–9, 1949. doi:10.1113/
Investigation of a role for ghrelin signaling in binge-like feeding in jphysiol.1949.sp004363.
mice under limited access to high-fat diet. Neuroscience 319: 233– 51. Foerster T. SableBase: Sable Systems Basic Functions. R package
245, 2016. doi:10.1016/j.neuroscience.2016.01.004. version 1.0.1. 2017.
35. Valdivia S, Cornejo MP, Reynaldo M, De Francesco PN, Perello M. 52. Kassambara A. rstatix: Pipe-Friendly Framework for Basic Statistical
Escalation in high fat intake in a binge eating model differentially Tests. R package version 0.6.0. 2020.
engages dopamine neurons of the ventral tegmental area and 53. Ayala JE, Samuel VT, Morton GJ, Obici S, Croniger CM, Shulman
requires ghrelin signaling. Psychoneuroendocrinology 60: 206–216, GI, Wasserman DH, McGuinness OP; Consortium NIHMMPC.
2015. doi:10.1016/j.psyneuen.2015.06.018. Standard operating procedures for describing and performing meta-
36. Yamada C. Relationship between orexigenic peptide ghrelin signal, bolic tests of glucose homeostasis in mice. Dis Model Mech 3: 525–
gender difference and disease. Int J Mol Sci 22: 3763, 2021. 534, 2010. doi:10.1242/dmm.006239.
doi:10.3390/ijms22073763. 54. Ma X, Lin L, Qin G, Lu X, Fiorotto M, Dixit VD, Sun Y. Ablations of
37. Clegg DJ, Brown LM, Zigman JM, Kemp CJ, Strader AD, Benoit ghrelin and ghrelin receptor exhibit differential metabolic pheno-
SC, Woods SC, Mangiaracina M, Geary N. Estradiol-dependent types and thermogenic capacity during aging. PLoS One 6: e16391,
decrease in the orexigenic potency of ghrelin in female rats. 2011. doi:10.1371/journal.pone.0016391.
Diabetes 56: 1051–1058, 2007 [Erratum in Diabetes 56:2649, 2007]. 55. Egecioglu E, Jerlhag E, Salome N, Skibicka KP, Haage D, Bohlooly
doi:10.2337/db06-0015. YM, Andersson D, Bjursell M, Perrissoud D, Engel JA, Dickson SL.
38. Yamada C, Iizuka S, Nahata M, Hattori T, Takeda H. Vulnerability to Ghrelin increases intake of rewarding food in rodents. Addict Biol 15:
psychological stress-induced anorexia in female mice depends on 304–311, 2010. doi:10.1111/j.1369-1600.2010.00216.x.
blockade of ghrelin signal in nucleus tractus solitarius. Br J 56. Fernandez G, Cabral A, Andreoli MF, Labarthe A, M'Kadmi C,
Pharmacol 177: 4666–4682, 2020. doi:10.1111/bph.15219. Ramos JG, Marie J, Fehrentz JA, Epelbaum J, Tolle V, Perello M.
39. Frazao R, Dungan Lemko HM, da Silva RP, Ratra DV, Lee CE, Evidence supporting a role for constitutive ghrelin receptor signaling
Williams KW, Zigman JM, Elias CF. Estradiol modulates Kiss1 neuro- in fasting-induced hyperphagia in male mice. Endocrinology 159:
nal response to ghrelin. Am J Physiol Endocrinol Metab 306: E606– 1021–1034, 2018. doi:10.1210/en.2017-03101.
E614, 2014. doi:10.1152/ajpendo.00211.2013. 57. Cornejo MP, Castrogiovanni D, Schioth HB, Reynaldo M, Marie
40. Farkas I, Vastagh C, Sarvari M, Liposits Z. Ghrelin decreases firing J, Fehrentz JA, Perello M. Growth hormone secretagogue re-
activity of gonadotropin-releasing hormone (GnRH) neurons in an ceptor signalling affects high-fat intake independently of plasma
estrous cycle and endocannabinoid signaling dependent manner. levels of ghrelin and LEAP2, in a 4-day binge eating model. J
PLoS One 8: e78178, 2013. doi:10.1371/journal.pone.0078178. Neuroendocrinol 31: e12785, 2019. doi:10.1111/jne.12785.

E506 AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.
SEX-DEPENDENT EFFECTS OF GHRELIN DEFICIENCY IN MICE

58. Cornejo MP, Mustafa ER, Cassano D, Baneres JL, Raingo J, Perello 73. Jerlhag E, Egecioglu E, Dickson SL, Andersson M, Svensson L,
M. The ups and downs of growth hormone secretagogue receptor Engel JA. Ghrelin stimulates locomotor activity and accumbal dopa-
signaling. FEBS J 24: 7213–7229, 2021. doi:10.1111/febs.15718. mine-overflow via central cholinergic systems in mice: implications
59. Kern A, Albarran-Zeckler R, Walsh HE, Smith RG. Apo-ghrelin re- for its involvement in brain reward. Addict Biol 11: 45–54, 2006.
ceptor forms heteromers with DRD2 in hypothalamic neurons and is doi:10.1111/j.1369-1600.2006.00002.x.
essential for anorexigenic effects of DRD2 agonism. Neuron 73: 74. Silver Z, Abbott-Tate S, Hyland L, Sherratt F, Woodside B, Abizaid
317–332, 2012. doi:10.1016/j.neuron.2011.10.038. A. Ghrelin receptor signaling is not required for glucocorticoid-
60. Kern A, Mavrikaki M, Ullrich C, Albarran-Zeckler R, Brantley AF, induced obesity in female mice. J Endocrinol 250: 37–48, 2021.
Smith RG. Hippocampal dopamine/DRD1 signaling dependent on doi:10.1530/JOE-20-0579.
the ghrelin receptor. Cell 163: 1176–1190, 2015. doi:10.1016/j.cell. 75. Konig C, Plank AC, Kapp A, Timotius IK, von Horsten S,
2015.10.062. Zimmermann K. Thirty mouse strain survey of voluntary physical
61. Perello M, Cabral A, Cornejo MP, De Francesco PN, Fernandez activity and energy expenditure: influence of strain, sex and day-
G, Uriarte M. Brain accessibility delineates the central effects of night variation. Front Neurosci 14: 531, 2020. doi:10.3389/fnins.
circulating ghrelin. J Neuroendocrinol 31: e12677, 2019. doi:10. 2020.00531.
1111/jne.12677. 76. Ros-Simo C, Valverde O. Early-life social experiences in mice affect
62. Abizaid A, Liu ZW, Andrews ZB, Shanabrough M, Borok E, emotional behaviour and hypothalamic-pituitary-adrenal axis func-
Elsworth JD, Roth RH, Sleeman MW, Picciotto MR, Tschop MH, tion. Pharmacol Biochem Behav 102: 434–441, 2012. doi:10.1016/j.
Gao XB, Horvath TL. Ghrelin modulates the activity and synaptic pbb.2012.06.001.
input organization of midbrain dopamine neurons while promoting 77. Yamada C, Saegusa Y, Nahata M, Sadakane C, Hattori T, Takeda
appetite. J Clin Invest 116: 3229–3239, 2006. doi:10.1172/JCI29867. H. Influence of aging and gender differences on feeding behavior
63. Cabral A, Valdivia S, Fernandez G, Reynaldo M, Perello M. and ghrelin-related factors during social isolation in mice. PLoS One
Divergent neuronal circuitries underlying acute orexigenic effects of 10: e0140094, 2015. doi:10.1371/journal.pone.0140094.
peripheral or central ghrelin: critical role of brain accessibility. J 78. Westerterp KR. Control of energy expenditure in humans. Eur J Clin
Neuroendocrinol 26: 542–554, 2014. doi:10.1111/jne.12168. Nutr 71: 340–344, 2017. doi:10.1038/ejcn.2016.237.
64. Rhea EM, Salameh TS, Gray S, Niu J, Banks WA, Tong J. Ghrelin 79. Lin L, Saha PK, Ma X, Henshaw IO, Shao L, Chang BHJ, Buras ED,
transport across the blood-brain barrier can occur independently of Tong Q, Chan L, McGuinness OP, Sun Y. Ablation of ghrelin recep-
the growth hormone secretagogue receptor. Mol Metab 18: 88–96, tor reduces adiposity and improves insulin sensitivity during aging
2018. doi:10.1016/j.molmet.2018.09.007. by regulating fat metabolism in white and brown adipose tissues.
65. Uriarte M, De Francesco PN, Fernandez G, Castrogiovanni D,
Aging Cell 10: 996–1010, 2011. doi:10.1111/j.1474-9726.2011.00740.x.
D'Arcangelo M, Imbernon M, Cantel S, Denoyelle S, Fehrentz JA,
80. Reynolds TH, Dalton A, Calzini L, Tuluca A, Hoyte D, Ives SJ. The
Praetorius J, Prevot V, Perello M. Circulating ghrelin crosses the
impact of age and sex on body composition and glucose sensitivity
blood-cerebrospinal fluid barrier via growth hormone secretagogue
in C57BL/6J mice. Physiol Rep 7: e13995, 2019. doi:10.14814/
receptor dependent and independent mechanisms. Mol Cell
phy2.13995.
Endocrinol 538: 111449, 2021. doi:10.1016/j.mce.2021.111449.
81. Zigman JM, Nakano Y, Coppari R, Balthasar N, Marcus JN, Lee CE,
66. Zhang W, Waise TMZ, Toshinai K, Tsuchimochi W, Naznin F, Islam
Jones JE, Deysher AE, Waxman AR, White RD, Williams TD,
MN, Tanida R, Sakoda H, Nakazato M. Functional interaction
between Ghrelin and GLP-1 regulates feeding through the vagal Lachey JL, Seeley RJ, Lowell BB, Elmquist JK. Mice lacking ghrelin
afferent system. Sci Rep 10: 18415, 2020. doi:10.1038/s41598-020- receptors resist the development of diet-induced obesity. J Clin
75621-5. Invest 115: 3564–3572, 2005. doi:10.1172/JCI26002.
67. Blanco AM, Bertucci JI, Valenciano AI, Delgado MJ, Unniappan S. 82. Sun Y, Asnicar M, Saha PK, Chan L, Smith RG. Ablation of ghrelin
Ghrelin suppresses cholecystokinin (CCK), peptide YY (PYY) and improves the diabetic but not obese phenotype of ob/ob mice. Cell
glucagon-like peptide-1 (GLP-1) in the intestine, and attenuates the Metab 3: 379–386, 2006. doi:10.1016/j.cmet.2006.04.004.
anorectic effects of CCK, PYY and GLP-1 in goldfish (Carassius aura- 83. Gray SM, Page LC, Tong J. Ghrelin regulation of glucose metabo-
tus). Horm Behav 93: 62–71, 2017. doi:10.1016/j.yhbeh.2017.05.004. lism. J Neuroendocrinol 31: e12705, 2019. doi:10.1111/jne.12705.
68. Engelstoft MS, Schwartz TW. Opposite regulation of ghrelin and 84. Longo KA, Charoenthongtrakul S, Giuliana DJ, Govek EK,
glucagon-like peptide-1 by metabolite G-protein-coupled receptors. McDonagh T, Qi Y, DiStefano PS, Geddes BJ. Improved insulin sen-
Trends Endocrinol Metab 27: 665–675, 2016. doi:10.1016/j.tem. sitivity and metabolic flexibility in ghrelin receptor knockout mice.
2016.07.001. Regul Pept 150: 55–61, 2008. doi:10.1016/j.regpep.2008.03.011.
69. Lamont EW, Bruton J, Blum ID, Abizaid A. Ghrelin receptor-knock- 85. Wortley KE, del Rincon JP, Murray JD, Garcia K, Iida K, Thorner
out mice display alterations in circadian rhythms of activity and feed- MO, Sleeman MW. Absence of ghrelin protects against early-onset
ing under constant lighting conditions. Eur J Neurosci 39: 207–217, obesity. J Clin Invest 115: 3573–3578, 2005. doi:10.1172/JCI26003.
2014. doi:10.1111/ejn.12390. 86. Lin L, Lee JH, Buras ED, Yu K, Wang R, Smith CW, Wu H, Sheikh-
70. LeSauter J, Hoque N, Weintraub M, Pfaff DW, Silver R. Stomach Hamad D, Sun Y. Ghrelin receptor regulates adipose tissue inflam-
ghrelin-secreting cells as food-entrainable circadian clocks. Proc mation in aging. Aging (Albany NY) 8: 178–191, 2016. doi:10.18632/
Natl Acad Sci USA 106: 13582–13587, 2009. aging.100888.
71. Blum ID, Patterson Z, Khazall R, Lamont EW, Sleeman MW, 87. Li H, Wu G, Fang Q, Zhang M, Hui X, Sheng B, Wu L, Bao Y, Li P,
Horvath TL, Abizaid A. Reduced anticipatory locomotor responses Xu A, Jia W. Fibroblast growth factor 21 increases insulin sensitivity
to scheduled meals in ghrelin receptor deficient mice. Neuroscience through specific expansion of subcutaneous fat. Nat Commun 9:
164: 351–359, 2009. doi:10.1016/j.neuroscience.2009.08.009. 272, 2018. doi:10.1038/s41467-017-02677-9.
72. Mifune H, Tajiri Y, Sakai Y, Kawahara Y, Hara K, Sato T, Nishi Y, 88. Schipper L, Harvey L, van der Beek EM, van Dijk G. Home alone: a
Nishi A, Mitsuzono R, Kakuma T, Kojima M. Voluntary exercise is systematic review and meta-analysis on the effects of individual
motivated by ghrelin, possibly related to the central reward circuit. J housing on body weight, food intake and visceral fat mass in
Endocrinol 244: 123–132, 2020. doi:10.1530/JOE-19-0213. rodents. Obes Rev 19: 614–637, 2018. doi:10.1111/obr.12663.

AJP-Endocrinol Metab  doi:10.1152/ajpendo.00432.2021  www.ajpendo.org E507


Downloaded from journals.physiology.org/journal/ajpendo at Univ of Texas Arlington (129.107.136.108) on May 26, 2022.

You might also like