scispace - formally typeset
Open AccessJournal ArticleDOI

Ultradian feeding in mice not only affects the peripheral clock in the liver, but also the master clock in the brain.

Reads0
Chats0
TLDR
The additional impact of hypocaloric conditions on the SCN are mainly due to the metabolic and not the timing effects of restricted daytime feeding.
Abstract
Restricted feeding during the resting period causes pronounced shifts in a number of peripheral clocks, but not the central clock in the suprachiasmatic nucleus (SCN). By contrast, daily caloric restriction impacts also the light-entrained SCN clock, as indicated by shifted oscillations of clock (PER1) and clock-controlled (vasopressin) proteins. To determine if these SCN changes are due to the metabolic or timing cues of the restricted feeding, mice were challenged with an ultradian 6-meals schedule (1 food access every 4 h) to abolish the daily periodicity of feeding. Mice fed with ultradian feeding that lost 10% body mass (i.e. hypocaloric) became more diurnal, hypothermic in late night, and displayed larger (3.5 h) advance of body temperature rhythm, more reduced PER1 expression in the SCN, and further modified gene expression in the liver (e.g. larger phase-advance of Per2 and up-regulated levels of Pgc-1α). While glucose rhythmicity was lost under ultradian feeding, the phase of daily rhythms in liver glycogen and plasma corticosterone (albeit increased in amplitude) remained unchanged. In conclusion, the additional impact of hypocaloric conditions on the SCN are mainly due to the metabolic and not the timing effects of restricted daytime feeding.

read more

Content maybe subject to copyright    Report

HAL Id: hal-03366396
https://hal.archives-ouvertes.fr/hal-03366396
Submitted on 19 Oct 2021
HAL is a multi-disciplinary open access
archive for the deposit and dissemination of sci-
entic research documents, whether they are pub-
lished or not. The documents may come from
teaching and research institutions in France or
abroad, or from public or private research centers.
L’archive ouverte pluridisciplinaire HAL, est
destinée au dépôt et à la diusion de documents
scientiques de niveau recherche, publiés ou non,
émanant des établissements d’enseignement et de
recherche français ou étrangers, des laboratoires
publics ou privés.
Ultradian feeding in mice not only aects the peripheral
clock in the liver, but also the master clock in the brain
Satish Sen, Hélène Raingard, Stéphanie Dumont, Andries Kalsbeek, Patrick
Vuillez, Etienne Challet
To cite this version:
Satish Sen, Hélène Raingard, Stéphanie Dumont, Andries Kalsbeek, Patrick Vuillez, et al.. Ul-
tradian feeding in mice not only aects the peripheral clock in the liver, but also the mas-
ter clock in the brain. Chronobiology International, Taylor & Francis, 2017, 34 (1), pp.17-36.
�10.1080/07420528.2016.1231689�. �hal-03366396�

Ultradian feeding in mice not only affects the peripheral clock in the liver, but
also the master clock in the brain
Satish Sen
a,b,c
, Hélène Raingard
a
, Stéphanie Dumont
a
, Andries Kalsbeek
b,c,d
, Patrick Vuillez
a,c
, and
Etienne Challet
a,d
a
5 Regulation of Circadian Clocks team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche
Scientifique (CNRS), University of Strasbourg, France
Q1 ;
b
Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN),
Amsterdam, The Netherlands;
c
International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and
The Netherlands
Q2 ;
d
Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, The
Netherlands.
Q3
10 ABSTRACT
Q4 Restricted feeding during the resting period causes pronounced shifts in a number of
peripheral clocks, but not the central clock in the suprachiasmatic nucleus (SCN). By contrast,
daily caloric restriction impacts also the light-entrained SCN clock, as i ndicated by shifted
oscillations of clock (PER1) and clock-con trolled (vasopressin) proteins. To determine if these
15 SCN changes are due to the metabolic or timing cues of the restricted feeding, mice were
challenged with an ultradian 6-meals schedule (1 food access every 4 h) to abolish the daily
periodicity of feeding. Mice fed with ultradian feeding that lost <10% body mass (i.e.
isocaloric) displayed 1.5-h phase-advance of body temperature rhythm, but remained mostly
nocturnal, together with up-regulated vasopressin and down-regulated PER1 and PER2 levels
20 in the SCN. Hepatic expression of clock genes (P er2, Rev-erb α,andClock)andFgf21 was,
respectively, phase-advanced and up-regulated by ultradian feeding. Mice fed with ultradian
feeding that lost >10% body mass (i.e. hypocalori c) became more diurnal, hypothermic in
late night, and displayed larger (3.5 h) advance of body temperature rhythm, m ore reduced
PER1 expression in the SCN, and further modified gene expression in the liver (e.g. larger
25 phase-advance of Per2 and up-regulated levels of Pgc-1α). While glucose rhythmicity was lost
under ultrad ian fe eding, the p hase of daily rhythms in li ver glycogen and plasma c orticos-
terone (albeit increased in amplitude) remained unchanged. In conclusion, the additional
impact of hypocaloric conditions on the SCN are mainly due to the metabolic and n ot the
timing effects o f restricted daytime feeding.
KEYWORDS
30 circadian rhythm; feeding;
6-meal schedule; clock gene;
suprachiasmatic nucleus
Introduction
Biological rhythms are under the control of circadian
os
cillators, including a master circadian clock
located in the suprachiasmatic nucleus (SCN) of
35 the anterior hypothalamus and peripheral oscillators
present in almost every cell of the body (Bray &
Young, 2009). The underlying molecular mechanism
of the clock is based on transcriptional and
translational feedback loops consisting of positive
40 and negative elements (Reppert & Weaver, 2001 ).
When heterodimerized, the positive limb elements
BMAL1 and CLOCK activate transcription of the
negative elements (Period (Per)1, 2, 3 and
Cryptochrome (Cry)1, 2) that in turn inhibit
45 BMAL1/CLOCK transactivation. In parallel, other
clock genes such as Rev-erbα, β and Ror α, β,whose
transcription is also activated by BMAL1/CLOCK,
modulate Bmal1 and Clock transcription (Cho et al.,
2012; Crumbley & Burris, 2011; Preitner et al., 2002).
50Light perceived by the retina is the most potent
synchronizer of the circadian rhythm produced by
the molecular clock mechanism within the SCN. The
molecular clockwork regulates the rhythmic
transcription of clock-controlled genes, such as the
55gene coding for neuropeptide Arginine Vasopressin
(Avp) (Jin et al., 1999 ). The output of the SCN con-
trols the timing of peripheral clocks via nervous,
hormonal and behavioral cues (Froy, 2011). Food
access restricted to the usual resting period can
60phase-shift circadian oscillations in a number of
peripheral organs and brain regions outside the
SCN, while the SCN master clock remains
CONTACT Etienne Challet challet@inci-cnrs.unistra.fr INCI, CNRS UPR3212, 5 rue Blaise Pascal, 67084 Strasbourg, France. Tel: +33 388456693.Q5
CHRONOBIOLOGY INTERNATIONAL
2016, VOL. 00, NO. 00, 120
http://dx.doi.org/10.1080/07420528.2016.1231689
© 2016 Taylor & Francis

synchronized to the light-dark cycle (Damiola et al.,
2000;Feilletetal.,2008;Stokkanetal.,2001).
65 However, when daytime restricted feeding is com-
bined with caloric restriction, the master clock is
affected, as assessed by phase-advances in daily
rhythms of body temperature, activity rhythm, and
pineal melatonin, as well as by altered photic reset-
70 ting (Challet, 2010). Moreover, daily caloric restric-
tion leads to phase-shifts in daily oscillations of clock
(PER1) and clock-controlled (AVP) proteins in the
SCN (Mendoza et al., 2007). To avoid the synchro-
nizing effects of daily restricted feeding, a protocol
75 has been developed using a feeding regimen of six
10-min food accesses equally distributed over 24 h
(i.e. one 10-min meal every 4 h) (Kalsbeek &
Strubbe, 1998). In nocturnal rats under light-dark
conditions, this ultradian 6-meals-a-day feeding
80 schedule does not modify the phase of locomotor
activity rhythm, but if food access to the 6-meals is
shortened to cause body mass loss, rats become
partially active during daytime due to a phase-
advance of the rest/activity rhythm (Mendoza et al.,
85 2008).
One recent rat study showed that peripheral
clock gene rhythms are still present during ultra-
dian 6-meals-a-day feeding, despite changes in
amplitude and phase (Su et al., 2016b). Another
90 study suggested that in mice, the peripheral clocks
remain unaffected by meal timing when each meal
is given equally spaced either 2, 3, 4 or 6 times per
day. However, if meal frequency is unevenly dis-
tributed, i.e. with unequal intervals between the
95 meals, then the phase of peripheral clock genes
changes, especially in the kidney. Moreover, that
study also showed that ultradian 6-meal feeding
coupled to caloric restriction was able to produce
phase-advances of peripheral clocks inversely pro-
100 portional to the degree of energy intake (Kuroda
et al., 2012).
In the present study, we aimed at investigating
further whether it is the daily timing of feeding
and fasting or metabolic cues associated with calo-
105 ric restriction that affects the central and periph-
eral clocks. For that purpose, we challenged mice
with a 6-meals-a-day feeding schedule (combined
with isocaloric or hypocaloric conditions) and stu-
died their behavioural and physiological changes,
110 as well as expression of clock and clock-controlled
genes in the master clock and liver.
Materials and methods
Animals and housing
Seventy-six5-weekoldmaleC57BL/6Jmice
115(Janvier labs, Le Genest-Saint-Isle, France) were
usedforthisstudy.Theanimalswerehousedin
individual cages equipped with a wheel, at an
ambient temperature of 23 ± 2°C under 12:12 h
light-dark conditions (lights on at 7:00 AM
120(defining Zeitgeber Time (ZT) 0) and off at
19:00 PM (=ZT12)). In a group of 46 animals,
access to food was automatically controlled by
electronic timers for six cages at a time. Thirty
animals served as controls and had ad libitum
125access to food.
All experiments were performed in accordance
with the U.S. National Institutes of Health Guide
for the Care and Use of Laboratory Animals
(1996)
Q6, the French National Law (implementing
130the European Communities Council Directive 86/
609/EEC) and approved in advance by the
Regional Ethical Committee of Strasbourg for
Animal Experimentation (AL/50/57/02/13) and in
compliance with the ethical standards of the jour-
135nal (Portaluppi et al., 2010).
Surgery
Mice were anesthetized with isoflurane (Vetflurane,
Virbac 3% powered by 0.2 l/min O2) to implant a
transponder (Minimitter, Vitalview, Sunriver, OR,
140USA) in the abdominal cavity to record body
temperature and general cage activity. The abdo-
men was shaved and sprayed with antiseptic
(DermaSpray, Bayer) before an incision in the
skin and muscle (810 mm) was made. Once the
145transponder was inserted into the abdominal cavity,
the muscle layer was stitched with surgical sutures
(Filapeau, 3.0) and anti-inflammatory medication
was provided in drinking water (Metacam, 0.2
mg/ml, 0.1 ml) for 2 days.
150Experimental procedure
After surgery, the animals were placed in experimen-
tal cages for 2 weeks with drinking water and food ad
libitum. After this, mice were habituated to an ultra-
dian schedule of six meals each day with one food
155access every 4 h (ZT2, ZT6, ZT10, ZT14, ZT18 and
2 S. SEN ET AL.

ZT22). Access to food during restricted feeding was
set automatically by the Food Planning system based
on a food basket allowing and preventing food access
in the low and upper position, respectively
160 (Intellibio, Seichamps, France). Lifting and fall of
the food basket being associated with a brief motor
noise, these auditory cues may have signaled food
availability to the mice. The smaller mesh size of the
trough compared to the size of the food pellets pre-
165 vented any food hoarding in the cage. The duration
of food access was reduced every 4 days gradually
from6×1h,via6×30min,6×20minto6×15
min. This protocol was based on previous studies in
rats (Kalsbeek & Strubbe, 1998 ;Mendozaetal.,
170 2008). The fact th at mic e were fed every day at the
same times has pr obably improved their ability to
adjust to ultradian feeding, as opposed to irregular
meal times (Valle, 1981 ). Food int ake during day-
time and nighttime was measured twice (at the steps
175 of 6 × 1 h and 6 × 15 min) to evaluate the day-night
pattern of food intake. The body mass was measured
weekly. At the end of two weeks of feeding according
to the 6 × 15 min protocol, two groups were
categorized according to individual adaptation to
180 the paradigm, eventually leading to body mass loss.
A cut-off at 10% body mass loss allowed to distin-
guish an isocaloric group including animals with less
than 10% of body mass loss (mean: 5.4 ± 0.5%; n total
=24;n = 4 per ZT) and a hypocaloric group in which
185 animals lost 10% or more (up to 25%) body mass
(mean: 15.5 ± 1.1%; n total = 22; n =34perZT).
Animals of the control group were kept with food
and wa ter ad libitum (n =5perZT).
Immunohistochemistry
190 At the end of the experiment, animals were sacrificed
with an overdose of pentobarbital. Mice fed with
ultradian 6-meals schedule were sampled every 4 h
betweenfoodaccesses(i.e.ZT0,ZT4,ZT8,ZT12,
ZT16 and ZT20) to limit direct effects of feeding
195 while avoiding prolonged fasting. Control mice fed
ad libitum were sacrificed at the same times. Blood
was sampled by intracardiac puncture, liver was
sampled in the right lobe, and the heart was perfused
with 50 mL of 0.9% saline followed by 50 mL of 4%
200 paraformaldehyde in phosphate buffer (0.1 M, pH
7.4). Brains were removed, postfixed overnight in
4% paraformaldehyde (4°C) and transferred to a
cryoprotectant buffered sucrose solution (30% at
C) for at least 24 h till brains sank to the bottom due to
205the sucrose density gradient. Brains were then frozen
in isopentane around 50°C and stored at 80°C . Five
series of 30-μm coronal SCN sections were prepared
on a cryostat and collected in Phosphate-Buffered
Saline (0.1 M PBS, 1×) and washed with Tris
210Buffer Saline pH 7.6 (0.1 M TBS 1×). Then, sections
were incubated in 3% H
2
O
2
in TBS (30 min) to
suppress endogenous peroxidase activity, thereby
reducing background staining. Again brain sections
were rinsed in TBS 1×. Brain sections were then
215transferred in a solution containing 10% normal
serum (either goat or horse according to the host
species of the primary antibody) and Triton X-100
(0.1 %) in TBS for 2 h, followed by incubation in the
primary antibody (48 h at 4°C). We used rabbit poly-
220clonal anti arginine-vasopressin (AVP) (1:20000,
Truus, a gift from Dr. Ruud Buijs, Netherlands
Institute for Brain Research, Amsterdam, the
Netherlands), goat polyclonal anti-PER1 (1:750; SC-
7724, Santa Cruz Biotechnologies, Santa Cruz, CA,
225USA) and rabbit polyclonal anti-PER2 (1:3000, #PER-
21A; Alpha Diagnostic International, San Antonio
TX, USA; note that for anti-PER2 immunohisto-
chemistry, PBS indicated below was always replaced
with TBS). The sections were washed in PBS 1×, then
230incubated (2 h at 4°C) with biotinylated goat anti-
rabbit IgG (1:500, PK6101; Vectastain Standard Elite
ABC Kit Vector Laboratories, Inc., Burlingame, CA,
USA) for AVP and PER2 and with biotinylated anti-
goat IgG made in horse (1:500, BA-9500; Vector labs)
235for PER1 immunostaining. After this, sections were
rinsed in PBS and incubated (2 h) in a solution
containing avidinbiotin peroxidase complex
(Vectastain Elite ABC kit; Vector Laboratories Inc.).
Following incubation with ABC reagents, sections
240were rinsed 4 times in PBS, and incubated with
H
2
O
2
(0.015%, Sigma-Aldrich, St Louis, MO, USA)
and 3,3-diaminobenzidine tetrahydrochloride (0.5
mg/ml, Sigma-Aldrich) diluted in water. Thereafter,
sections were rinsed with PBS, wet mounted on slides
245coated with gelatin, dehydrated through a series of
alcohols, soaked in xylene, and cover slipped.
Photomicrographs were taken on Leica DMRB
microscope (Leica Microsystems) with an Olympus
DP50 digital camera (Olympus France). The number
250of immunopositive cells was counted on one section
in both SCNs and averaged.
CHRONOBIOLOGY INTERNATIONAL 3

mRNA extraction and quantitative real-time PCR
RNA was extracted from frozen liver samples by
homogenizing liver samples in lysis buffer supplemen-
255 ted with β-mercaptoethanol and using absolutely
RNA miniprep kit (Agilent Technologies, USA. The
sampleswerepurifiedbyprecipitationwithsodium
acetate and isopropyl alcohol. The quality of RNA was
measured on NanoDrop ND-100 spectrophotometer
260 (NanoDrop Technologies, Wilmington, DE, USA;
A260/A280, and A260/A230 values were > 1.8) RNA
integrity was assessed using (Agilent RNA 6000 Nano
Kit) on Aligent 2100 bio-analyzer for all the liver
samples (RIN Value were >7) bio-analyzer. cDNA
265 was synthesized with the High Capacity RNA to
cDNA kit (Applied Biosystem, Foster city CA, USA)
using 1 µg of RNA. Measurement of relative
abundance was performed by real-time PCR analysis
using of TaqMan Gene Expression Master Mix
270 (Life Technologies, Foster city, CA, USA). The
following TaqMan probes (Per2: Mm00478113_m1,
Clock: Mm00455950_m1, Sirt1: Mm00490758_m1,
Fgf21: Mm00840165_g1, Nr1d1 (Rev-erb α):
Mm00520708_m1, Pparα: Mm01208835 m1 and
275 Pgc-1α: Mm00440939_m1) were used for all the
genes with 1µl of cDNA in the reaction mixture of
20 μl. Each reaction PCR was done in duplicate. A
dilution curve was prepared of pooled cDNA samples
using log10 standards to calculate the amplification
280 efficiency for each primer set (values were between
1.851.99). Data were normalized to Tbp
(Mm00446971_m1) and analysed the comparative
cycle threshold (Ct) method RQ = 2-ΔΔCt. ΔΔCt =
ΔCt sample ΔCt reference (Pfaffl, 2001) with effi-
285 ciency corrections. Transcript levels were calculated
relative to the mean of ZT 0 samples.
Plasma metabolic parameters
Plasma samples were obtained after centrifuga-
tion of fresh blood collected with 4% EDTA (10
290 µL for 1 mL of blood) and centrifuged for 10
min (5000 rpm at 4 °C). Plasma gl ucose was
evaluated with GOD-PAP kit (Biola bo, Maizy,
France). The ACS-ACOD method (NEFA-HR2;
Wako, Osaka, Japan) was used for assaying
295 plasma non-esterified fatty acids (NEFA).
Plasma concentrations of corticosterone were
determined by a Rat/Mouse Corticosterone EIA
kit (AC-14F 1, IDS EURL, Paris, FRA NCE). The
limit of s ensitivity of the assay was 0.55 ng/mL.
300Hepatic glycogen assay
Samples of fresh liver were flash-frozen in liquid
nitrogen. Hepatic glycogen was quantified accord-
ing to the method developed by Murat and Serfaty
(Murat & Serfaty, 1974).
305Statistical analysis
Data are presented as mean ± standard error of the
mean (SEM). Statistical analysis was performed by
SigmaPlot (version 12, SPSS Inc, Chicago, IL,
USA). Significance was defined at p < 0.05.
310Two-way analysis of variance (ANOVA) with or
without repeated measures (RM) were performed
to assess the effect of Feeding conditions (food ad
libitum, ultradian iso- or ultradian hypo-caloric
feeding) and the effect of Time (either baseline
315versus experimental condition, or time of day),
and the Interaction between these factors. One-
way analysis of variance (ANOVA) was performed
to assess the effect of Time in the separate feeding
groups. When appropriate, post-hoc analysis was
320performed using Fisher LSD method. For assessing
daily rhythmicity, we used a cosinor analysis to
determine mean level, amplitude and acrophase of
the considered parameter with SigmaPlot software
(Jandel Scientific,Chicago, IL). Data were fitted to
325the following regression: [y = A + B·cos(2π(x C)/
24)], where A is the mean level, B the amplitude
and C the acrophase of the rhythm.
Results
Food intake
330When mice were fed with 6 meals of 1 h each, they ate
most of their food during their active period (night
time) and less during their resting period (p<0.001).
Post-hoc analysis showed a significant effect of Time
(nightvs.day;FisherLSDtest,p<0.05) (Figure 1A).
335Mice on the 15-min 6-meals-a-day feeding schedule,
both the hypocaloric and isocaloric group, ate equal
amounts of food during day and night. However, the
amount of food ingested by the hypocaloric group
was significantly lower (~20%) than that by the
4 S. SEN ET AL.

Citations
More filters
Journal ArticleDOI

The circadian regulation of food intake.

TL;DR: In this Review, Etienne Challet provides insight into the dual modulation of food intake by homeostatic and circadian processes, describes the mechanisms regulating feeding time and highlights the beneficial effects of correctly timed eating, as opposed to the negative metabolic consequences of mistimed eating.
Journal ArticleDOI

Feeding Rhythms and the Circadian Regulation of Metabolism

TL;DR: The aim of this review is to synthesize the work on the mechanisms and metabolic effects of circadian misalignment, highlighting the timing of food intake as a powerful environmental cue with the potential to destroy or restore the synchrony of circadian rhythms in metabolism.
Journal ArticleDOI

Circadian clock network desynchrony promotes weight gain and alters glucose homeostasis in mice.

TL;DR: Data reveal the importance of an overall synchronized circadian clockwork for the maintenance of metabolic homeostasis and restore peripheral clock rhythmicity and synchrony by time-restricted feeding normalizes body weight and glucose metabolism.
Journal ArticleDOI

Circadian Influences of Diet on the Microbiome and Immunity

TL;DR: Current knowledge linking the circadian clock and dietary timing to host immune-microbiota interactions is reviewed, exemplifying how this axis may impact on host immunity in health and in a variety of immune-mediated diseases.
Journal ArticleDOI

Diurnal regulation of RNA polymerase III transcription is under the control of both the feeding-fasting response and the circadian clock

TL;DR: Gen expression arrays are used to measure mRNA accumulation during the diurnal cycle in the livers of wild-type mice, mice fed at regular intervals during both night and day, and mice lacking the Maf1 gene to provide a comprehensive view of the changes in cyclic mRNA accumulation occurring in these different systems.
References
More filters
Journal ArticleDOI

A new mathematical model for relative quantification in real-time RT-PCR.

TL;DR: This study enters into the particular topics of the relative quantification in real-time RT-PCR of a target gene transcript in comparison to a reference gene transcript and presents a new mathematical model that needs no calibration curve.
Journal ArticleDOI

Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1.

TL;DR: It is shown that the Sir2 homologue, SIRT1 controls the gluconeogenic/glycolytic pathways in liver in response to fasting signals through the transcriptional coactivator PGC-1α, and this findings have strong implications for the basic pathways of energy homeostasis, diabetes and lifespan.
Journal ArticleDOI

Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus

TL;DR: It is shown that temporal feeding restriction under light-dark or dark-dark conditions can change the phase of circadian gene expression in peripheral cell types by up to 12 h while leaving thephase of cyclic gene expressionIn the SCN unaffected.
Journal ArticleDOI

The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator.

TL;DR: The orphan nuclear receptor REV-ERBalpha is identified as the major regulator of cyclic Bmal1 transcription, and constitutes a molecular link through which components of the negative limb drive antiphasic expression of component of the positive limb.
Journal ArticleDOI

Entrainment of the Circadian Clock in the Liver by Feeding

TL;DR: It is demonstrated that feeding cycles can entrain the liver independently of the SCN and the light cycle, and the need to reexamine the mammalian circadian hierarchy is suggested, raising the possibility that peripheral circadian oscillators like those in the liver may be coupled to theSCN primarily through rhythmic behavior, such as feeding.
Related Papers (5)
Frequently Asked Questions (21)
Q1. What are the contributions in "Ultradian feeding in mice not only affects the peripheral clock in the liver, but also the master clock in the brain" ?

Challet et al. this paper showed that the effects of daily restricted feeding on the master clock of a rat rat can be traced back to the number of 10-min food accesses over 24 hours ( i.e., one 10min meal every 4 h ). 

Daytime restricted feeding in rats, however, results not only in increased gluconeogenesis, but also in a 12-h shift in the daily variations of hepatic glycogen content 735(Perez-Mendoza et al., 2014), thus indicating that daily restricted feeding can impact timing of glycogen synthesis. 

In mice fed according to the ultradian feeding schedule, the daily rhythm of plasma corticosterone is increased in amplitude and not shifted, while AVP protein expression is markedly increased. 

The 985 weaker oscillations of AVP during ultradian feeding may thus feedback within the SCN to the molecular clockwork and participate in dampenedSCN oscillations, as observed here for a reduced amplitude of PER1 and PER2 expression. 

SIRT1 also regulates Pparα expression (Purushotham et al., 2009) and interacts with Pgc-1α in the liver 870 (Rodgers et al., 2005). 

The lack of rhythmicity in the isocaloric 460 6-meals schedule groups indicates that the ultradian feeding schedule may have a major impact on SCN functioning. 

Because FGF21 can be released in the bloodstream and act on the central clock (Bookout et al., 2013), the authors assayed plasma FGF21 in mice challenged with the ultradian 6-meal schedule. 

At the end of two weeks of feeding according to the 6 × 15 min protocol, two groups were categorized according to individual adaptation to 180 the paradigm, eventually leading to body mass loss. 

When diurnal grass rats (Arvicanthis ansorgei) are fed with daily hypocaloric feeding at night, they become partially nocturnal (Mendoza et al., 2012). 

In the present study performed in mice, the daily rhythm of plasma corticos-750 terone also showed a profound increase in amplitude during ultradian feeding conditions, but no shift of its acrophase. 

In nocturnal rats under light-dark conditions, this ultradian 6-meals-a-day feeding 80 schedule does not modify the phase of locomotor activity rhythm, but if food access to the 6-meals is shortened to cause body mass loss, rats become partially active during daytime due to a phaseadvance of the rest/activity rhythm (Mendoza et al., 85 2008). 

The fact that the rest-activity rhythm was only phase-advanced in the hypocaloricgroup may be due to the deeper hypothermia in the calorie-restricted mice, i.e. the SCN and/or its downstream structures (e.g. secondary brain clocks) 705would be sensitive to the shifting effects of this deeper hypothermia. 

The authors used rabbit poly220clonal anti arginine-vasopressin (AVP) (1:20000, Truus, a gift from Dr. Ruud Buijs, Netherlands Institute for Brain Research, Amsterdam, the Netherlands), goat polyclonal anti-PER1 (1:750; SC7724, Santa Cruz Biotechnologies, Santa Cruz, CA, 225USA) and rabbit polyclonal anti-PER2 (1:3000, #PER21A; Alpha Diagnostic International, San Antonio TX, USA; note that for anti-PER2 immunohistochemistry, PBS indicated below was always replaced with TBS). 

The quality of RNAwas measured on NanoDrop ND-100 spectrophotometer 260 (NanoDrop Technologies, Wilmington, DE, USA; A260/A280, and A260/A230 values were > 1.8) RNA integrity was assessed using (Agilent RNA 6000 Nano Kit) on Aligent 2100 bio-analyzer for all the liver samples (RIN Value were >7) bio-analyzer. 

Previous studies have already shown that day975 time restricted feeding in rats modifies the dailypattern of AVP release from the SCN (i.e. delayed onset and earlier offset; Kalsbeek et al., 1998). 

This modulatory effect could partly explain the dampened amplitude of hepatic expression of Per2 during 6-meal schedule,though it did not abolish the daily rhythmicity (as would be expected if oxyntomodulin was the sole845 factor involved because food intake would trigger Per2 transcription every 4 h, leading to constitutive levels throughout 24 h). 

The molecular clockwork regulates the rhythmic transcription of clock-controlled genes, such as the 55gene coding for neuropeptide Arginine Vasopressin (Avp) (Jin et al., 1999). 

A dilution curve was prepared of pooled cDNA samples using log10 standards to calculate the amplification 280 efficiency for each primer set (values were between 1.85–1.99). 

For AVP and PER2, the reducing effects of ultradian feeding on the amplitude of their oscillations werecomparable between the isocaloric and hypocaloric 940 groups, while the magnitude of the downregula-tion of PER1 was more pronounced in the hypocaloric group. 

The fact that mice were fed every day at the same times has probably improved their ability to adjust to ultradian feeding, as opposed to irregular meal times (Valle, 1981). 

To avoid the synchronizing effects of daily restricted feeding, a protocol 75 has been developed using a feeding regimen of six 10-min food accesses equally distributed over 24 h (i.e. one 10-min meal every 4 h) (Kalsbeek & Strubbe, 1998).