You are on page 1of 10

Molecular and Cellular Biochemistry (2018) 449:63–72

https://doi.org/10.1007/s11010-018-3343-7

Low-grade neuroinflammation due to chronic sleep deprivation


results in anxiety and learning and memory impairments
Shaffi Manchanda1 · Harpal Singh1 · Taranjeet Kaur1 · Gurcharan Kaur1

Received: 28 December 2017 / Accepted: 7 March 2018 / Published online: 16 March 2018
© Springer Science+Business Media, LLC, part of Springer Nature 2018

Abstract
Chronic sleep loss/fragmentation prevalent in the current 24/7 society is associated with irreversible consequences on health
and overall wellbeing. Various studies have well documented the ill effects of acute sleep loss on cognitive functions of
individuals; however, the underlying mechanism behind the chronic sleep loss is yet to be explored. The present study was
aimed to investigate whether chronic sleep deprivation (CSD) triggers anxiety-like behaviour and memory decline in male
Wistar rats. Rats were sleep deprived by placing them over slowly rotating drum (2 rpm) for 18 h (between 4 pm and 10 am)
followed by 6 h of recovery sleep for 21 consecutive days. Post CSD regimen, rats were subjected to behavioural tests such
as elevated plus maze (EPM), Novel Object Recognition (NOR) and Rotarod performance test and then sacrificed to remove
brain for further molecular studies. The study demonstrated that CSD rats showed anxiogenic behaviour along with recogni-
tion memory decline compared to control rats. CSD rats further showed elevated levels of inflammatory cytokines (TNFα,
IL-1β) along with activation of NFκB and AP1 transcription factors in hippocampus and piriform cortex (PC) regions of
brain. These observations were also accompanied by enhanced expression of GFAP and Iba1 in the two brain regions. The
data suggest that CSD triggered low-grade neuroinflammation which caused anxiogenic response and recognition memory
impairment. The study provides preliminary leads to further explore the role of astrocytes/microglial cells and inflammatory
cytokines in mediating these neurobehavioural consequences of chronic sleep loss and to develop effective interventions to
combat them.

Keywords  Chronic sleep deprivation · Neuroinflammation · Inflammatory mediators · Astrogliosis · Microgliosis

Introduction chronic periods of total sleep loss and sleep fragmentation


are associated with immune system dysfunction (cellular
Chronic sleep loss, defined as sleep obtained ≤ 6-h/24-h and humoral) [2], but the mechanism behind this is not well
period, is prevalent in the modern societal setup due to pro- understood under different sleep loss conditions. In addition
fessional commitments and lifestyle factors. Serious conse- to neurons, non-neuronal cells such as oligodendrocytes,
quences of sleep loss documented so far include compro- astrocytes and microglia also play critical role in synaptic
mised neurogenesis, cognitive functioning (poor vigilance maintenance and homeostasis [3–5]. Recent studies provide
and performance, deficits in learning and memory), meta- evidence that both chronic sleep loss and fragmentation can
bolic and cardiovascular problems, immune disturbances activate these cell types and influence the behavioural and
and blood–brain barrier disruption [1]. Both acute and physiological state [6–9]. Astrocytes demonstrate more
pronounced response to wakefulness than oligodendrocytes
with multifold activation in the genes during wake period
Shaffi Manchanda, Harpal Singh, Taranjeet Kaur have contributed than sleep [8]. Impact of short-term sleep loss on learning
equally. and memory impairment has now been firmly established
and demonstrated by various studies [10–14]. Acute sleep
* Gurcharan Kaur
kgurcharan.neuro@yahoo.com loss has also been reported to lead to anxiety-like behaviour
and impairment in learning and memory as reported by our
1
Medical Biotechnology Laboratory, Department lab [9, 13, 14] as well as others [15, 16]. Activation of sym-
of Biotechnology, Guru Nanak Dev University, Amritsar, pathetic nervous system and hypothalamic–pituitary–adrenal
Punjab, India

13
Vol.:(0123456789)

64 Molecular and Cellular Biochemistry (2018) 449:63–72

axis after sleep disturbance [17] lead to the heightened libitum food and water supply. For experimental study, ani-
pro-inflammatory state [18]. Various clinical studies [19, mals were grouped into two groups—(I) control rats allowed
20] have also documented the occurrence of inflammation undisturbed sleep during light phase and (II) Chronic Sleep
caused due to sleep loss. Sleep disturbance has also been Deprived (CSD) rats deprived of sleep for duration of 18 h
reported to induce microglial activation in the mouse hip- between 4 pm and 10 am for 21 days, Forced locomotion
pocampus region along with elevated levels of IL-6 [21]. method was used for sleep deprivation by placing animals
Literature reports on acute sleep deprivation regimen have on slowly rotating drum with dimensions (40 cm diame-
suggested the link between sleep loss and neuroinflammation ter × 30 cm height) operating at a speed of 2 rpm and were
but whether chronic sleep loss causes impairments to same allowed to sleep for 6-h duration between 10 am and 4 pm
extent as acute sleep loss is yet to be explored. for 21 consecutive days. All the animals were then subjected
The present study was aimed to investigate whether to behavioural studies to test their memory and anxiety sta-
chronic sleep deprivation (CSD) of 18 h for 21 consecutive tus after the regimen. All animal experimental protocols
days in male Wistar rats triggers anxiety-like behaviour and were approved by Institutional Animal Ethical Committee,
memory decline accompanied by activation of inflammatory Guru Nanak Dev University, Amritsar, India (permission
mediators with reactive astrogliosis and microgliosis. Rats number 226/CPCSEA-A/2014/37) and performed in accord-
were sleep deprived for 18 h (between 4 pm and 10 am) fol- ance with the guidelines of ‘Animal Care and Use’ laid down
lowed by 6 h of recovery sleep for 21 consecutive days. We by Institutional Animal Ethical Committee, Guru Nanak Dev
have used rotating drum method to induce sleep deprivation University.
in rats. This method induces sleep fragmentation rather than
total sleep loss as animals may have microsleeps between the Behavioural studies
time, rotating drum disc. Post CSD regimen, rats were first
subjected to behavioural tests such as elevated plus maze Elevated plus maze (EPM) test
(EPM) test to assess the anxiety levels, Novel Object Recog-
nition (NOR) test to elucidate learning and memory impair- The animals (n = 8 per group) were subjected to EPM test
ments and Rotarod performance test for motor coordination post-regimen on 22nd day starting from 10:00 am onwards,
and then sacrificed to remove brain for further molecular which consisted of two opposing open (50 × 10 cm) and
studies. To investigate the underlying mechanisms of chronic closed arms (50 × 10 × 50 cm) extending from the central
sleep deprivation-associated neurobehavioural alterations, platform (10 × 10 cm), crossing each other in the form of a
we studied the expression of pro-inflammatory mediators, plus, elevated 50 cm above the floor. At the beginning of the
viz. TNFα (tumour necrosis factor α), IL-1β (Interleukin-1β) test, the individual rats were placed on the central platform
and IL-6 (Interleukin-6) and their downstream transcrip- facing one of the closed arms and were allowed to freely
tional activation of NFκB (nuclear factor-κB) and AP1 explore the apparatus for 5 min. Whole experiment was per-
(activator protein 1). NFκB occupies a vital position in the formed in dim light to encourage rats to explore open arms.
inflammatory cascade, where it controls the production of The behaviour of each rat was monitored using a video cam-
numerous pro-inflammatory mediators [22, 23], whereas era and their movements were registered and analysed. The
AP1 has been implicated in mediating cell survival by whole apparatus was thoroughly cleaned using 5% ethanol
inducing apoptosis under stressful conditions [24]. Occur- before placing next rat. An entry into any arm was defined as
rence of reactive astrogliosis and microgliosis under CSD entry of all four limbs into that arm. A crossing was defined
conditions was ascertained by studying the expression of as animals emerging from one arm and entering with all four
astroglial cell-specific marker GFAP (glial fibrillary acidic paws, to its opposite arm. Head dipping is defined as looking
protein) and microglial cell-specific marker Iba1 (Ionized down from the edge of an open arm or the central platform.
calcium-binding adapter molecule 1) from hippocampus and Videos were analysed by multiple observers blinded to the
piriform cortex (PC) regions of brain. experiment.

Novel object recognition (NOR) test


Materials and methods
Rats (n = 8 per group) were subjected to NOR test for testing
Animals and sleep deprivation regimen their object recognition and working memory. The rats were
habituated to the empty box (100 × 50 × 50 cm) for 5 min
Wistar albino male rats in the age group of 4–8 months, over 2 days at the start of dark phase. The activities of the
weighing 260–275 g were caged in the group of three under rats were recorded using a video camera. This was followed
controlled environmental conditions (constant temperature by the familiarization phase during which rats were placed in
of 25 ± 2 °C and constant dark/light cycle of 12:12 h) with ad the box with two similar sample objects without having any

13
Molecular and Cellular Biochemistry (2018) 449:63–72 65

specific odour and were allowed to explore both the objects centrifuged at 5000 rpm for 10 min at 4 ̊C. Supernatant was
for 5 min up to 3 days. The objects were cleaned thoroughly collected after centrifugation and used for protein estima-
with 70% ethanol to ensure the absence of olfactory cues. tion by Bradford method. 50 µg of protein from each sample
On the test phase, the least preferred sample object was was mixed with 6X sample buffer. Protein samples were
replaced by a novel object to check the recognition memory resolved in 7–10% SDS-PAGE followed by transfer onto a
of the rats. Between each phase, there was a gap of 24 h. The 0.45 µm pore size PVDF membrane (Hybond-P from Amer-
exploration of object was defined by sniffing, licking, chew- sham Biosciences UK Limited) using the semi-dry Novablot
ing by rats or by moving vibrissae while directing the nose system (Amersham Biosciences UK Limited) at 90 mA for
towards and less than 1 cm from the object. The number of 90 min. Transferred membranes were then blocked with
episodes and time spent in exploration of each object by each 5% skimmed milk in 0.1% TBS-Tween 20 for 2 h. Further,
animal was recoded. The preference index between the two membranes were probed with mouse monoclonal anti-Iba1
objects was calculated as (1:1000) (EMD Millipore, Catalogue number MABN92),

Preference index = Time spent in exploring new object∕Total time in exploration.

Grooming and rearing anti-GFAP (1:5000) (Sigma-Aldrich, Catalogue number


G3893), anti-AP1 (1:2000) (Sigma-Aldrich, Catalogue num-
We further analysed grooming behaviour in these animals in ber A5968), anti-TNFα (1:1000) (Sigma-Aldrich, Catalogue
the open field box during NOR test to correlate the effect of number SAB1404480), anti-IL-1β (1:1000) (Sigma-Aldrich,
stress and anxiety caused by extended wakefulness on mem- Catalogue number SRP8033), anti-IL-6 (1:1000) (Sigma-
ory and cognition. The grooming behaviour in rats is defined Aldrich, Catalogue number SAB5300276) and anti- rabbit
as the duration of time spent by the animal in licking various NFκB (1:2000) (Cell Signaling Technology, Catalogue num-
body parts including paw licking, head washing, body fur ber 8242) for overnight at 4 °C. This was followed by three
grooming with hind paws, leg and genital scratching. Any washings with 0.1% TBS-Triton-X 100 of 10 min each and
grooming bout of duration more than 5 s was recorded as incubation with HRP-labelled anti-mouse IgG (1:2500) or
actual grooming bout. Total time spent in grooming activity anti-rabbit IgG (1:2500) secondary antibodies (Genei) for
and latency of grooming i.e. grooming bouts was recorded 2 h at 25 °C. Membranes were again washed 0.1% TBS-
using video recording and averaged for each group. Rearing Triton-X 100 thrice for 10 min each. Immunoreactive bands
is a frequent behavioural act defined as standing up on the were detected by Amersham ECL Plus Western blot detec-
hind limbs with or without keeping the forelimbs on the tion system (GE Healthcare Life Sciences UK Limited)
wall. It is a measure of environmental novelty used to meas- using Image Quant LAS 4000 (GE Healthcare Life Sciences
ure learning and memory. Number of rearing episodes were UK Limited). α-tubulin has been used as an endogenous
recorded for each rat for every 5-min session during NOR. control for normalizing the expression of the proteins probed
on the membrane. Change in expression of protein of interest
Rotarod performance test was taken as the average of integrated density values (IDV)
obtained from at least three independent experiments.
Rotarod apparatus is an automatic motor-driven tread-
mill (Rotamex-5; Columbus Instruments) consisting of a
7.0 × 9.5 cm spindle diameter with a fall height of 44.5 cm Statistical analysis
from the centre. The Rotarod performance test was per-
formed on each animal (n = 8 per group) at a fixed speed of Values were expressed as mean ± SEM of the values
10 rpm for duration of 300 s. The number of falls and the obtained from at least three independent experiments.
time spent on the rotating rod was recorded as a measure of The Sigma Stat for Windows (version 3.5) was used to
running performance for each trial and averaged for each analyse the results by Student’s t test and one-way ANOVA
group. (Holm–Sidak post hoc method), in order to determine
the significance of the mean values. Two-way ANOVA
was performed on EPM test data and NOR test data to
Western blotting
determine the level of significance for the parameters
studied within the group and between the groups. One-
Rats (n = 4 for each group) were anesthetized using thio-
way ANOVA was used to analyse the data of rotarod
pentone injection (1 unit per 10 g) and sacrificed to dissect
performance test and Western blotting. Values with p
out their brain. PC and hippocampus regions were dissected
value ≤ 0.05 were considered as significant.
out and lysed in lysing buffer using tissue homogenizer and

13

66 Molecular and Cellular Biochemistry (2018) 449:63–72

(a) (b)
300 Open Arm 7 Open Arm
Closed Arm #
Closed Arm

No. of entries in open and closed


#
Time spent in open and closed

250 Central Open Area # 6


#
5
200
arm (seconds)

arm
150
3
100 $
$ 2
50 1
*
0 0
Control CSD Control CSD

(c) 5 Open Arm (d)


Closed Arm 7
#
No. of crossings in open and

4 6

No. of head dips 5


closed arm

3
4

2 *# 3

2
1 *
1

0 0
Control CSD Control CSD

(e) (f)
2.5 350
300
2
250
Time spent (sec.)
No. of Falls

1.5 200
150
1
100
0.5
50

0 0
Control CSD Control CSD

Results impairment and elicited inflammatory response and caused


reactive gliosis in hippocampus and PC regions of brain.
The current data suggest that CSD for 21  days in male During EPM test, although both control and CSD animals
Wistar rats induced anxiety-like behaviour and memory spent significant duration of time in closed arm (p ≤ 0.001)

13
Molecular and Cellular Biochemistry (2018) 449:63–72 67

◂Fig. 1  Chronic sleep loss resulted in anxiety-like behaviour and poor of brain, which is indicative of astrogliosis and microgliosis
grip strength: a histogram represents average time spent by the rats in under CSD conditions. Further activation of NFκB (which
open and closed arms of EPM apparatus. CSD animals spent maxi-
mum time in the closed arm than open arm and central open area as
is implicated in sleep regulation) was observed in CSD rats
compared to control rats. The exploratory activity of animals of each in hippocampus (p ≤ 0.05) and PC regions of brain as com-
group is show by the number of entries (b) and number of crossings pared to control rats (Fig. 3a third panel, b, c). This was also
(c) in open and closed arms. CSD rats showed complete avoidance of accompanied by activation of AP1 (dimer of c-Jun and c-fos)
the open arm compared to control rats. d Histogram represents the
number of head dips by the rats of each group. e, f Histograms repre-
in CSD rats in both the brain regions (p ≤ 0.05) (Fig. 3a
senting number of falls and time spent by rats on rotarod. Values are fourth panel, b, c). We further evaluated the inflammatory
expressed as  mean ± SEM, *p ≤ 0.05 control versus CSD, #p ≤ 0.05 molecules expression such as TNFα, IL-1β and IL-6 using
open versus closed arm and, $p ≤ 0.05 closed versus central open area. Western blotting. CSD rats showed enhanced expression of
Holm–Sidak method after one-way and two-way ANOVA. (Color fig-
ure online)
TNFα in both the brain regions (p ≤ 0.05) (Fig. 4a upper
panel, b, c) as well as IL-6 in hippocampus region (p ≤ 0.05)
(Fig. 4a second panel, b). However, a marginal decrease in
but CSD rats completely prohibited themselves from enter- the expression of IL-6 was observed in PC region which was
ing the open arm and spent no time at all in open arm unlike not statistically significant (Fig. 4a second panel, c). CSD
control rats (p ≤ 0.05) (Fig. 1a). These observations are in rats showed no change in the expression of IL-1β in both the
line with the number of entries and crossings in an arm, brain regions studied (Fig. 4a third panel, b, c).
where CSD rats entered closed arm with more number of
entries and crossings than open arm (p ≤ 0.001) as compared
to control rats (Fig. 1b, c). However, both control and CSD Discussion
rats demonstrated no significant difference for the time spent
in central open area (Fig. 1a). Further the number of head Chronic sleep deprivation for 21  days was observed to
dips shown by CSD rats was significantly lesser (p ≤ 0.01) induce anxiety-like behaviour and recognition memory
than the control rats (Fig. 1d). These observations suggest impairment in young adult male Wistar rats. Animals were
that CSD induced anxiety-like behaviour in male rats. We subjected to EPM test to ascertain their anxiety levels. CSD
also evaluated motor activity of these rats and found that group rats showed minimum number of entries and time
CSD rats after 21 days of chronic sleep deprivation had spent in open arm as compared to closed arm, which indi-
33.33% more number of falls (Fig. 1e). There was no differ- cates their higher anxiety levels triggered due to chronic
ence in the time spent by the CSD rats on rotating rod from sleep loss. Complete avoidance for the open arm demon-
that of control rats (Fig. 1f). strated by CSD rats as evident form the number of crossings
We further analysed recognition memory response of rats in open arm further supports their anxiogenic response after
during NOR test and found that CSD rats had lesser pref- CSD. CSD rats also showed fear-like response as evident
erence index score (15% lesser) for the novel object than from the marked reduction in the number of head dips as
control rats (Fig. 2c) and spent 25% lesser time in exploring compared to control rats. Various sleep deprivation para-
the novel object as compared to control rats (Fig. 2b). In digms have shown time-dependent feature of anxiety-like
parallel, time spent by CSD rats in exploring the old object behaviour triggered due to SD as evaluated by EPM [15, 25,
was higher (52.83%) as compared to control rats (Fig. 2b). 26]. A recent study by Yin et al. [27] has shown that mice
However, there was marginal difference between the two exhibited anxiety-like behaviour following 3 days of inter-
groups for the number of episodes towards both the objects mittent and paradoxical SD with more pronounced increase
(Fig. 2a) and the rearing activity of CSD rats was also 75% when deprived for 7 days of SD paradigm. Sleep deprivation
higher as compared to control rats (Fig. 2d). Furthermore, and anxiety are further known to affect the motor perfor-
we analysed grooming behaviour of rats and found that CSD mance of animals. More number of falls from the rotating
rats had more grooming bouts (percentage increase was rod indicated the impaired motor coordination of CSD rats.
62.5%) compared to control rats (Fig. 2e). However, they However, we did not find any difference in the length of time
spent 20% lesser time in grooming themselves as compared spent by CSD rats on rotating rod from that of control rats.
to control rats (Fig. 2f). The current data suggest that in CSD regimen of 18 h of
sleep deprivation, although the animals were daily allowed
CSD and neuroinflammation undisturbed sleep for 6 h, but there was sleep debt due to
curtailment of daily sleep dose, which may have led to anxi-
Chronic sleep deprivation resulted in upregulation in the ety and low-grade inflammatory response.
expression of astrocytic cell-specific protein GFAP (Fig. 3a Since anxiety is further associated with poor performance,
upper panel, b, c) and microglial cell specific marker Iba1 alertness and cognitive dysfunction, therefore we analysed
(Fig. 3a second panel, b, c) in hippocampus and PC regions the recognition memory and learning response of the rats to

13

68 Molecular and Cellular Biochemistry (2018) 449:63–72

(a) (b)
6 Old New 20 # Old New
18
5 16

Time spent (sec.)


14
No. of episodes

4
12
3 10
8
2 6
4
1
2
0 0
Control CSD Control CSD

(c) (d)

0.9 12
0.8
10
0.7
Preference Index

0.6 8
Rearings

0.5
6
0.4
0.3 4
0.2
2
0.1
0 0
Control CSD Control CSD

(e) (f)

6 80
70
5
60
Grooming time (sec.)
Grooming bouts

4
50
3 40
30
2
20
1
10
0 0
Control CSD Control CSD

Fig. 2  Chronic sleep loss caused learning and recognition memory rats. e, f Histograms representing grooming bouts and time spent in
deficits: histograms represent several parameters analysed dur- grooming by the two groups. CSD rats had more number of grooming
ing NOR test a number of episodes, b time spent in exploration of bouts (p = 0.07) with lesser time spent in grooming (p = 0.45) as com-
objects, c preference index for the novel object, d number of rearings. pared to control rats. Values are expressed as mean ± SEM, #p ≤ 0.05
CSD rats spent relatively lesser time in exploring the novel object old versus new. Holm–Sidak method after one-way and two-way
with lower preference score for the novel object compared to control ANOVA. (Color figure online)

13
Molecular and Cellular Biochemistry (2018) 449:63–72 69

(a)
Hippocampus Piriform Cortex
Control CSD Control CSD
GFAP 50 kDa

Iba1 17 kDa

NFKB 65 kDa

AP1 39 kDa

α-tubulin 52 kDa

(b) (c)
180 * Control 160 Control
CSD
160 140 CSD
140 * Relative expression in PC region *
120
in hippocampus region

*
Relave expression

120
100
*
100
80
80
60
60
40
40
20 20

0 0
GFAP Iba1 NFκB AP-1 GFAP Iba1 NFκB AP-1

Fig. 3  Chronic sleep loss activated astrocytes and microglial cells of all the markers were upregulated in response to chronic sleep loss
and inflammatory pathway: a representative western blot images of in CSD rats. Values are expressed as mean ± SEM, *p ≤ 0.05 control
GFAP, Iba1, NFκB and AP1. b, c Quantitative densitometric analysis versus CSD, Holm–Sidak method after one-way ANOVA. (Color fig-
of the markers in the hippocampus and piriform cortex regions of rat ure online)
brain among the two groups (n = 4 for each group). The expression

a novel environment. Reduced preference of CSD rats for and non-stressful conditions [28–31]. Rats perform dif-
the novel object as evident from the preference index score ferent types of grooming behaviour depending on envi-
(Fig. 2c) and lesser time spent by CSD rats in exploration of ronmental conditions [32]. Under non-stressful conditions
novel object as compared to control rats (Fig. 2b) indicated such as home cage, rats show spontaneous self-grooming
their impaired recognition memory and learning response pattern characterized by a typical cephalocaudal sequence
to the novel environment. Recent study has revealed that [28, 29]. Under anxiogenic conditions such as the open-
paradoxical SD for 3 consecutive days did not cause work- field test (OFT) or the EPM, rats show interrupted and dis-
ing memory impairment as shown on Y-maze task, whereas organized grooming pattern that depends on their anxiety
7 days of paradoxical SD significantly impaired working levels [30, 31, 33]. In the present study, we have analysed
memory indicating that SD-induced impairment is degree self-grooming behaviour of rats under non-stressful condi-
dependent [27]. These findings may suggest that chronically tions of environmental novelity (NOR). Thus, reduced time
reducing daily quota of sleep also impairs the learning and spent by CSD rats in self-grooming supported their anxi-
memory function along with anxiety. ogenic behaviour triggered due to sleep debt and chronic
Self-grooming is an innate self-care behaviour for the sleep fragmentation, whereas control rats showed normal
cleaning of skin and fur, adopted under various stressful grooming behaviour attained as a result of adequate sleep.

13

70 Molecular and Cellular Biochemistry (2018) 449:63–72

(a) Hippocampus Piriform Cortex mice to water-based environment may be due to long-term
Control CSD Control CSD utilization of flower pot method of sleep deprivation.
TNF-α 17 kDa
Chronic sleep deprivation caused
IL-1β 17 kDa neuroinflammation by activating inflammatory
mediators, astrocytes and microglial cells
IL-6 21 kDa

α-tubulin
Sleep deprivation, sleep curtailment and sleep fragmentation
52 kDa
are known to cause low-grade inflammation and blood–brain
(b) barrier disruption mediated by inflammatory molecules,
140
*
astrocytes and microglia. For instance, chronic sleep depri-
Control CSD
120 * vation for period of 72 h elevated the levels of TNFα, IL-6
and IL-1β in hippocampus and basal forebrain of rats [34].
Relative expression in
hippocampus region

100 Similarly, in the present study, chronic sleep deprivation also


80 resulted in activation of pro-inflammatory cytokines in rats
as evident from the marked increase in the expression of
60
TNFα in both the brain regions and IL-6 in hippocampus
40 region (Fig. 4a–c). A recent study has shown that intermit-
20 tent or paradoxical sleep for 3 and 7 days repetitively for
3 months caused sustained increase in expression of TNFα
0
TNFα IL-1β IL-6 without any significant change in IL-1β and IL-6 compared
to control mice [27]. Further, we evaluated the expression
(c) of NFκB and AP1 which are known to be involved in sleep
140
regulation [35, 36]. NFκB activation converges in cellular
Control CSD
* pathways considered as hallmark of inflammation [37]. Acti-
Relative expression in PC region

120 vation of NFκB and its subsequent translocation to nucleus


100 may promote the transcriptional activation of inflammation-
related genes such as TNFα, IL-1β [1, 17]. Although NFκB
80
showed marginal increase in its expression in the two brain
60 regions but consistent change may suggest the role of this
40 transcription factor in mediating the inflammatory response
to CSD. Several studies based on SD have shown induction
20
of AP1 and c-fos (immediate early gene) in young adult rats
0 due to extended wakefulness [36, 38, 39]. Enhanced expres-
TNFα IL-1β IL-6
sion of AP1 in the current study may be the outcome of
increased sleep debt in CSD rats as long-term lack of sleep
Fig. 4  Chronic sleep loss ameliorated the base levels of inflamma- disrupts the homeostatic balance of sleep–wake cycle lead-
tory cytokines: a representative western blot images of inflamma-
tory cytokines TNFα, IL-1β and IL-6. b, c Quantitative densitomet- ing to cognitive impairments.
ric analysis of the markers in the hippocampus and piriform cortex Astrocyte/microglial activation along with release of
regions of rat brain among the two groups (n = 4 for each group). inflammatory cytokines is considered to be the main source
Values are expressed as mean ± SEM, *p ≤ 0.05 control versus CSD, of neuroinflammation among other factors [40]. Various ani-
Holm–Sidak method after one-way ANOVA. (Color figure online)
mal studies from our lab and others have shown that both
acute and chronic periods of sleep loss induce astroglial and
microglial activation [12, 14, 27]. In a recent study by Hur-
Further, the rearing activity of CSD rats for the open area tado-Alvarado et al. [41], it was reported that 20 h of sleep
was higher compared to control rats which could be due deprivation for 10 consecutive days induced overexpression
to their increased familiarity to the open area which was of markers of reactive astroglia (GFAP) and microglia (Iba1)
similar to the area used in rotating drum method for SD. and disrupts the blood–brain barrier via adenosine-mediated
This is in line with a recent study by Yin et al. [27] which signalling. Similarly, in our recent lab studies of 12 h of
has revealed that intermittent paradoxical sleep deprived acute sleep deprivation in rats, we observed reactive gliosis
mice took lesser time in reaching the hidden/visible plat- in astrocytic and microglial population along with secretion
form during Morris water maze test compared to control of pro-inflammatory mediators such as TNFα, IL-6 [12, 14].
mice. The study indicated concluded that familiarity of SD Activation of inflammatory cytokines along with profound

13
Molecular and Cellular Biochemistry (2018) 449:63–72 71

astrogliosis and microgliosis (Fig. 3a–c) and anxiety-like 4. Argente-Arizón P, Guerra-Cantera S, Garcia-Segura LM


behaviour (Fig. 1a–d) triggered due to chronic sleep depriva- et al (2017) Glial cells and energy balance. J Mol Endocrinol
58(1):R59–R71
tion is further indicative of neuroinflammation. 5. Wolf SA, Boddeke HWGM., Kettenmann H (2017) Microglia
in physiology and disease. Annu Rev Physiol 79:619–643
6. Bellesi M, Pfister-Genskow M, Maret S et al (2013) Effects
of sleep and wake on oligodendrocytes and their precursors. J
Neurosci 33(36):14288–14300
Conclusion 7. Zhao Z, Zhao X, Veasey SC (2017) Neural consequences of
chronic short sleep: reversible or lasting? Front Neurol 8:235
The current data suggest that inflammatory mediators 8. Bellesi M, de Vivo L, Tononi G, Cirelli C (2015) Effects of
increase during chronic sleep deprivation and trigger the sleep and wake on astrocytes: clues from molecular and ultras-
tructural studies. BMC Biol 13(1):66
activation of glial and microglial cells in discrete brain 9. Huber R, Ghilardi MF, Massimini M, Tononi G (2004) Local
regions. Upregulation in the protein expression of inflamma- sleep and learning. Nature 430(6995):78
tory cytokines, transcription factors as well as astroglial and 10. Tononi G, Cirelli C (2006) Sleep function and synaptic homeo-
microglial cells activation may explain the higher anxiety stasis. Sleep Med Rev 10(1):49–62
11. Yang G, Lai CSW, Cichon J et  al (2014) Sleep promotes
levels and learning and memory impairment associated with branch-specific formation of dendritic spines after learning.
chronic sleep deprivation and may prove to be the potential Sci 344(6188):1173–1178
targets to develop effective therapeutic interventions to over- 12. Mishra R, Manchanda S, Gupta M et al (2016) Tinospora cordi-
come these conditions. folia ameliorates anxiety-like behavior and improves cognitive
functions in acute sleep deprived rats. Sci Rep 6:25564
13. Manchanda S, Mishra R, Singh R et al (2017) Aqueous leaf
Acknowledgements  This work was supported by Department of Sci- extract of Withania somnifera as a potential neuroprotective
ence and Technology (DST), Government of India (GOI) project Grant agent in sleep-deprived rats: a mechanistic study. Mol Neurobiol
[Grant No. C/639/(IFD)/2015–2016] to Gurcharan Kaur. Shaffi Man- 54(4):3050–3061
chanda is thankful to Council of Scientific and Industrial Research 14. Kaur T, Singh H, Mishra R et al (2017) Withania somnifera
(CSIR), GOI for fellowship grant during the entire course of study. as a potential anxiolytic and immunomodulatory agent in
Harpal Singh and Taranjeet Kaur are thankful to University Grants acute sleep deprived female Wistar rats. Mol Cell Biochem
Commission (UGC), GOI and its scheme CPEPA (Centre with Poten- 427(1–2):91–101
tial for Excellence in Particular Area) for fellowship grant. Infrastruc- 15. Silva RH, Kameda SR, Carvalho RC et al (2004) Anxiogenic
ture provided by University Grants Commission (UGC), India under effect of sleep deprivation in the elevated plus-maze test in
University with Potential for Excellence (UPE) and CPEPA schemes mice. Psychopharmacology 176:115–122
and Department of Biotechnology (DBT), India under DISC facility is 16. Su CL, Chen CH, Lu HY et al (2004) The involvement of PTEN
highly acknowledged. The funding source had no role in study design; in sleep deprivation-induced memory impairment in rats. Mol
collection, analysis and interpretation of data; in writing of report; and Pharmacol 66:1340–1348
in decision to submit the article for publication. 17. Vgontzas AN, Fernandez-Mendoza J, Liao D et al (2013) Insom-
nia with objective short sleep duration: the most biologically
Compliance with ethical standards  severe phenotype of the disorder. Sleep Med Rev 17:241–254
18. Irwin MR (2015) Why sleep is important for health: a psycho-
Conflict of interest  The authors declare that they have no conflict of neuroimmunology perspective. Annu Rev Psychol 66:143–172
interest. 19. Irwin MR, Wang M, Ribeiro D et al (2008) Sleep loss activates
cellular inflammatory signaling. Biol Psychiatr 64:538–540
Ethical approval  All applicable international, national and/or institu- 20. Haack M, Sanchez E, Mullington JM (2007) Elevated inflam-
tional guidelines for the care and use of animals were followed. All matory markers in response to prolonged sleep restriction are
procedures performed in studies involving animals were in accordance associated with increased pain experience in healthy volunteers.
with the ethical standards of the institution or practice at which the Sleep 30:1145–1152
studies were conducted. 21. Zhu B, Dong Y, Xu Z et al (2012) Sleep disturbance induces
neuroinflammation and impairment of learning and memory.
Neurobiol Dis 48:348–355
22. Makarov SS (2001) NF-kappaB in rheumatoid arthritis: a piv-
otal regulator of inflammation, hyperplasia, and tissue destruc-
tion. Arthritis Res 3:200–206
References 23. Killeen MJ, Linder M, Pontoniere P, Crea R (2014) NF-κB sign-
aling and chronic inflammatory diseases: exploring the potential
1. Hurtado-Alvarado G, Domínguez-Salazar E, Pavon L et al (2016) of natural products to drive new therapeutic opportunities. Drug
Blood-brain barrier disruption induced by chronic sleep loss: low- Discov Today 19:373–378
grade inflammation may be the link. J Immunol Res. https​://doi. 24. Jacobs-Helber SM, Wickrema A, Birrer MJ, Sawyer ST (1998)
org/10.1155/2016/45760​12 AP1 regulation of proliferation and initiation of apoptosis
2. Besedovsky L, Lange T, Born J (2012) Sleep and immune func- in erythropoietin-dependent erythroid cells. Mol Cell Biol
tion. Pflug Arch Eur J Physiol 463:121–137 18:3699–3707
3. Roth AD, Núñez MT (2016). Oligodendrocytes: functioning 25. Gonzalez-Castañeda RE, Galvez-Contreras AY, Martínez-
in a delicate balance between high metabolic requirements and Quezada CJ et al (2016) Sex-related effects of sleep deprivation
oxidative damage. Glial cells in health and disease of the CNS, on depressive-and anxiety-like behaviors in mice. Exp Anim
Springer, New York, pp 167–181 65(1):97–107

13

72 Molecular and Cellular Biochemistry (2018) 449:63–72

26. Vollert C, Zagaar M, Hovatta I et al (2011) Exercise prevents sleep 35. Jhaveri KA, Ramkumar V, Trammell RA, Toth LA (2006) Spon-
deprivation-associated anxiety-like behavior in rats: potential role taneous, homeostatic, and inflammation-induced sleep in NF-κB
of oxidative stress mechanisms. Behav Brain Res 224(2):233–240 p50 knockout mice. Am J Physiol Regul Integr Comp Physiol
27. Yin M, Chen Y, Zheng H et al (2017) Assessment of mouse cog- 291(5):R1516–R152
nitive and anxiety-like behaviors and hippocampal inflammation 36. Basheer R, Shiromani PJ (2001) Effects of prolonged wakefulness
following a repeated and intermittent paradoxical sleep depriva- on c-fos and AP1 activity in young and old rats. Mol Brain Res
tion procedure. Behav Brain Res 321:69–78 89(1):153–157
28. Bolles RC (1960) Grooming behavior in the rat. J Comp Physiol 37. Trickler WJ, Mayhan WG, Miller DW (2005) Brain microvessel
Psychol 53:306–310 endothelial cell responses to tumor necrosis factor-alpha involve
29. Spruijt BM, Van Hooff JA, Gispen WH (1992) Ethology and neu- a nuclear factor kappa B (NF-κB) signal transduction pathway.
robiology of grooming behavior. Physiol Rev 72(3):825–852 Brain Res 1048(1):24–31
30. Kalueff AV, Tuohimaa P (2004) Grooming analysis algorithm for 38. Cirelli C, Pompeiano M, Tononi G (1993) Fos-like immunoreac-
neurobehavioural stress research. Brain Res Protoc 13(3):151–158 tivity in the rat brain spontaneous wakefulness and sleep. Arch
31. Kalueff AV, Aldridge JW, LaPorte JL et al (2007) Analyzing Ital Biol 131(4):327–330
grooming microstructure in neurobehavioral experiments. Nat 39. Cirelli C, Pompeiano M, Tononi G (1995) Sleep deprivation and
Protoc 2(10):2538 c-fos expression in the rat brain. J Sleep Res 4(2):92–106
32. Shiota N, Narikiyo K, Masuda A et al (2016) Water spray-induced 40. Jha MK, Lee WH, Suk K (2016) Functional polarization of neu-
grooming is negatively correlated with depressive behavior in the roglia: Implications in neuroinflammation and neurological dis-
forced swimming test in rats. J Physiol Sci 66(3):265–273 orders. Biochem Pharmacol 103:1–16
33. Kalueff AV, Tuohimaa P (2004) Experimental modeling of anxiety 41. Hurtado-Alvarado G, Domínguez-Salazar E, Velázquez-Mocte-
and depression. Acta Neurobiol Exp 64(4):439–448 zuma J, Gómez-González B (2016) A2A adenosine receptor
34. Zielinski MR, Kim Y, Karpova SA et al (2014) Chronic sleep antagonism reverts the blood-brain barrier dysfunction induced
restriction elevates brain interleukin-1 beta and tumor necrosis by sleep restriction. PLoS ONE 11(11):e0167236
factor-alpha and attenuates brain-derived neurotrophic factor
expression. Neurosci Lett 580:27–31

13

You might also like