Skip to main content
. 2024 May 2;25(9):4972. doi: 10.3390/ijms25094972

Table 1.

Synthesis of included studies [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41].

Scheme 1. Aim Animal Model/Cell Line Methods Results
1.
Basalay
et al.
2019
[25]
-Examine the
Neuroprotective
Effects of liraglutide
And semaglutide
Male nondiabetic Sprague-Dawley rats -A model of acute ischemic stroke (middle cerebral artery occlusion)
was used to assess the effects of
liraglutide or semaglutide administered i.v. at onset or s.c. before it.
-Infarct size and functional status
were evaluated after 24 or 72 h
of reperfusion.
-Liraglutide reduced brain damage
dose-dependent.
-Reperfusion delay is a limiting factor
For neuroprotection by liraglutide.
-Neuroprotection by semaglutide is at least as potent as by liraglutide.
-The principal clinic-related difference between semaglutide and liraglutide is the substantially longer half-life.
2.
Chen
et al.
2023
[26]
-Investigate differences in cognitive function and hippocampal phosphorylated protein expression in high-fat diet-induced obese mice
after treatment with
semaglutide and
empagliflozin.
C57BL/6JC male mice -Control (C) and high-fat diet (H)
-After 12 weeks, Group H
Was divided into:
-Group H
-Group semaglutide:
Intraperitoneal injections of 30 nmol/kg/d bodyweight of
Semaglutide for 12 weeks
-Group empagliflozin: gavage
administration of 10 mg/kg/d
bodyweight of empagliflozin
for 12 weeks
-Groups C and H received equal
volumes of saline via the same routes.
-The use of semaglutide treatment decreased escape latency and
Increased the total percentage of time spent swimming.
-Semaglutide and empagliflozin increased the phosphorylation of CACNA1D, CACNA1A, and CACNA1B proteins.
3.
Wang et
al. 2021
[27]
-Investigate
the functions of
semaglutide in
epilepsy and
inflammation
models
C57BL/6J mice -In vitro: inflammation model
Using lipopolysaccharide (LPS)
and nigericin stimulation in BV2
cells.
-In vivo: chronic epilepsy model
using a pentylenetetrazole (PTZ)
kindling method.
-Semaglutide decreased inflammation and reduced cell damage by blocking NLRP3 inflammasome activation in BV2 cells treated with LPS and nigericin.
-Semaglutide reduced seizure severity, prevented hippocampal neuron death, improved cognition, blocked NLRP3 inflammasome activation, and lowered inflammatory cytokine levels.
4.
M.A.
Sadek et
al. 2023
[28]
-Investigate the
Influence of
semaglutide on
experimental
autoimmune
encephalomyelitis (EAE)-induced
multiple sclerosis
(MS) in mice.
Adult male Swiss albino mice -The standard control group received saline (0.2 ml; intraperitoneal [i.p.]).
-The semaglutide-control group received semaglutide (25 nmol/kg/day; i.p.)
-The EAE group was subjected
to EAE induction and treated
with saline (0.2 ml; i.p.).
-The semaglutide-treated group
was subjected to EAE induction
and treated with semaglutide
(25 nmol/kg/day i.p.)
Two hours after EAE induction.
-All treatments were administered for two weeks starting from EAE induction.
-Semaglutide: reduced EAE-induced clinical signs
-Mitigates EAE-induced weight changes in CNS organs
-Attenuated EAE-induced motor impairment, cognitive dysfunction
-Amended EAE-induced histological alterations in the corpus callosum and brain atrophy and demyelination
-Lessened EAE-induced oxidative stress and neuroinflammation
5.
Zhang Q.
et al.
2022
[29]
-Explore the effect of
A1 astrocytes on BBB integrity after
ischemic stroke
Adult male ICR mice -Transient middle cerebral
artery occlusion (TMCAO).
-Immunohistochemical staining
of A1 (c3d) and A2 (S100A10)
were performed to characterize
astrocyte phenotypic changes
after TMCAO.
-Intraperitoneal injection with semaglutide to inhibit A1
astrocytes.
-The number of c3d+/GFAP+ A1 astrocytes increased within 14 days after S100A10+/GFAP+ TMCAO, while A2 astrocytes decreased.
-Treatment with semaglutide decreased the count of microglia expressing CD16/32 and astrocytes expressing C3d and GFAP, which are characteristic of A1 astrocytes.
-Semaglutide reduced brain infarct
volume and neuroinflammation; improved neurobehavioral outcomes.
6.
Chen et
al. 2023
[30]
-Investigate the effects
of semaglutide on
phosphorylated
protein expression
and its
neuroprotective
mechanism in
hippocampi of
high-fat-diet-
induced obese
mice
C57BL/6J male normal mice -Control group (c) regular diet
-Model group (H) high-fat
diet + saline
-Semaglutide group (H+
semaglutide, group S)
semaglutida 30 nmol/kg/d
for 12 weeks
-Morris water maze assay to detect cognitive function changes
-Phosphorylated proteomic
analysis to detect the hippocampal protein profile in mice
Semaglutide intervention on high-fat diet-induced obese mice:
-reduced body weight
-improved oxidative stress indexes
-shortened the water maze platform latency
-Semaglutide reduces phosphorylation and increases neuroprotection in obese mice.
7.
Z.J. Wang
et al.
2023
[31]
-Correlate the GLP-1R/SIRT1/GLUT4
pathway with
glucose metabolism.
-Investigate the
mechanism of action
For improving glucose metabolism in the
3xtg transgenic
mouse model of
Alzheimer’s disease and cultured neurons.
-male APP/PS1/Tau transgenic mice (3xtg)

-C57B6/129 wild-type mice (WT)
-WT + saline group
-3xtg + saline group
-3xtg + semaglutida group
-3xtg + semaglutide + EX527
(SIRT1 inhibitor) group
-15 mice in each group were
injected with semaglutide
(0.1 mg/kg, i.p.) or saline
(0.9%, i.p.) every other
day for 30 days before the
behavioral experiment.
Semaglutide increased the expression levels of SIRT1 and GLUT4 in the hippocampus of 3xtg mice, → improved learning and memory, and decreased Aβ plaques and neurofibrillary tangles.
-Improved glucose metabolism, promoted glycolysis, and improved glycolytic disorders
-Increased the membrane translocation of GLUT4 in cultured HT22 cells.
8.
Liu et al.
2022
[40]
-Explore the
neuroprotective
effects of
semaglutide in PD
-Compared the
effect of semaglutide with liraglutide at
the same dose
-SH-SY5Y
The cell line of human neuroblastoma
-Treated the human
neuroblastoma SH-SY5Y
the cell line with 6-hydroxydopamine
(6-OHDA) as a PD in vitro model
-Semaglutide and liraglutide protect against 6-OHDA cytotoxicity by increasing autophagy flux and decreasing oxidative stress and mitochondrial dysfunction in SH-SY5Y cells.
-Semaglutide was superior to
liraglutide for most parameters
measured
9.
L. Zhang
et al.
2018
[32]
-Report the protective effects of semaglutide (25 nmol/kg ip, once daily for seven days)
in the MPTP mouse model of PD.
-Compare the
neuroprotective effects of semaglutide and
liraglutide when given
at the exact dosage.
-Male C57BL/6
Mice
-Control group treated with saline
-Liraglutide group treated with saline and liraglutide
-Semaglutide group treated with
saline and semaglutide
-MPTP group treated with MPTP
-MPTP (once daily 20 mg/kg i.p.
for seven days) followed
immediately by liraglutide-treated group (25 nmol/kg i.p. once daily for seven days)
-MPTP (20 mg/kg i.p. once daily
for seven days) followed
immediately by semaglutide-treated group (25 nmol/kg i.p. once daily for seven days).
-Semaglutide and liraglutide were found to have improved the motor impairments caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP).
-Semaglutide was found to be better than liraglutide in most of the
measurements taken.
10.
L. Zhang
et al.
2019
[33]
-Investigate the neuroprotective effects of semaglutide
(25 nmol/kg i.p. once every two days for 30 days) and liraglutide
(25 nmol/kg i.p. once daily for 30 days) in the chronic MPTP mouse model of PD.
-Male C57BL/6
Mice
-Control group treated with saline
-Liraglutide group (25 nmol/kg i.p. once daily for 30 days)
-Semaglutide group (25 nmol/kg i.p. once every two days for 30 days)
-MPTP group treated with MPTP alone (once daily 20 mg/kg i.p.
for 30 days)
-MPTP (once daily 20 mg/kg i.p.
for 30 days) + liraglutide-treated
group (25 nmol/kg i.p. once
daily for 30 days)
-MPTP (20 mg/kg i.p. once daily
for 30 days) + semaglutide-treated group (25 nmol/kg i.p. once every two days for 30 days).
MPTP-induced increase in alfa-syn expression in the brain is reduced by semaglutide and liraglutide. Semaglutide is more potent than once daily liraglutide in most parameters measured.
11.
L. Zhang
et al.
2022
[34]
-Developed a dual
GLP-1/GIP receptor
agonist (DA5-CH)
that can cross the
blood-brain barrier
at a higher rate than
semaglutide.
-Tested semaglutide
and DA5-CH in the
6-OHDA-lesion rat
model of PD
-Adult male
Sprague-Dawley (SD) rats
-Stereotactic surgery
-A sham + saline group
-A 6-OHDA+ saline group
-A 6-OHDA+ semaglutide
group
-A 6-OHDA+DA5-CH group
-Treatment was semaglutide
or DA5-CH (25 nmol/kg, i.p.)
daily for 30 days postlesion.
-Both drugs protected dopaminergic neurons and increased TH expression in the substantia nigra.
-The level of monomer and aggregated α-synuclein was reduced.
12.
X. Yang
et al.
2019
[35]
-Providing new
insight
into the
semaglutide
neuroprotective
effects
in the rat brain
-Adult male
Sprague
–Dawley
-Permanent middle cerebral artery occlusion (PMCAO) model
-Sham-operated group
-Vehicle control group
-Semaglutide group—2 h after
MCAO intraperitoneally
(10 nmol/kg),
-Injected every second day
for 1, 7, 14, and 21 days
-Tissue sampling after 21
days of observation
-After ischemia, semaglutide treatment improved the functional recovery of rats without affecting blood glucose levels.
-The drug reduced inflammation and apoptosis and normalized cell growth signaling and neurogenesis.
13.
Poupon-
Bejuit
et al.
2022
[36]
-Evaluate the
therapeutic efficacy
of semaglutide
in a mouse model
of INAD
(infantile
neuroaxonal
dystrophy)
-Pla2g6−/− mice -In this study, a mouse model
with the Pla2g6 knock-in was
given semaglutide at three
different doses through
intraperitoneal injection once
a week. The treatment began
when the mice were three
weeks old, and their survival was
monitored and compared to age-matched untreated Pla2g6−/− mice and wild-type mice (WT), which were used as controls.
-Weekly delivery of high-dose semaglutide improved lifespan and locomotor function in juvenile INAD mice.
Semaglutide has been shown to increase the levels of molecules that protect the brain while simultaneously decreasing those molecules that promote neurodegeneration.
-The expression of mediators in the apoptotic and necroptotic pathways was significantly reduced in mice treated with semaglutide.
14.
T.S.
Salameh
et al.
2020
[37]
-Compare the
pharmacokinetics
of 125I-labeled single IRAs (exendin-4,
liraglutide,
lixisenatide,
semaglutide) and
125I-labeled dual
IRAs
-Male CD-1 mice -Consequently, compared
brain uptake pharmacokinetics
of intravenous 125I-labeled
IRAs in adult CD-1 mice
over 60 min
-The nonacylated and
nonpegylated IRAs (exendin-4, lixisenatide, Peptide 17, DA3-CH, and DA-JC4) had significant rates of blood-to-brain influx (Ki).
-The acylated IRAs (liraglutide, semaglutide, and Peptide 18) did not measurably cross the BBB.
The capillaries completely segregated semaglutide and Peptide 18, and none of the 125I-semaglutide or Peptide 18 was found in the brain tissue.
15.
Yan-fang Chang
et al.
2020
[41]
-Explore the
potential
mechanisms of
semaglutide
against AD
-SH-SY5Y cells -SH-SY5Y cells damaged by
Ab25–35 were treated by
semaglutide.
-Autophagy-related proteins
and apoptosis-related proteins
were measured to explore
molecular mechanisms.
-Semaglutide inhibited apoptosis
and increased the expression of Bcl2 inhibited by Ab25–35.
-Semaglutide activity in preventing
SH-SY5Y cells against Ab25–35 present a potential mechanism for enhancing autophagy and inhibiting apoptosis.
16.
Thornton
et al.
2024
[38]
-Identify specific NLRP3-sensitive
mechanismscontributing to
obesity-induced
inflammation
-Compare an
NLRP3 inhibitor
to semaglutide
to evaluate its
association with
systemic inflammatory response, and cerebral gliosis
-Male C57BL/6J mice -To investigate whether NLRP3
activation contributes to
the pathogenesis of diet-induced
obesity (DIO) in mice, two
different clinical-stage
NLRP3 inflammasome inhibitors
were tested.
-After being on a high-fat diet
for 15 weeks, mice induced with
diet-induced obesity (DIO) weighed significantly more than the mice fed a regular chow-based diet.
To understand the effect of
the LRP3 inhibitor NT-0249, it was compared with semaglutide or calorie restriction for an additional 28 days.
-NLRP3 inhibitors can block the cellular inflammatory response to obesity-related molecular patterns.
-In pre-existing obese states in mice, inhibiting NLRP3 can reduce systemic inflammation and astrogliosis.
When dosed to achieve brain exposure, NLRP3 inhibitors can reverse pre-established obesity at a similar efficacy to the GLP-1RA semaglutide.
17.
Shnaien
et al.
2023
[39]
-Examine the
neuroprotective
effects of
semaglutide
during
endotoxemia
and its role in
modulating
pro-inflammatory
mediators.
-Adult male Swiss
albino mice
-Laparotomy without/with cecal ligation and puncture (CLP)
-Semaglutide administration before CLP
-Sham group: laparotomy surgery
without CLP.
-CLP group: (sepsis group)
•Vehicle group: mice were given
an equal volume of distilled water
(DW) by subcutaneous injection
Daily for seven days before CLP.
•Semaglutide group: mice
received 40 µg/kg of semaglutide
-The brain tissue level of TLR4\STAT3 of the semaglutide-treated group was significantly lower.
-The brain tissues obtained from mice treated with semaglutide showed significantly less cellular injury.
-Semaglutide can protect against sepsis and prevent brain dysfunction.