ArticlePDF Available

Exendin-4 alleviates β-Amyloid peptide toxicity via DAF-16 in a Caenorhabditis elegans model of Alzheimer's disease

Authors:

Abstract and Figures

Epidemiological analyses indicate that type 2 diabetes mellitus (T2DM) is a risk factor for Alzheimer's disease (AD). They share common pathophysiological mechanisms. Thus, it has been increasingly suggested that several anti-T2DM drugs may have therapeutic potential in AD. Exendin-4, as a glucagon-like peptide-1 (GLP-1) receptor agonist, is an approved drug used to treat T2DM. In this research, the neuroprotective effect of Exendin-4 was investigated for the first time using transgenic Caenorhabditis elegans. Our results demonstrated that Exendin-4 attenuated the amyloid-β (1-42) (Aβ1-42) toxicity via multiple mechanisms, such as depressing its expression on protein and mRNA and reducing Aβ (1-42) accumulation. Exendin-4 at 0.5 mg/ml had been shown to extend life by 34.39% in CL4176 and delay the onset of paralysis in CL4176 and CL2006 which were increased by 8.18 and 8.02%, respectively. With the treatment of Exendin-4, the nuclear translocation of DAF-16 in the transgenic nematode TJ356 was enhanced. Superoxide dismutase-3 (SOD-3), as a downstream target gene regulated by DAF-16, was upregulated on mRNA level and activity. The reactive oxygen species (ROS) level was decreased. In contrast, we observed that the ability of Exendin-4 to regulate SOD was decreased in CL4176 worms with the DAF-16 gene silenced. The activity of SOD and the mRNA level of sod-3 were downregulated by 30.45 and 43.13%, respectively. Taken together, Exendin-4 attenuated Aβ (1-42) toxicity in the C. elegans model of AD via decreasing the expression and the accumulation of Aβ (1-42). Exendin-4 exhibited the ability of antioxidant stress through DAF-16. With continuous research, Exendin-4 would become a potential therapeutic strategy for treating AD.
Content may be subject to copyright.
TYPE Original Research
PUBLISHED 05 August 2022
DOI 10.3389/fnagi.2022.955113
OPEN ACCESS
EDITED BY
Michael Wink,
Heidelberg University, Germany
REVIEWED BY
Chatrawee Duangjan,
University of Southern California,
United States
Shaoxiong Zhang,
Fujian Agriculture and Forestry
University, China
*CORRESPONDENCE
Xiangwei Song
songxiangwei@ccsfu.edu.cn
Xueli Wang
xueliwang101@hotmail.com
SPECIALTY SECTION
This article was submitted to
Alzheimer’s Disease and Related
Dementias,
a section of the journal
Frontiers in Aging Neuroscience
RECEIVED 28 May 2022
ACCEPTED 11 July 2022
PUBLISHED 05 August 2022
CITATION
Song X, Sun Y, Wang Z, Su Y, Wang Y
and Wang X (2022) Exendin-4
alleviates β-Amyloid peptide toxicity
via DAF-16 in a Caenorhabditis
elegans model of Alzheimer’s disease.
Front. Aging Neurosci. 14:955113.
doi: 10.3389/fnagi.2022.955113
COPYRIGHT
©2022 Song, Sun, Wang, Su, Wang
and Wang. This is an open-access
article distributed under the terms of
the Creative Commons Attribution
License (CC BY). The use, distribution
or reproduction in other forums is
permitted, provided the original
author(s) and the copyright owner(s)
are credited and that the original
publication in this journal is cited, in
accordance with accepted academic
practice. No use, distribution or
reproduction is permitted which does
not comply with these terms.
Exendin-4 alleviates β-Amyloid
peptide toxicity via DAF-16 in a
Caenorhabditis elegans model
of Alzheimer’s disease
Xiangwei Song1*, Yingqi Sun1, Zhun Wang2, Yingying Su1,
Yangkun Wang1and Xueli Wang3*
1School of Life Sciences, Changchun Normal University, Changchun, China, 2Plant Inspection and
Quarantine Laboratory, Changchun Customs Technical Center, Changchun, China, 3School of
Grain, Jilin Business and Technology College, Changchun, China
Epidemiological analyses indicate that type 2 diabetes mellitus (T2DM) is a risk
factor for Alzheimer’s disease (AD). They share common pathophysiological
mechanisms. Thus, it has been increasingly suggested that several anti-T2DM
drugs may have therapeutic potential in AD. Exendin-4, as a glucagon-like
peptide-1 (GLP-1) receptor agonist, is an approved drug used to treat T2DM. In
this research, the neuroprotective eect of Exendin-4 was investigated for the
first time using transgenic Caenorhabditis elegans. Our results demonstrated
that Exendin-4 attenuated the amyloid-β(1-42) (Aβ1-42) toxicity via multiple
mechanisms, such as depressing its expression on protein and mRNA and
reducing Aβ(1-42) accumulation. Exendin-4 at 0.5 mg/ml had been shown
to extend life by 34.39% in CL4176 and delay the onset of paralysis in
CL4176 and CL2006 which were increased by 8.18 and 8.02%, respectively.
With the treatment of Exendin-4, the nuclear translocation of DAF-16 in the
transgenic nematode TJ356 was enhanced. Superoxide dismutase-3 (SOD-3),
as a downstream target gene regulated by DAF-16, was upregulated on mRNA
level and activity. The reactive oxygen species (ROS) level was decreased.
In contrast, we observed that the ability of Exendin-4 to regulate SOD was
decreased in CL4176 worms with the DAF-16 gene silenced. The activity of
SOD and the mRNA level of sod-3 were downregulated by 30.45 and 43.13%,
respectively. Taken together, Exendin-4 attenuated Aβ(1-42) toxicity in the
C. elegans model of AD via decreasing the expression and the accumulation of
Aβ(1-42). Exendin-4 exhibited the ability of antioxidant stress through DAF-16.
With continuous research, Exendin-4 would become a potential therapeutic
strategy for treating AD.
KEYWORDS
Alzheimer’s disease, Exendin-4, Caenorhabditis elegans, DAF-16, Amyloid-βpeptide,
neuroprotection
Frontiers in Aging Neuroscience 01 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
Introduction
As a chronic neurodegenerative disease, Alzheimer’s disease
(AD) poses a serious threat to individuals’ health. Despite
years of massive investigation, the pathogenic mechanism of
AD is still in the stage of hypothesis theories, such as the
hyperphosphorylated tau protein hypothesis, amyloid toxicity
hypothesis, oxidative stress hypothesis, and acetylcholine
hypothesis. However, the amyloid toxicity hypothesis is widely
accepted. Amyloid-βpeptides (Aβ1-40 and Aβ1-42) are
produced due to aberrant cleavage of the amyloid precursor
protein (APP), which accumulates on the neuron. Furthermore,
Aβ(1-42), as a mediator of oxidative stress, has been proposed
to play a central role in the pathogenesis of AD (Butterfield and
Boyd-Kimball, 2004), such as decreasing the levels of superoxide
dismutase (SOD) and increasing the levels of MDA and reactive
oxygen species (ROS) (Wang et al., 2021). As a target, amyloid is
often applied to drug screening and clinical diagnosis.
The data of epidemiological investigation suggest that the
correlation between type 2 diabetes mellitus (T2DM) and AD is
highly significant. Diabetic patients are twice as likely to suffer
from AD as the normal population. AD and T2DM always
share common pathophysiological symptoms (Caberlotto et al.,
2019), such as hyperglycemia, hypercholesterolemia, and insulin
signaling dysfunction.
Thus, it has been strongly suggested that some drugs treated
with T2DM may have therapeutic potential in AD (Sebastiao
et al., 2014). Exendin-4, as a glucagon-like peptide-1 (GLP-1)
receptor agonist, has been approved for the treatment of T2DM.
However, the strong preclinical evidence suggests that Exendin-
4 is neuroprotective in AD (Mullins et al., 2019). For instance,
Exendin-4 appears to prevent the hyperphosphorylation of
AD-associated tau protein due to the increased insulin signaling
pathway in the brain (Xu et al., 2015). Exendin-4 significantly
attenuated Aβ-induced memory deficits in the Morris water
maze and Y-maze test (Garabadu and Verma, 2019).
Many kinds of transgenic Caenorhabditis elegans models
(Wu and Luo, 2005) of AD had been used to research the
pathogenesis of AD (Link, 2006) due to their short lifetime
and progressive paralysis phenotype. Moreover, the transgenic
C. elegans, such as CL4176 (Wu and Luo, 2005; Zhang et al.,
2016) and CL2006 (Smith and Luo, 2003), promotes the onset
of the ROS, and it is similar to the onset of ROS observed in
patients with AD, which is consistent with the amyloid cascade
hypothesis on oxidative stress. Oxidative stress is extensive in the
AD brain. Aβ(1-42) has been shown to induce oxidative stress
and neurotoxicity in vitro and in vivo. Therefore, transgenic
C. elegans strains accelerate the pace of understanding the
mechanisms of Aβ(1-42) toxicity in biological systems. At the
same time, they can be used for anti-AD drug screening in vivo
(Ewald and Li, 2010; Wolozin et al., 2011).
Herewith, we mainly employed transgenic C. elegans
as the model to investigate the neuroprotective effects of
Exendin-4. Our conclusion presents that Exendin-4 develops
its neuroprotective effect by reducing the accumulation and
expression of Aβ(1-42) directly. Exendin-4 also showed the
excellent performance of antioxidant stress via the transcription
factors DAF-16.
Materials and methods
C. Elegans strains and maintenance
The wild-type N2 worms and the transgenic worms
CL4176 {smg-1ts [myo-3/Aβ1–42 long 3-untranslated
region (UTR)]} were obtained from Caenorhabditis Genetics
Center (University of Minnesota, Minneapolis, MN). TJ356
[daf-16p::daf-16a/b::GFP +rol-6], CL2006 {dvIs2 [pCL12(unc-
54/human Aβ1–42 minigene) +pRF4]}, the transgenic
CF1553 {muIs84 [(pAD76) sod-3p::GFP +rol-6(su1006)]},
and Escherichia coli were kindly donated by Dr. Liping Wang.
CL4176 was maintained at 16 C. The wild-type N2, CL2006,
CF1553, and TJ356 were maintained at 20 C.
All worms were on a solid nematode growth medium
(NGM) seeded with live E. coli (OP50) as a food source.
Brood size and body length assays
The wild-type N2 worms were used in brood size and
body length assays. For the brood size assay, each worm
(L4 stage) was transferred onto NGM plates treated with
different concentrations of Exendin-4 (0.1, 0.3, 0.5, and 1.2
mg/ml). The worms were cultivated at 20 C. The number of
all eggs that it hatched was recorded (Rangsinth et al., 2019).
For the body length assay, worms (L4 stage) were transferred
onto different NGM plates treated with different concentrations
of Exendin-4 (0.1, 0.3, 0.5, and 1.2 mg/ml) and cultured at 20 C.
Adult day-1 worms were paralyzed using 0.1% sodium azide and
mounted on a glass slide. At least thirty worms per group were
imaged using light microscopes (Leica, DM3000 LED, Germany)
and measured using the Leica Application Suite version 4.12
software (Leica, Germany).
Paralysis assays
We synchronized the transgenic C. elegans strain CL4176 on
different concentrations of Exendin-4 (0 mg/ml, 0.02 mg/ml, 0.1
mg/ml, 0.3 mg/ml, 0.5 mg/ml, and 1.2 mg/ml) and caffeine (1.2
mg/ml) treated plates at 16 C for 48 h. Then, worms (L2 stage)
were transferred from 16 C to 23 C. After cultivating 40 h at
23 C, the paralyzed worms had been counted at 2 h intervals
until all the worms were paralyzed (Dostal and Link, 2010;
Takahashi et al., 2014). The principle of paralysis is stipulated
Frontiers in Aging Neuroscience 02 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
that they moved their heads only or failed to move their bodies
by touching stimuli.
Life span assays
To obtain the synchronized worms, gravid adults (L1 stage)
of CL4176 were raised on different concentrations of Exendin-4
(0 mg/ml, 0.02 mg/ml, 0.1 mg/ml, 0.3 mg/ml, and 0.5 mg/ml)-
treated plates to lay eggs for 2–3 h. Generally, 280 worms
should be promised in one plate. After 48 h of synchronization,
Aβ(1-42) expression was induced by upshifting the temperature
to 23 C. We continued to collect the data for 20 days. All life
span assays were preceded independently at least three times.
Worms that were missing, attaching to walls or worm bags, were
not included during the life statistics.
TABLE 1 Lists of primers.
Gene Forward primer Reverse primer
actin-1 5AAGACCACGTCAT
CAAGG3
5TTCTCCATATCAT
CCCAGTT3
daf-16 5GCGAATCGGTTCCAGCAAT
TCCAA3
5ATCCACGGACACTGTT
CAACTCGT3
sod-3 TATTAAGCGCGACTTCGG
TTCCCT
CGTGCTCCCAAACGTCAAT
TCCAA3
Aβ(1-42) CCGACATGACTCAGGATA
TGAAGT
CACCATGAGTCCAATGA
TTGCA3
Western blotting
Worms were treated with Exendin-4 (0.5 mg/ml) and
induced for 44 h at 23 C. Worms (L4 stage) were harvested
and centrifuged at 12,000 rpm for 10 min. The pellets were
resuspended and sonicated after freeze-thawed as described
in the WormBook (http://www.wormbook.org). The gel was
transferred onto polyvinylidene fluoride (PVDF) membranes.
The monoclonal antibody (ab201060, ABCam, UK) was used to
detect Aβ(1-42). Experiments were repeated independently for
three times.
Staining of Aβ
Worms (L2 stage) were transferred from 16 C to 23 C
to cultivate for 40 h after synchronization. A total of 100
worms (L4 stage) were picked up and washed with M9
buffer. After centrifugation, worms were fixed with 150 µl of
4%paraformaldehyde/PBS buffer (pH =7.4) at 4 C for 24 h.
Fixed worms were treated with 150 µl 5% β-mercaptoethanol,
1% Triton X-100, and 125 mM Tris pH 7.4, at 37 C for 24 h.
Treated worms were washed with PBS buffer three times, and
200 µl of 0.125% thioflavin T (Sigma) in 50% ethanol was added
for 90 s and destained with 500 µl of 50% ethanol until the
suspension became transparent. Worms were resuspended with
100 µl M9 buffer. The animals were finally transferred to slides
using a drop of glycerol, and fluorescence images were acquired
by confocal laser scanning fluorescence microscopy (CarlZeiss
LSM710, Germany).
FIGURE 1
The eects of Exendin-4 on wild-type N2 Caenorhabditis elegans.(A) Body length assay and (B) brood size assay. There was no significant
dierence in the treatment groups on brood size and body length. Values are mean ±SEM of experiments repeated independently at least for
three times.
Frontiers in Aging Neuroscience 03 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
FIGURE 2
Exendin-4 alleviated Aβ-induced paralysis in transgenic C. elegans strains CL4176 and CL2006. (A) In CL4176. (B) In CL2006. Curves show the
process of Aβ-induced paralysis in transgenic C. elegans treated with a vehicle control (H2O) or dierent concentrations of Exendin-4 (0.02
mg/ml, 0.1 mg/ml, 0.3 mg/ml, 0.5 mg/ml, and 1.2 mg/ml). Caeine (1.2 mg/ml) is positive control. The onset of paralysis was prolonged in
CL4176 and CL2006 worms in a dose-dependent manner (at least 40 worms were tested in each group, and the experiments were repeated
independently for three times).
FIGURE 3
The life span assay administrated Exendin-4 in transgenic
C. elegans strain CL4176. CL4176 was treated with a vehicle
control (H2O) or dierent concentrations of Exendin-4 (0.02
mg/ml, 0.1 mg/ml, 0.3 mg/ml, and 0.5 mg/ml). The median
survival of worms treated with 0.5 mg/ml Exendin-4 was 13
days, which had a significant expansion compared with 8 days
of control worms (p<0.001). The experiment was repeated
independently for three times.
ROS assays
Age-synchronized C. elegans (more than 30 eggs per
plate) were transferred to NGM plates containing vehicle
or 0.5 mg/ml Exendin-4 and incubated for 48 h at 16 C.
After incubation at 23 C for 44 h, ROS was determined
based on published studies (Wu et al., 2012). The examined
nematodes were transferred to 0.5 ml of M9 buffer containing
5µM CM-H2DCFDA (D6883, Sigma-Aldrich, USA) and
preincubated for 3 h at 20 C. Later, nematodes were mounted
on 2 % agar pads and examined using a fluorescence
microscope (Nikon, SMZ 18, Japan) at 495 nm of excitation
wavelength and 537 nm of emission filter. More than 30
animals were counted for the statistical analysis. The relative
fluorescence intensities of the worm were semi-quantified
using the ImageJ software. The semi-quantified ROS was
expressed as relative fluorescence units (RFU). Three replicates
were performed.
SOD activity assays
The transgenic C. elegans strain CL4176 was synchronized
on 0.5 mg/ml Exendin-4 treated and untreated plates at 16
C for 48 h, respectively. Then, t he worms (L2 stage) were
cultivated for another 40 h at 23 C. A total of 200 worms
(L4 stage) were collected and centrifuged with M9 buffer.
The pellets were disrupted according to the Western blotting
assay protocol. After protein quantification using bicinchoninic
acid (BCA) assay, the analysis of SOD activity was proceeded
using nitrotetrazolium blue chloride (NBT) method (Total
Superoxide Dismutase Assay Kit with NBT, #S0109, Beyotime
Biotechnology, China). Four replicates were performed.
Real-time PCR analysis
Worms were treated according to the Western blotting
protocol. The worms (L4 stage) were collected by washing the
plate with 1 ml M9 buffer. The pellets were washed three times by
Frontiers in Aging Neuroscience 04 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
FIGURE 4
Exendin-4 attenuated the protein and mRNA levels of Aβ(1-42) in C. elegans CL4176. (A) Western blotting assays of experiments repeated
independently for three times are shown. Control groups were not treated with Exendin-4. The test groups were treated with 0.5 mg/ml of
Exendin-4. (B) The relative intensity of Aβ(1-42) expression was analyzed using the ImageJ software. After treatment with 0.5 mg/ml Exendin-4,
the protein expression level was reduced by 39.78%. p<0.01 (**).(C) The mRNA levels of Aβ(1-42) were analyzed by real-time PCR. mRNA levels
were reduced by 29.67%. p<0.001 (***).
centrifugation at 25,000 rpm for 5 min. The worms were freeze-
thawed and transferred directly into 1 ml of TRIzol reagent
(Thermo Fisher Scientific, Shanghai, China). The total nematode
RNA was extracted first and measured using a Nanophotometer
(IMPLEN N60, German). The cDNAs were synthesized using
the Transcription Kit (Promega, A5000, USA). The expression
of genes was determined by real-time PCR performed on 7500
Real-Time PCR System (ABI) with SYBR Green real-time PCR
kit (Roche). According to the instruction of the real-time PCR
kit, the cycling conditions were as follows: (1) holding stage at 50
C for 2 min; (2) holding stage at 95 C for 10 min; (3) cycling
stage at 95 C for 15 s, at 55 C for 1 min; the number of cycling
for 40 cycles; and (4) melt curve stage at 95 C for 10 s, at
60 C for 1 min, at 95 C for 10 s. The relative levels of gene
expression were calculated using the 2 11CT method using
the gene actin-1 as the internal control. The experiment was
repeated in triplicate. Primers are listed in Table 1.
DAF-16 nuclear translocation analysis
Age-synchronized nematodes of the transgenic strain TJ356
stably expressing a DAF-16::GFP fusion protein were treated
with Exendin-4 (0.5 mg/ml) and kept at 20 C for 48 h. Later,
the worms (L4 stage) were anesthetized with 0.1% sodium and
transferred to slides using a drop of glycerol. The subcellular
DAF-16 distribution among twenty worms per group was
analyzed using a Nikon SMZ 1500 fluorescence microscope. The
experiments were performed more than 3 times. The amount of
DAF-16::GFP was analyzed using the ImageJ software.
The C. Elegans RNA interference assay
Worms (L1 stage) were treated with 0.5 mg/ml Exendin-4.
DAF-16 was knocked down by feeding the C. elegans CL4176
with E. coli strain R13H8.1 bacteria carrying daf-16 dsRNA.
Worms fed with R13H8.1 bacteria with the empty vector L4440
were used as negative controls. Notably, 1 mM isopropyl β-D-1-
thiogalactopyranoside (IPTG) was used on NGM plates. CL4176
was maintained at 16 C after synchronization. After 48 h, more
than 50 worms (L2 stage) were cultured for another 40 h at
23 C. Then, the process of paralysis assays was carried out. The
experiments were performed more than 3 times.
Statistical analysis
Statistical analyses of differences between groups in the
paralysis assays and life span assay were performed using
Frontiers in Aging Neuroscience 05 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
FIGURE 5
Aβ(1-42) accumulation assay with thioflavin T staining in transgenic C. elegans CL4176. (A) Control groups were not treated with Exendin-4. Aβ
(1-42) deposits stained with thioflavin-T show abundant fluorescence patches near the pharynx in transgenic worm CL4176. (B–D) Transgenic
worms fed with 0.3 mg/ml, 0.6 mg/ml, and 1.2 mg/ml Exendin-4. They show a significant reduction in the deposition of Aβ(1-42) in a
dose-dependent way. The yellow arrows show the sites of Aβ(1-42). (E) The number of Aβ(1-42) patches in dierent groups. p<0.01 (**) and
p<0.001 (***).
the log-rank test. Data other than paralysis and life span
were analyzed using Student’s t-test. Results were expressed
as the mean ±standard deviation of experiments repeated
independently for three times. The GraphPad Prism software 5.0
(GraphPad, La Jolla, CA, United States) was used for statistical
analyses. The difference in statistical data is shown with p-value;
p<0.05 (), p<0.01 (∗∗), and p<0.001 (∗∗∗ ) were regarded
as significant.
Results
Eects of exendin-4 on the development
and reproduction of C. Elegans
To investigate the toxicity of Exendin-4, we performed body
length and brood size assays to monitor the development and
fertility rate of wild-type N2 worms, respectively. The data
showed that Exendin-4 with 0.1, 0.3, 0.5, and 1.2 mg/ml has no
toxic effects on nematodes in terms of spawning and body length
(Figures 1A,B).
Exendin-4 delays the progression of
paralysis in AD worms
To investigate the potential influence of Exendin-4 on the
progression of paralysis induced by A β(1-42), we treated
CL4176 and CL2006, respectively. The onset of paralysis
was dramatically delayed in a dose-dependent manner after
treatment of Exendin-4 (Figures 2A,B). The low dose of
Exendin-4 (0.02 mg/ml) cannot slow down the process
of paralysis significantly. Notably, 1.2 mg/ml of Exendin-
4 displayed the best effect among all groups. The mean
paralysis time was increased by 8.18% (CL4176) and 8.02%
(CL2006), respectively. In CL4176, the duration of paralysis
in the 1.2 mg/ml Exendin-4 group was prolonged by 3.67 h.
The average paralysis time was 0.7 h longer than that of the
positive control (Supplementary Table 1). Exendin-4 showed
better performance than the positive control. In CL2006, the
duration of paralysis in the 1.2 mg/ml Exendin-4 group was
prolonged by 3.58 h. The average paralysis time was 0.11 h
longer than that of the positive control (Supplementary Table 2).
Combined with the experimental results in the two mutant
nematodes, these observations suggested that Exendin-4 may
Frontiers in Aging Neuroscience 06 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
FIGURE 6
The eect of antioxidant stress of Exendin-4 in C. elegans CL4176 and CL2006. ROS was analyzed using CM-H2DCFDA. Photographs were
taken using a fluorescence microscope at 495 nm of excitation wavelength and 537 nm of emission filter. (A,D) The untreated CL4176 and
CL2006. (B,E) The treated CL4176 and CL2006 with 0.5 mg/ml Exendin-4. (C,F) The fluorescence intensity analyzed using the ImageJ software.
(G,H) The mRNA levels of superoxide dismutase-3 (sod-3). (I,J) The activity of sod-3. All the experiments were repeated independently for three
times. p<0.05 (*), p<0.01 (**).
have the potential to relieve the paralysis caused by Aβ(1-
42).
Exendin-4 increases life span in AD
worms
The life span assay was examined under different Exendin-
4 concentrations (0.02, 0.1, 0.3, and 0.5 mg/ml). We collected
the data for 20 days. Exendin-4 treatment showed a significant
increase in the life span in a dose-dependent manner (Figure 3).
Median survival time was prolonged in each treated group.
Notably, 0.3 mg/ml and 0.5 mg/ml of Exendin-4 displayed
remarkable performance in extending life span. They almost
increased the median survival time to 13 days. Compared
with the control group, lifetime was increased by 32.05 and
34.39%, respectively (Supplementary Table 3). Results implied
that Exendin-4 treatment attenuated the harmful effect of
longevity induced by Aβ(1-42) toxicity in worms.
Exendin-4 reduced the levels of Aβ(1-42)
in AD worms
To clarify why Exendin-4 can relieve paralysis in CL4176,
we monitored the Aβ(1-42) levels on protein and mRNA.
All worms were collected after being induced to paralysis
for 44 h, and parallel populations were processed for real-
time PCR and the Western blotting assay. The group treated
with 0.5 mg/ml Exendin-4 resulted in a 39.78% decrease in
the protein expression of Aβ(1-42) (Figures 4A,B) and a
significant reduction by 29.67% in the mRNA level of Aβ(1-42)
(Figure 4C). Thus, the ability of Exendin-4 to delay paralysis
is directly demonstrated by downregulating the protein and
mRNA levels of Aβ(1-42). We detected the change of Aβ(1-
42) accumulation with a thioflavin T staining assay. We focused
on the area near the pharynx of C. elegans CL4176 to count
the number of deposits. Lots of Aβ(1-42) deposits appeared
after being induced (Figures 5A–D). Instead, worms treated with
Exendin-4 displayed less Aβ(1-42) deposits in a dose-dependent
Frontiers in Aging Neuroscience 07 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
FIGURE 7
Exendin-4 lost the ability to alleviate Aβ-induced paralysis when the daf-16 gene is knockout in transgenic C. elegans strain CL4176. DAF-16
expression was knocked down by feeding the C. elegans CL4176 with Escherichia coli strain R13H8.1 bacteria carrying daf-16 dsRNA. Curves
show that the eect of Exendin-4 (0.5 mg/ml) on inhibiting the paralysis rate of C. elegans vanished after daf-16 gene knockout. (At least 50
worms were tested in each group, and the experiments were repeated independently for three times.).
manner (Figure 5E). Exendin-4 possessed the positive effect to
inhibit the accumulation of Aβ(1-42).
Exendin-4 increases oxidative stress
resistance in AD worms
The ROS will be increased by triggering the expression of Aβ
(1-42). The increased production of ROS associated with age and
neurotransmission in neurons leads to cognitive dysfunction
in AD.
To test the antioxidant effect of Exendin-4, two kinds of
strains (CL2006 and CL4176) were treated with 0.5 mg/ml
of Exendin-4.
Reactive oxygen species was measured using the
CM-H2-DCFDA method. The worms treated with Exendin-4
showed lower production of ROS in CL2006 and CL4176
(Figures 6A,B,D,E). The fluorescence intensity was analyzed
using the ImageJ software. A volume of 0.5 mg/ml Exendin-4
decreased ROS level by 13.57% (p<0.01) and 17.22% (p<0.05)
in CL4176 and CL2006, respectively (Figures 6C,F). A volume
of 0.5 mg/ml Exendin-4 improved the mRNA levels of sod-3
by 37.33% (p<0.01) and 63.33% (p<0.05) in CL4176 and
CL2006, respectively (Figures 6G,H). A volume of 0.5 mg/ml
Exendin-4 improved the activity of sod-3 by 36.04% (p<0.05)
and 27.11% (p<0.05) in CL4176 and CL2006, respectively
(Figures 6I,J).
We have a primary judgment that Exendin-4 is provided
with delaying the process of AD induced by Aβtoxicity through
its antioxidative stress activity.
DAF-16 was essential to the
neuroprotective eect of exendin-4
DAF-16, which is a major regulator in the insulin/insulin-
like growth factor 1 signaling (IIS) pathway, integrates
signals from upstream pathways to elicit transcriptional
changes in many genes involved in aging, development,
stress, metabolism, and immunity. Exendin-4 has a profound
effect on the insulin signaling pathway to prevent the
process of AD (Xu et al., 2015; Yang et al., 2016). Hence,
we investigated whether Exendin-4 can delay the process
of paralysis in worms when daf-16 was knocked down
using RNA interference (RNAi) (Supplementary Figure 2).
Exendin-4 at 0.5 mg/ml lost its ability to delay Aβ-induced
paralysis significantly when daf-16 of the worms was knockout
(Figure 7).
Meanwhile, we examined whether Exendin-4 affected the
translocation of DAF-16. The transgenic strain TJ356 can
stably express a DAF-16::GFP fusion protein. The indication of
fluorescence showed that Exendin-4 enhanced the translocation
of DAF-16 from the cytoplasm to nuclei. The subcellular
distribution includes “cytosol, “intermediate, and “nucleus
(Figures 8A–C). The amount of DAF-16 in nuclei was improved
obviously by estimating the fluorescence particles using the
ImageJ software (Figure 8D). The ratio of nucleus location was
increased from 15 to 45% compared with control worms.
However, Exendin-4 was still able to improve the mRNA
expression levels of the daf-16 gene (Figure 8E). These findings
indicated that DAF-16 was required for the protective effects of
Exendin-4 on Aβ(1-42) toxicity.
Frontiers in Aging Neuroscience 08 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
FIGURE 8
The eect of Exendin-4 on DAF-16. The subcellular distribution of DAF-16 was examined in approximately 20 worms for each group by
fluorescence microscopy. (A) Cytosol, (B) intermediate, (C) nucleus, and (D) the treated worms with 0.5 mg/ml Exendin-4 significantly increase
the ratio of nucleus translocation. The histogram represents the ratio of each subcellular distribution in the whole population in each group.
(E) The mRNA levels of daf-16 were analyzed by real-time PCR. mRNA levels was increased by 69.00% (p<0.001).
Exendin-4 regulates SOD via DAF-16
SOD-3 is a typical downstream target regulated by
DAF-16. To explore whether Exendin-4 regulates SOD via
DAF-16, SOD was detected in the activity and mRNA
levels after knocking down the gene of DAF-16 by RNAi.
We observed that the ability of Exendin-4 to regulate
SOD decreased in CL4176 worms with the DAF-16 gene
silenced. The activity of SOD and mRNA level of sod-
3 was downregulated by 30.45 and 43.13%, respectively
(Figures 9A,B). The CL4176 worms with empty vector L4440
were administrated with or without Exendin-4, and the same
trends as the above results in Figure 6 were displayed due to
the existence of DAF-16. All results indicated that Exendin-
4 regulated the SOD at least partially through a DAF-16-
based mechanism.
Discussion
Epidemiological studies showed that diabetes is a high-
risk factor for Alzheimer’s disease. Common pathophysiological
features exist between T2DM and AD, including oxidative stress
and insulin resistance. It suggests that effective drugs for T2DM
could provide an effective treatment option for AD. Exendin-4 is
a GLP-1R agonist approved to treat T2DM. Recently, Exendin-
4 has shown neuroprotective effects in vitro and in vivo model
systems (Wang et al., 2015, 2016; Qiu et al., 2016; Rocha-Ferreira
et al., 2018). However, the neuroprotective effect of Exendin-4
on AD has never been reported in the C. elegans models.
In this research, we investigated the neuroprotective effects
of Exendin-4 in C. elegans model of AD for the first
time. We found that Exendin-4 alleviated Aβ(1-42) toxicity
via DAF-16.
Frontiers in Aging Neuroscience 09 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
FIGURE 9
Exendin-4 regulates SOD via DAF-16. CL4176 worms with empty vector L4440 as control were treated with or without 0.5 mg/ml Exendin-4.
CL4176 worms were knocked down the daf-16 gene by RNAi and treated with 0.5 mg/ml Exendin-4. (A) The total SOD activity in worms was
investigated using the NBT method. Compared with the control group treated with Exendin-4, the SOD activity was decreased by 30.45% after
knocking down the daf-16 gene (p<0.01). (B) Compared with the control group treated with Exendin-4, the mRNA levels of sod-3 were
decreased by 43.13% after knocking down the daf-16 gene (p<0.05).
We utilized transgenic C. elegans strains CL4176 and
CL2006 induced expressing Aβ(1-42) to imitate the onset
of paralysis. Our results indicated that Exendin-4 attenuated
the process of paralysis and enhanced life span in a dose-
dependent manner.
So far, it has not been reported that Exendin-4 can inhibit
the onset of paralysis. In this research, we clarified that Exendin-
4 possesses the effect of inhibiting paralysis in C. elegans model
of AD for the first time.
However, it has also been reported that Exendin-4 possesses
the ability to increase life span in Huntington’s disease mice
(Martin et al., 2012). We have proved again that Exendin-
4 has the effect of prolonging life span in C. elegans
model of AD.
We hypothesized that exendin-4 can inhibit paralysis rates
or prolong the life of C. elegans due to its ability to attenuate Aβ
(1-42) toxicity. The content of Aβ(1-42) is the critical reason for
its toxicity.
In previous studies, Exendin-4 mainly focuses on the study
of protecting against cascading damage caused by Aβ(1-42),
such as inhibiting cell apoptosis (Qiu et al., 2016) and relieving
oxidative damage (An et al., 2019). Few studies were focused
on how Exendin-4 affects the change of Aβ(1-42) directly. Our
results of Western blotting and qPCR testified that Exendin-
4 attenuated the content of Aβ(1-42) on protein and mRNA
levels directly. Subsequently, Aβ(1-42) aggregation deposits
were reduced.
As we all know, ROS played a negative role in longevity
(Sanz, 2016; Dilberger et al., 2019; Huang et al., 2021) and
paralysis (Tang et al., 2022), and the aggregation of Aβ(1-42)
will lead to the accumulation of ROS, which will affect the onset
of paralysis and life span (Ewald, 2018; Kalmankar et al., 2021).
Our results showed that exendin-4 reduced ROS, which was
beneficial to prolong life and improve paralysis.
We traced the effect of SOD. We found that the SOD was
improved in its activity and mRNA level. It was a positive effect
on decreasing ROS content. This kind of effect is consistent with
previous studies that Exendin-4 upregulated the SOD on the
activity (Zhou et al., 2016) and gene expression in neuronal cells
(Hu et al., 2013; Yasuda et al., 2016).
The antioxidant effect of Exendin-4 has been described in
many oxidative stress models induced by different drugs in
vivo and in vitro. Exendin-4 may inhibit lipotoxicity-induced
oxidative stress in β-cells (Shen et al., 2020). Exendin-4 alleviates
oxidative stress by activating Nrf2/HO-1 in streptozotocin-
induced diabetic mice (Fang et al., 2019). This is the first study
to report that Exendin-4 alleviates the oxidative stress induced
by Aβ(1-42) in C. elegans.
As a known nuclear transcription factor, DAF-16 can induce
transcriptional regulation of many genes involved in aging,
development, and stress. SOD-3, as a downstream target gene,
is regulated by DAF-16. To study whether Exendin-4 regulates
SOD through DAF-16/FOXO, the nuclear translocation of
DAF-16 was detected in transgenic C. elegans TJ356 expressed
DAF-16::GFP. We found that Exendin-4 promoted the nuclear
translocation of DAF-16. The nuclear translocation of DAF-16
contributed to the upregulation of SOD-3, leading to the
antioxidant effect of Exendin-4. In contrast, worms knocked
down daf-16 gene were treated with Exendin-4. We found that
the ability of Exendin-4 to upregulate SOD was reduced.
Meanwhile, Exendin-4 lost the ability to alleviate the process
of paralysis after silencing the daf-16 gene. These results
Frontiers in Aging Neuroscience 10 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
indicated that DAF-16 played a pivotal role in alleviating Aβ
(1-42) toxicity when Exendin-4 exerted its neuroprotective
effects. It has been reported that Exendin-4 upregulated FoxO
1 expression both in vitro and in vivo (Wang et al., 2017), or
regulated the phosphorylation of FoxO 1 in cardiomyocytes (Eid
et al., 2021). So our results about the important roles of DAF-
16 in mediating the function of exendin-4 are consistent with
previous studies. We can draw a conclusion from the present
data that Exendin-4 has a positive effect on the FoxO family.
Conclusion
Although Exendin-4 was reported that possesses
neuroprotective effects in various AD models, we reported
for the first time that Exendin-4 exhibits the ability to delay
the development of paralysis and prolong the life span in the
C. elegans model of AD. In our research, we elucidated some
neuroprotective mechanisms of Exendin-4 on oxidative stress
and Aβ(1-42) toxicity. The DAF-16 is required for Exendin-4
to display the antioxidant effects and neuroprotective effects.
Taken together, our study testified that Exendin-4 as a marketed
diabetes drug has the potential to be a kind of drug for AD.
Data availability statement
The original contributions presented in the study are
included in the article/Supplementary material, further inquiries
can be directed to the corresponding authors.
Author contributions
XS and XW designed this study. YSun, ZW, and YSu
performed the experiments. YSu and ZW collected and analyzed
the data. XS wrote the manuscript. XW contributed to the
manuscript review. All authors contributed to the article and
approved the submitted version.
Funding
This study was supported by the Scientific and Technological
Developing Scheme of Jilin Province (20190304051YY) and
the Scientific Research Project of Jilin Provincial Department
of Education (JJKH20210879KJ) and the Natural Science
Foundation of Changchun Normal University 2018 (012).
Acknowledgments
We would like to thank Dr. Liping Wang and Dr. Shunwen
Guan for the C. elegans strains.
Conflict of interest
The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Publisher’s note
All claims expressed in this article are solely those
of the authors and do not necessarily represent those of
their affiliated organizations, or those of the publisher,
the editors and the reviewers. Any product that may
be evaluated in this article, or claim that may be made
by its manufacturer, is not guaranteed or endorsed
by the publisher.
Supplementary material
The Supplementary Material for this article can be
found online at: https://www.frontiersin.org/articles/10.3389/
fnagi.2022.955113/full#supplementary-material
References
An, J., Zhou, Y., Zhang, M., Xie, Y., Ke, S., Liu, L., et al. (2019).
Exenatide alleviates mitochondrial dysfunction and cognitive impairment in the
5xFAD mouse model of Alzheimer’s disease. Behav. Brain Res. 370, 111932.
doi: 10.1016/j.bbr.2019.111932
Butterfield, D. A., and Boyd-Kimball, D. (2004). Amyloid beta-peptide(1-42)
contributes to the oxidative stress and neurodegeneration found in Alzheimer
disease brain. Brain Pathol. 14, 426–432. doi: 10.1111/j.1750-3639.2004.tb00087.x
Caberlotto, L., Nguyen, T. P., Lauria, M., Priami, C., Rimondini, R., Maioli,
S., et al. (2019). Cross-disease analysis of Alzheimer’s disease and type-2 diabetes
highlights the role of autophagy in the pathophysiology of two highly comorbid
diseases. Sci. Rep. 9, 3965. doi: 10.1038/s41598-019-39828-5
Dilberger, B., Baumanns, S., Schmitt, F., Schmiedl, T., Hardt, M., Wenzel,
U., et al. (2019). Mitochondrial oxidative stress impairs energy metabolism and
reduces stress resistance and longevity of C. elegans. Oxid. Med. Cell Longev. 2019,
6840540. doi: 10.1155/2019/6840540
Dostal, V., and Link, C. D. (2010). Assaying beta-amyloid toxicity
using a transgenic C. elegans model. J. Vis. Exp. 44, 2252. doi: 10.37
91/2252
Eid, R. A., Bin-Meferij, M. M., El-Kott, A. F., Eleawa, S. M., Zaki, M.
S. A., Al-Shraim, M., et al. (2021). Exendin-4 Protects Against Myocardial
Ischemia-Reperfusion Injury by Upregulation of SIRT1 and SIRT3 and Activation
of AMPK. J. Cardiovasc. Transl. Res. 14, 619–635. doi: 10.1007/s12265-020-
09984-5
Ewald, C. Y. (2018). Redox Signaling of NADPH oxidases regulates
oxidative stress responses, immunity and aging. Antioxidants 7, 130.
doi: 10.3390/antiox7100130
Frontiers in Aging Neuroscience 11 frontiersin.org
Song et al. 10.3389/fnagi.2022.955113
Ewald, C. Y., and Li, C. (2010). Understanding the molecular basis of Alzheimer’s
disease using a Caenorhabditis elegans model system. Brain Struct. Funct. 214,
263–283. doi: 10.1007/s00429-009-0235-3
Fang, S., Cai, Y., Li, P., Wu, C., Zou, S., Zhang, Y., et al. (2019). [Exendin-
4 alleviates oxidative stress and liver fibrosis by activating Nrf2/HO-1 in
streptozotocin-induced diabetic mice]. Nan Fang Yi Ke Da Xue Xue Bao
39, 464–470. doi: 10.12122/j.issn.1673-4254.2019.04.13
Garabadu, D., and Verma, J. (2019). Exendin-4 attenuates brain mitochondrial
toxicity through PI3K/Akt-dependent pathway in amyloid beta (1-42)-induced
cognitive deficit rats. Neurochem. Int. 128, 39–49. doi: 10.1016/j.neuint.2019.04.006
Hu, G., Zhang, Y., Jiang, H., and Hu, X. (2013). Exendin-4 attenuates myocardial
ischemia and reperfusion injury by inhibiting high mobility group box 1 protein
expression. Cardiol. J. 20, 600–604. doi: 10.5603/CJ.2013.0159
Huang, Q., Li, R., Yi, T., Cong, F., Wang, D., Deng, Z., et al. (2021).
Phosphorothioate-DNA bacterial diet reduces the ROS levels in C.
elegans while improving locomotion and longevity. Commun. Biol. 4, 1335.
doi: 10.1038/s42003-021-02863-y
Kalmankar, N. V., Hari, H., Sowdhamini, R., and Venkatesan, R. (2021).
Disulfide-rich cyclic peptides from clitoria ternatea protect against beta-amyloid
toxicity and oxidative stress in transgenic Caenorhabditis elegans.J. Med. Chem.
64, 7422–7433. doi: 10.1021/acs.jmedchem.1c00033
Link, C. D. (2006). C. elegans models of age-associated neurodegenerativediseases:
lessons from transgenic worm models of Alzheimer’s disease. Exp. Gerontol. 41,
1007–1013. doi: 10.1016/j.exger.2006.06.059
Martin, B., Chadwick, W., Cong, W. N., Pantaleo, N., Daimon, C.
M., Golden, E. J., et al. (2012). Euglycemic agent-mediated hypothalamic
transcriptomic manipulation in the N171-82Q model of Huntington disease
is related to their physiological efficacy. J. Biol. Chem. 287, 31766–31782.
doi: 10.1074/jbc.M112.387316
Mullins, R. J., Mustapic, M., Chia, C. W., Carlson, O., Gulyani, S., Tran, J., et al.
(2019). A pilot study of exenatide actions in Alzheimer’s disease. Curr. Alzheimer
Res. 16, 741–752. doi: 10.2174/1567205016666190913155950
Qiu, C., Wang, Y. P., Pan, X. D., Liu, X. Y., Chen, Z., and Liu, L. B. (2016).
Exendin-4 protects Abeta(1-42) oligomer-induced PC12 cell apoptosis. Am. J.
Transl. Res. 8, 3540–3548.
Rangsinth, P., Prasansuklab, A., Duangjan, C., Gu, X., Meemon, K., Wink, M.,
et al. (2019). Leaf extract of Caesalpinia mimosoides enhances oxidative stress
resistance and prolongs lifespan in Caenorhabditis elegans. BMC Complement.
Altern. Med. 19, 164. doi: 10.1186/s12906-019-2578-5
Rocha-Ferreira, E., Poupon, L., Zelco, A., Leverin, A. L., Nair, S., Jonsdotter,
A., et al. (2018). Neuroprotective exendin-4 enhances hypothermia therapy
in a model of hypoxic-ischaemic encephalopathy. Brain 141, 2925–2942.
doi: 10.1093/brain/awy220
Sanz, A. (2016). Mitochondrial reactive oxygen species: do they extend
or shorten animal lifespan? Biochim. Biophys. Acta. 1857, 1116–1126.
doi: 10.1016/j.bbabio.2016.03.018
Sebastiao, I., Candeias, E., Santos, M. S., de Oliveira, C. R., Moreira, P. I., and
Duarte, A. I. (2014). Insulin as a bridge between type 2 diabetes and Alzheimer
Disease - how anti-diabetics could be a solution for dementia. Front. Endocrinol. 5,
110. doi: 10.3389/fendo.2014.00110
Shen, X., Luo, L., Yang, M., Lin, Y., Li, J., and Yang, L.
(2020). Exendin4 inhibits lipotoxicityinduced oxidative stress in
betacells by inhibiting the activation of TLR4/NFkappaB signaling
pathway. Int. J. Mol. Med. 45, 1237–1249. doi: 10.3892/ijmm.
2020.4490
Smith, J. V., and Luo, Y. (2003). Elevation of oxidative free radicals in Alzheimer’s
disease models can be attenuated by Ginkgo biloba extract EGb 761. J. Alzheimers
Dis. 5, 287–300. doi: 10.3233/JAD-2003-5404
Takahashi, A., Watanabe, T., Fujita, T., Hasegawa, T., Saito, M., and Suganuma,
M. (2014). Green tea aroma fraction reduces beta-amyloid peptide-induced toxicity
in Caenorhabditis elegans transfected with human beta-amyloid minigene. Biosci.
Biotechnol. Biochem. 78, 1206–1211. doi: 10.1080/09168451.2014.921553
Tang, X., Zhao, Y., Liu, Y., Liu, Y., Liu, Y., Niu, F., et al. (2022). 3,6’-disinapoyl
sucrose attenuates Abeta1-42 - induced neurotoxicity in Caenorhabditis elegans
by enhancing antioxidation and regulating autophagy. J. Cell Mol. Med. 26,
1024–1033. doi: 10.1111/jcmm.17153
Wang, A., Li, T., An, P., Yan, W., Zheng, H., Wang, B., et al. (2017). Exendin-
4 upregulates adiponectin level in adipocytes via Sirt1/Foxo-1 signaling pathway.
PLoS ONE 12, e0169469. doi: 10.1371/journal.pone.0169469
Wang, J., Ding, Y., Zhuang, L., Wang, Z., Xiao, W., and Zhu, J. (2021).
Ginkgolide binduced AMPK pathway activation protects astrocytes by regulating
endoplasmic reticulum stress, oxidative stress and energy metabolism induced by
Abeta142. Mol. Med. Rep. 23, 457. doi: 10.3892/mmr.2021.12096
Wang, X., Wang, L., Jiang, R., Xu, Y., Zhao, X., and Li, Y. (2016). Exendin-4
antagonizes Abeta1-42-induced attenuation of spatial learning and memory ability.
Exp. Ther. Med. 12, 2885–2892. doi: 10.3892/etm.2016.3742
Wang, X., Wang, L., Jiang, R., Yuan, Y., Yu, Q., and Li, Y. (2015). Exendin-
4 antagonizes Abeta1-42-induced suppression of long-term potentiation by
regulating intracellular calcium homeostasis in rat hippocampal neurons. Brain
Res. 1627, 101–108. doi: 10.1016/j.brainres.2015.09.015
Wolozin, B., Gabel, C., Ferree, A., Guillily, M., and Ebata, A. (2011). Watching
worms whither: modeling neurodegeneration in C. elegans.Prog. Mol. Biol. Transl.
Sci. 100, 499–514. doi: 10.1016/B978-0-12-384878-9.00015-7
Wu, Q., Qu, Y., Li, X., and Wang, D. (2012). Chromium exhibits adverse
effects at environmental relevant concentrations in chronic toxicity assay
system of nematode Caenorhabditis elegans.Chemosphere 87, 1281–1287.
doi: 10.1016/j.chemosphere.2012.01.035
Wu, Y., and Luo, Y. (2005). Transgenic C. elegans as a model in
Alzheimer’s research. Curr. Alzheimer Res. 2, 37–45. doi: 10.2174/1567205052
772768
Xu, W., Yang, Y., Yuan, G., Zhu, W., Ma, D., and Hu, S. (2015). Exendin-4, a
glucagon-like peptide-1 receptor agonist, reduces Alzheimer disease-associated tau
hyperphosphorylation in the hippocampus of rats with type 2 diabetes. J. Investig.
Med. 63, 267–272. doi: 10.1097/JIM.0000000000000129
Yang, Y., Ma, D., Xu, W., Chen, F., Du, T., Yue, W., et al. (2016). Exendin-
4 reduces tau hyperphosphorylation in type 2 diabetic rats via increasing
brain insulin level. Mol. Cell Neurosci. 70, 68–75. doi: 10.1016/j.mcn.2015.
10.005
Yasuda, H., Mizukami, K., Hayashi, M., Kamiya, T., Hara, H., and Adachi,
T. (2016). Exendin-4 promotes extracellular-superoxide dismutase expression
in A549 cells through DNA demethylation. J. Clin. Biochem. Nutr. 58, 34–39.
doi: 10.3164/jcbn.15-16
Zhang, X. G., Wang, X., Zhou, T. T., Wu, X. F., Peng, Y., Zhang, W.
Q., et al. (2016). Scorpion venom heat-resistant peptide protects transgenic
Caenorhabditis elegans from beta-amyloid toxicity. Front. Pharmacol. 7, 227.
doi: 10.3389/fphar.2016.00227
Zhou, Q., Wang, Y. P., Liu, X. Y., Liu, X. H., Pan, X. D., Chen, Z.,
et al. (2016). [Effects of exendin-4 on methylglyoxal-induced oxidative stress
in PC12 cells]. Zhongguo Ying Yong Sheng Li Xue Za Zhi 32, 426–430.
doi: 10.13459/j.cnki.cjap.2016.05.011
Frontiers in Aging Neuroscience 12 frontiersin.org
... For instance, flavonoids isolated from Sc ameliorate pelvic inflammation by promoting autophagic cell reprogramming via the NLRP3 inflammation-mediated autophagy pathway [6]. The methanol extract of Sc root has neuroprotective effects against excitatory poisoning and cerebral ischemic injury, as well as Aβ (25)(26)(27)(28)(29)(30)(31)(32)(33)(34)(35)-induced cortical nerve injury in rats [7,8]. Sc polyphenols can also be used to prevent obesity and related metabolic diseases in mice [9]. ...
... Wild-type N2 C. elegans was used for reproductive capacity and body length assay [25]. N2 hermaphrodite C. elegans specimens synchronized to L4 were randomly assigned to NGM plates coated with control (0.1% DMSO) or ScE (1, 10, 50 and 100 µg/mL) groups. ...
... C. elegans skn-1, which plays an important role in regulatory pathways and treatments to increase life span, is significantly homologous to Nrf2, a regulatory protein for antioxidant and xeno defense [33]. Daf-16 and skn-1 play necessary roles in lowering Aβ aggregation, and daf-16 regulates the insulin/insulin-like growth factor receptor signaling system (IIS), which has been linked to transcriptional alterations in genes related to aging, development, stress, metabolism, and immunity [25]. Hsf-1 reduces Aβ proteotoxicity in the AD model of C. elegans [34]. ...
Article
Full-text available
Smilax china L. (Chinaroot) is a natural herb that has multiple uses, such as being used to make tea and food. Both its roots and leaves have different uses due to their unique components. In this study, we analyzed the extract of S. china. roots using LC-HRMS and evaluated the neuroprotective effects and metabolic regulation of S. china on Caenorhabditis elegans. Chinaroot extract prolonged the life span of healthy nematodes, delayed the paralysis time of transgenic CL4176, and reduced the level of β-amyloid deposition in transgenic CL2006. The comprehensive analysis of metabolomics and qRT-PCR revealed that Chinaroot extract exerted neuroprotective effects through the valine, leucine and isoleucine degradation and fatty acid degradation pathways. Moreover, we first discovered that the expressions of T09B4.8, ech-7, and agxt-1 were linked to the neuroprotective effects of Chinaroot. The material exerted neuroprotective effects by modulating metabolic abnormalities in AD model C. elegans. Our study provides a new foundation for the development of functional food properties and functions.
... And oxidative stress occurs in the early stages of AD pathology, when the organism is exposed to a variety of harmful stimuli, the excessive production of ROS in the body leads to an imbalance in the oxidative and antioxidant systems (Shukla et al., 2011), which not only affects the mitochondrial energy metabolism, accelerates the phosphorylation of tau proteins (Sierra-Fonseca and Gosselink, 2018) and plays a role in neurotoxicity through the formation of neurofibrillary tangles and lowering of the excitability threshold, but also it also causes hyperoxidation of functional cell membrane proteins and lipids, destabilizing the cell membrane bilayer and contributing to the accumulation of Aβ protein, which in turn accelerates the generation of reactive oxygen species, induces neuronal cell damage and death, and leads to the occurrence of AD (Alafuzoff et al., 2008;Mohsenzadegan and Mirshafiey, 2012). At the same time, many studies have shown (Reddy et al., 2016;Feng et al., 2019;Zhou et al., 2019;Song et al., 2022) that effective inhibition of Aβ aggregation and improvement of the body's antioxidant level are important tools in the prevention and treatment of AD. ...
Article
Full-text available
Introduction Gastrodia elata is the dried tuber of the orchid Gastrodia elata Bl. It is considered a food consisting of a source of precious medicinal herbs, whose chemical composition is relatively rich. Gastrodia elata and its extracted fractions have been shown to have neuroprotective effects. P -hydroxybenzaldehyde ( p -HBA), as one of the main active components of Gastrodia elata , has anti-inflammatory, antioxidative stress, and cerebral protective effects, which has potential for the treatment of Alzheimer’s disease (AD). The aim of this study was to verify the role of p -HBA in AD treatment and to investigate its mechanism of action in depth based using the Caenorhabditis elegans ( C. elegans ) model. Methods In this study, we used paralysis, lifespan, behavioral and antistress experiments to investigate the effects of p -HBA on AD and aging. Furthermore, we performed reactive oxygen species (ROS) assay, thioflavin S staining, RNA-seq analysis, qPCR validation, PCR Array, and GFP reporter gene worm experiment to determine the anti-AD effects of p -HBA, as well as in-depth studies on its mechanisms. Results p -HBA was able to delay paralysis, improve mobility and resistance to stress, and delay aging in the AD nematode model. Further mechanistic studies showed that ROS and lipofuscin levels, Aβ aggregation, and toxicity were reduced after p -HBA treatment, suggesting that p -HBA ameliorated Aβ-induced toxicity by enhancing antioxidant and anti-aging activity and inhibiting Aβ aggregation. p -HBA had a therapeutic effect on AD by improving stress resistance, as indicated by the down-regulation of NLP-29 and UCR-11 expression and up-regulation of PQN-75 and LYS-3 expression. In addition, the gene microarray showed that p -HBA treatment played a positive role in genes related to AD, anti-aging, ribosomal protein pathway, and glucose metabolism, which were collectively involved in the anti-AD mechanism of p -HBA. Finally, we also found that p -HBA promoted nuclear localization of DAF-16 and increased the expression of SKN-1, SOD-3, and GST-4, which contributed significantly to inhibition of Aβ toxicity and enhancement of antioxidative stress. Conclusion Our work suggests that p -HBA has some antioxidant and anti-aging activities. It may be a viable candidate for the treatment and prevention of Alzheimer’s disease.
... In AD model animals, it has been shown that these events contribute to Aβ accumulation, which, in turn, enhances ROS production and promotes neuronal damage and death, leading to the occurrence of AD. Accordingly, the determination of ROS levels is crucial in the assessment of the effects of AD-targeting drugs (Song et al., 2022). Here, we used paraquat (5 mM) to induce an increase in ROS contents in AD model worms and found that the average ROS fluorescence intensity in the drug administration group was significantly reduced compared with that in the Control group (p < 0.01), indicating that 4,4′-methylenediphenol inhibits the production of free radicals in worms to a certain extent, thereby reducing Aβ-induced toxicity ( Figures 3E-G). ...
Article
Full-text available
Introduction Gastrodia elata Blume is a widely used medicinal and edible herb with a rich chemical composition. Moreover, prescriptions containing Gastrodia elata are commonly used for the prevention and treatment of cardiovascular, cerebrovascular, and aging-related diseases. Recent pharmacological studies have confirmed the antioxidant and neuroprotective effects of Gastrodia elata , and, in recent years, this herb has also been used in the treatment of Alzheimer’s disease (AD) and other neurodegenerative disorders. We have previously shown that 4,4′-methylenediphenol, a key active ingredient of Gastrodia elata , can mitigate amyloid-β (Aβ)-induced paralysis in AD model worms as well as prolong the lifespan of the animals, thus displaying potential as a treatment of AD. Methods We investigated the effects of 4,4′-methylenediphenol on AD and aging through paralysis, lifespan, and behavioral assays. In addition, we determined the anti-AD effects of 4,4′-methylenediphenol by reactive oxygen species (ROS) assay, lipofuscin analysis, thioflavin S staining, metabolomics analysis, GFP reporter gene worm assay, and RNA interference assay and conducted in-depth studies on its mechanism of action. Results 4,4′-Methylenediphenol not only delayed paralysis onset and senescence in the AD model worms but also enhanced their motility and stress tolerance. Meanwhile, 4,4′-methylenediphenol treatment also reduced the contents of reactive oxygen species (ROS) and lipofuscin, and decreased Aβ protein deposition in the worms. Broad-spectrum targeted metabolomic analysis showed that 4,4′-methylenediphenol administration had a positive effect on the metabolite profile of the worms. In addition, 4,4′-methylenediphenol promoted the nuclear translocation of DAF-16 and upregulated the expression of SKN-1, SOD-3, and GST-4 in the respective GFP reporter lines, accompanied by an enhancement of antioxidant activity and a reduction in Aβ toxicity; importantly, our results suggested that these effects of 4,4′-methylenediphenol were mediated, at least partly, via the activation of DAF-16. Conclusion We have demonstrated that 4,4′-methylenediphenol can reduce Aβ-induced toxicity in AD model worms, suggesting that it has potential for development as an anti-AD drug. Our findings provide ideas and references for further research into the anti-AD effects of Gastrodia elata and its active ingredients.
... In one study, researchers injected exendin-4 (EX-4), a GLP-1R analogue, into transgenic C. elegans and observed the amelioration of Aβ1-42 toxicity via an EX-4 antioxidant effect through DAF-16 as well as its reduction in Aβ1-42 expression and accumulation [107]. Another study demonstrated the neuroprotective and neurotrophic effects of EXE-4 in the brain of mice that had undergone mild traumatic brain injury [108]. ...
Article
Full-text available
Type 2 diabetes mellitus (T2DM) and Alzheimer’s disease (AD) are chronic, progressive disorders affecting the elderly, which fosters global healthcare concern with the growing aging population. Both T2DM and AD have been linked with increasing age, advanced glycosylation end products, obesity, and insulin resistance. Insulin resistance in the periphery is significant in the development of T2DM and it has been posited that insulin resistance in the brain plays a key role in AD pathogenesis, earning AD the name “type 3 diabetes”. These clinical and epidemiological links between AD and T2DM have become increasingly pronounced throughout the years, and serve as a means to investigate the effects of antidiabetic therapies in AD, such as metformin, intranasal insulin, incretins, DPP4 inhibitors, PPAR-γ agonists, SGLT2 inhibitors. The majority of these drugs have shown benefit in preclinical trials, and have shown some promising results in clinical trials, with the improvement of cognitive faculties in participants with mild cognitive impairment and AD. In this review, we have summarize the benefits, risks, and conflicting data that currently exist for diabetic drugs being repurposed for the treatment of AD.
... Exendin-4 is a polypeptide comprising 39 amino acids that is a long-acting glucagon-like peptide-1 receptor agonist. Many studies have shown that exenatide can play a neuroprotective role in a variety of diseases (45)(46)(47)(48). Therefore, we supposed that exendin-4 may also have a protective effect on the nerve cells of the inner ear. ...
Article
Full-text available
Objective(s) The protection of spiral ganglion neurons (SGNs) is crucial for hearing loss. Exendin-4 has been shown to have neuroprotective effects in several neurological disorders. Therefore, this study aimed to investigate the effect of the glucagon-like protein-1 receptor (GLP-1R) agonist exendin-4 on kanamycin-induced injury in mouse SGNs in vitro. Materials and Methods In this study, GLP-1R expression in SGNs was verified by immunofluorescence and immunohistochemical staining. In vitro-cultured SGNs and the organ of Corti were exposed to kanamycin with or without exendin-4 treatment. The cell survival rate was measured using the cell counting kit-8 assay, and the damage to auditory nerve fibers (ANF) projecting radially from the SGNs was evaluated using immunofluorescence staining. Reactive oxygen species (ROS) content was determined by flow cytometry, and glutathione peroxidase (GSH-Px) content, superoxide dismutase (SOD) activity, and malondialdehyde (MDA) content were determined by spectrophotometry. Protein expression of nuclear factor erythroid-2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) was detected using western blotting. Results GLP-1R was expressed in SGNs. Treatment with 1 mM kanamycin for 24 hr induced SGN damage. Exendin-4 (100 nM) had a protective effect against kanamycin-induced SGN cell injury, improved cell survival rate, reduced nerve fiber injury, increased SOD activity and GSH-Px level, and reduced MDA and ROS contents. The Nrf2/HO-1 pathway was activated. Conclusion Exendin-4 alleviates oxidative damage and exerts neuroprotective effects in kanamycin-induced SGN injury through the Nrf2/HO-1 signaling pathway. Exendin-4 has the potential to prevent or treat hearing loss due to SGN damage.
Article
The average age of the population is increasing worldwide, which has a profound impact on our society. This leads to an increasing demand for medicines and requires the development of new strategies to promote health during the additional years. In the search for resources and therapeutics for improved health during an extended life span, attention has to be paid to environmental exposure and ecosystem burdens that inevitably emerge with the extended consumption of medicines and drug development, even in the preclinical stage. The hereby introduced sustainable strategy for drug discovery is built on 3Rs, “Robustness, Reliability, and saving Resources”, inspired by both the 3Rs used in animal experiments and environmental protection, and centers on the usefulness and the variety of the small model organism Caenorhabditis elegans for detecting health-promoting natural products. A workflow encompassing a multilevel screening approach is presented to maximize the amount of information on health-promoting samples, while considering the 3Rs. A detailed, methodology- and praxis-oriented compilation and discussion of proposed C. elegans health span assays and more disease-specific assays are presented to offer guidance for scientists intending to work with C. elegans, thus facilitating the initial steps towards the integration of C. elegans assays in their laboratories.
Article
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by memory loss and cognitive impairment. β-amyloid (Aβ) is one of the typical pathological features of AD, and its accumulation leads to neuronal death from oxidative stress. Here, we found that hederagenin (HG), a natural product, exhibits anti-tumor, anti-inflammatory, anti-depressant, anti-neurodegenerative biological activities. However, whether HG has anti-Aβ activity remains unclear. Based on the characteristics of HG, it is hypothesized that HG has biological activity against Aβ injury. Therefore, Aβ-injured SH-SY5Y cells were constructed, and the protective effect of HG against Aβ injury was further evaluated using C. elegans. The results showed that HG increased superoxide dismutase activity, effectively reduced Aβ-induced oxidative damage, and reduced apoptosis via the PI3 K/Akt signaling pathway. HG inhibited Aβ deposition and delayed senescence and paralysis in the C. elegans strain, CL4176. HG showed inhibitory effects on Aβ; therefore, more natural active products are expected to be applied in AD therapy.
Article
Neurodegenerative disorders, which affect millions worldwide, are marked by a steady decline of neurons that are selectively susceptible. Due to the complex pathological processes underlying neurodegeneration, at present, there is no viable therapy available for neurodegenerative disorders. Consequently, the establishment of a novel therapeutic approach for such conditions is a clinical void that remains. The potential significance of various peptides as neuroprotective interventions for neurodegenerative disorders is gaining increasing attention. In the past few years, there has been growing scientific interest in glucagon‐like peptide‐1 receptor agonists due to their claimed neuroprotective effects. Exendin‐4 is a glucagon‐like peptide‐1 receptor agonist that is known to possess anti‐diabetic effects and does not degrade for hours, making it a superior candidate for such disorders. Moreover, exendin‐4's neuroprotective effects have been reported in several preclinical studies. Exendin‐4's diverse therapeutic targets suggest its potential therapeutic uses in neurodegenerative ailments like Alzheimer's disease and Parkinson's disease and have garnered an increasing amount of attention. Given the substantial body of evidence supporting the neuroprotective potential of exendin‐4 in various research models, this article is dedicated to exploring the promising role of exendin‐4 as a therapeutic agent for the treatment and management of Alzheimer's disease and Parkinson's disease. This review draws insights from the findings of numerous preclinical and clinical studies to highlight the collective neuroprotective advantages of exendin‐4 and the potential mechanisms that underlie its neuroprotective effects.
Article
Full-text available
The aggregation of β-amyloid (Aβ) has the neurotoxicity, which is thought to play critical role in the pathogenesis of Alzheimer's disease (AD). Inhibiting Aβ deposition and neurotoxicity has been considered as an important strategy for AD treatment. 3,6'-Disinapoyl sucrose (DISS), one of the oligosaccharide esters derived from traditional Chinese medicine Polygalae Radix, possesses antioxidative activity, neuroprotective effect and anti-depressive activity. This study was to explore whether DISS could attenuate the pathological changes of Aβ1-42 transgenic Caenorhabditis elegans (C. elegans). The results showed that DISS (5 and 50 μM) treatment significantly prolonged the life span, increased the number of egg-laying, reduced paralysis rate, decreased the levels of lipofuscin and ROS and attenuated Aβ deposition in Aβ1-42 transgenic C. elegans. Gene analysis showed that DISS could up-regulate the mRNA expression of sod-3, gst-4, daf-16, bec-1 and lgg-1, while down-regulate the mRNA expression of daf-2 and daf-15 in Aβ1-42 transgenic C. elegans. These results suggested that DISS has the protective effect against Aβ1-42 -induced pathological damages and prolongs the life span of C. elegans, which may be related to the reduction of Aβ deposition and neurotoxicity by regulating expression of genes related to antioxidation and autophagy.
Article
Full-text available
DNA phosphorothioation (PT) is widely distributed in the human gut microbiome. In this work, PT-diet effect on nematodes was studied with PT-bioengineering bacteria. We found that the ROS level decreased by about 20–50% and the age-related lipofuscin accumulation was reduced by 15–25%. Moreover, the PT-feeding worms were more active at all life periods, and more resistant to acute stressors. Intriguingly, their lifespans were prolonged by ~21.7%. Comparative RNA-seq analysis indicated that many gene expressions were dramatically regulated by PT-diet, such as cysteine-rich protein ( scl -11/12/13), sulfur-related enzyme ( cpr -2), longevity gene ( jnk -1) and stress response ( sod -3/5, gps -5/6, gst -18/20, hsp -12.8). Both the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis suggested that neuroactivity pathways were upregulated, while phosphoryl transfer and DNA-repair pathways were down-regulated in good-appetite young worms. The findings pave the way for pro-longevity of multicellular organisms by PT-bacterial interference.
Article
Full-text available
Ginkgolide B (GB), the diterpenoid lactone compound isolated from the extracts of Ginkgo biloba leaves, significantly improves cognitive impairment, but its potential pharmacological effect on astrocytes induced by β‑amyloid (Aβ)1‑42 remains to be elucidated. The present study aimed to investigate the protective effect and mechanism of GB on astrocytes with Aβ1‑42‑induced apoptosis in Alzheimer's disease (AD). Astrocytes obtained from Sprague Dawley rats were randomly divided into control, Aβ, GB and GB + compound C groups. Cell viability and apoptosis were analyzed using Cell Counting Kit‑8 and flow cytometry assays, respectively. Protein and mRNA expression levels were analyzed using western blotting and reverse transcription‑quantitative PCR, respectively. The levels of superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH‑Px), reactive oxygen species (ROS) and ATP were determined using the corresponding commercial kits. The findings revealed that GB attenuated Aβ1‑42‑induced apoptosis and the 5' adenosine monophosphate‑ activated protein kinase (AMPK) inhibitor compound C reversed the protective effects of GB. In addition, GB reversed Aβ1‑42‑induced oxidative damage and energy metabolism disorders, including decreases in the levels of SOD, GSH‑Px and ATP and increased the levels of MDA and ROS in astrocytes, while compound C reversed the anti‑oxidative effect and the involvement of GB in maintaining energy metabolism in astrocytes. Finally, GB decreased the expression levels of the endoplasmic reticulum stress (ERS) proteins and the apoptotic protein CHOP and increased both mRNA and protein expression of the components of the energy metabolism‑related AMPK/peroxisome proliferator‑activated receptor γ coactivator 1α/peroxisome proliferator‑activated receptor α and anti‑oxidation‑related nuclear respiratory factor 2/heme oxygenase 1/NAD(P)H dehydrogenase (quinone 1) pathways and downregulated the expression of β‑secretase 1. However, compound C could antagonize these effects. In conclusion, the findings demonstrated that GB protected against Aβ1‑42‑induced apoptosis by inhibiting ERS, oxidative stress, energy metabolism disorders and Aβ1‑42 production probably by activating AMPK signaling pathways. The findings provided an innovative insight into the treatment using GB as a therapeutic in Aβ1‑42‑related AD.
Article
Full-text available
This study evaluated if the cardioprotective effect of Exendin-4 against ischemia/reperfusion (I/R) injury in male rats involves modulation of AMPK and sirtuins. Adult male rats were divided into sham, sham + Exendin-4, I/R, I/R + Exendin-4, and I/R + Exendin-4 + EX-527, a sirt1 inhibitor. Exendin-4 reduced infarct size and preserved the function and structure of the left ventricles (LV) of I/R rats. It also inhibited oxidative stress and apoptosis and upregulated MnSOD and Bcl-2 in their infarcted myocardium. With no effect on SIRTs 2/6/7, Exendin-4 activated and upregulated mRNA and protein levels of SIRT1, increased levels of SIRT3 protein, activated AMPK, and reduced the acetylation of p53 and PGC-1α as well as the phosphorylation of FOXO-1. EX-527 completely abolished all beneficial effects of Exendin-4 in I/R-induced rats. In conclusion, Exendin-4 cardioprotective effect against I/R involves activation of SIRT1 and SIRT3. Exendin-4 could scavenge free radical directly, upregulate p53, and through upregulation of SIRT1 and stimulating SIRT1 nuclear accumulation. In addition, Exendin-4 also upregulates SIRT3 which plays an essential role in the upregulation of antioxidants, inhibition of reactive oxygen species (ROS) generation, and prevention of mitochondria damage. Accordingly, SIRT1 induces the deacetylation of PGC-1α and p53 and is able to bind p-FOXO-1. This results in inhibition of cardiomyocyte apoptosis through increasing Bcl-2 levels, activity, and levels of MnSOD; decreasing expression of Bax; decreasing cytochrome C release; and improving mitochondria biogenesis through upregulation of Mfn-2.
Article
Full-text available
Introduction: Mitochondria supply cellular energy and are key regulators of intrinsic cell death and consequently affect longevity. The nematode Caenorhabditis elegans is frequently used for lifespan assays. Using paraquat (PQ) as a generator of reactive oxygen species, we here describe its effects on the acceleration of aging and the associated dysfunctions at the level of mitochondria. Methods: Nematodes were incubated with various concentrations of paraquat in a heat-stress resistance assay (37°C) using nucleic staining. The most effective concentration was validated under physiological conditions, and chemotaxis was assayed. Mitochondrial membrane potential (ΔΨm) was measured using rhodamine 123, and activity of respiratory chain complexes determined using a Clark-type electrode in isolated mitochondria. Energetic metabolites in the form of pyruvate, lactate, and ATP were determined using commercial kits. Mitochondrial integrity and structure was investigated using transmission electron microscopy. Live imaging after staining with fluorescent dyes was used to measure mitochondrial and cytosolic ROS. Expression of longevity- and mitogenesis-related genes were evaluated using qRT-PCR. Results: PQ (5 mM) significantly increased ROS formation in nematodes and reduced the chemotaxis, the physiological lifespan, and the survival in assays for heat-stress resistance. The number of fragmented mitochondria significantly increased. The ∆Ψm, the activities of complexes I-IV of the mitochondrial respiratory chain, and the levels of pyruvate and lactate were significantly reduced, whereas ATP production was not affected. Transcript levels of genetic marker genes, atfs-1, atp-2, skn-1, and sir-2.1, were significantly upregulated after PQ incubation, which implicates a close connection between mitochondrial dysfunction and oxidative stress response. Expression levels of aak-2 and daf-16 were unchanged. Conclusion: Using paraquat as a stressor, we here describe the association of oxidative stress, restricted energy metabolism, and reduced stress resistance and longevity in the nematode Caenorhabditis elegans making it a readily accessible in vivo model for mitochondrial dysfunction.
Article
Full-text available
Background: Caesalpinia mimosoides, a vegetable consumed in Thailand, has been reported to exhibit in vitro antioxidant properties. The in vivo antioxidant and anti-aging activities have not been investigated. The aim of this research was to study the antioxidant activity of C. mimosoides extracts in Caenorhabditis elegans, a widely used model organism in this context. Methods: C. elegans were treated with C. mimosoides extracts in a various concentrations. To investigate the protective effects of the extract against oxidative stress, wild-type N2 were used to determine survival rate under oxidative stress and intracellular ROS. To study underlying mechanisms, the mutant strains with GFP reporter gene including TJ356, CF1553, EU1 and LD4 were used to study DAF-16, SOD-3, SKN-1 and GST-4 gene, respectively. Lifespan and aging pigment of the worms were also investigated. Results: A leaf extract of C. mimosoides improved resistance to oxidative stress and reduced intracellular ROS accumulation in nematodes. The antioxidant effects were mediated through the DAF-16/FOXO pathway and SOD-3 expression, whereas the expression of SKN-1 and GST-4 were not altered. The extract also prolonged lifespan and decreased aging pigments, while the body length and brood size of the worms were not affected by the extract, indicating low toxicity and excluding dietary restriction. Conclusions: The results of this study establish the antioxidant activity of C. mimosoides extract in vivo and suggest its potential as a dietary supplement and alternative medicine to defend against oxidative stress and aging, which should be investigated in intervention studies.
Article
The present study aimed to investigate the relationship between the protective effects of exendin‑4 (EX‑4) on lipotoxicity‑induced oxidative stress and meta‑inflammation in β‑cells and the toll‑like receptor 4 (TLR4)/NF‑κB signaling pathway. Lipotoxicity, hydrogen peroxide (H2O2)‑induced oxidative stress in β cells, obese Sprague Dawley rats and TLR4 truncation rats were utilized in the present study. The expression levels were detected by western blotting; cell apoptosis was detected by TUNEL assay; and the intracellular reactive oxygen species (ROS) levels were analyzed using a ROS assay kit. The findings of the present study showed that EX‑4 inhibited the expression of TLR4, NF‑κB p65 subunit and p47phox in a concentration‑dependent manner, and decreased the intracellular level of ROS. Additionally, silencing of TLR4 expression enhanced the protective effects of EX‑4, while overexpression of TLR4 attenuated these protective influences. Simultaneously, it was demonstrated that TLR4 was involved in the process of EX‑4 intervention to inhibit H2O2‑induced oxidative stress in islet β‑cells. Moreover, it was found that EX‑4 also inhibited TLR4‑ or NF‑κB agonist‑induced oxidative stress. These results were also confirmed in an animal model of obese rats, in which EX‑4 was able to improve the function of β‑cells, attenuate oxidative stress, and inhibit the expression levels of TLR4 and NF‑κB p65 subunit in the pancreas of the diet‑induced obese rats. Furthermore, truncation of the TLR4 gene in SD rats delayed the aforementioned damage. In summary, EX‑4 may inhibit lipotoxicity‑induced oxidative stress in β‑cells by inhibiting the activation of the TLR4/NF‑κB signaling pathway.
Article
Background Strong preclinical evidence suggests that exenatide, a glucagon-like peptide-1 (GLP-1) receptor agonist used for treating type 2 diabetes, is neuroprotective and disease- modifying in Alzheimer’s disease (AD). Objective We performed an 18-month double-blind randomized placebo-controlled Phase II clinical trial to assess the safety and tolerability of exenatide and explore treatment responses for clinical, cognitive, and bio- marker outcomes in early AD. Method Eighteen participants with high probability AD based on cerebrospinal fluid (CSF) biomarkers completed the entire study prior to its early termination by the sponsor; partial outcomes were available for twenty-one. Results Exenatide was safe and well-tolerated, showing an expectedly higher incidence of nausea and decreased appetite compared to placebo and decreasing glucose and GLP-1 during Oral Glucose Tolerance Tests. Exenatide treatment produced no differences or trends compared to placebo for clinical and cognitive measures, MRI cortical thickness and volume, or biomarkers in CSF, plasma, and plasma neuronal extracellular vesicles (EV) except for a reduction of Aβ42 in EVs. Conclusion The positive finding of lower EV Aβ42 supports emerging evidence that plasma neuronal EVs provide an effective platform for demonstrating biomarker responses in clinical trials in AD. The study was underpowered due to early termination and therefore we cannot draw any firm conclusions. However, the analysis of secondary outcomes shows no trends in support of the hypothesis that exenatide is disease- modifying in clinical AD, and lowering EV Aβ42 in and of itself may not improve cognitive outcomes in AD.
Article
The role of mitochondrial dysfunction has been well-documented in Alzheimer's disease (AD). Glucagon-like peptide 1 (GLP-1) receptor agonists are being utilized as neuroprotectants in the treatment of various neurological disorders, including AD. We conducted this study to explore the effects of exenatide (a GLP-1 receptor agonist) on β-amyloid plaque (Aβ)-induced cognitive impairment and mitochondrial dysfunction in 5xFAD transgenic mice. Spatial memory test showed that exenatide administration (100 μg/kg twice per day) prevented cognitive decline after 16 weeks of treatment. Aβ 1-42 deposition and synapse damage in the hippocampus was significantly alleviated. Furthermore, exenatide treatment can improve mitochondrial morphology, relieve oxidative damage, correct mitochondrial energy crisis, and normalize mitochondrial dynamics. These findings suggest that exenatide, which has already been applied in clinical medicine, may be a promising agent for AD therapy via mitochondrial protection.