You are on page 1of 11

Neuroscience Letters 760 (2021) 136094

Contents lists available at ScienceDirect

Neuroscience Letters
journal homepage: www.elsevier.com/locate/neulet

Research article

microRNA-106b-containing extracellular vesicles affect autophagy of


neurons by regulating CDKN2B in Parkinson’s disease
Xue Bai *, Qiaoyun Dong , Li Zhao , Yan Yao , Bo Wang
Department Five of Neurology, Cangzhou Central Hospital, Cangzhou 061000, Hebei, PR China

A R T I C L E I N F O A B S T R A C T

Keywords: Parkinson’s disease (PD) is the second most frequent neurodegenerative disorder, and autophagy dysfunction is
Parkinson’s disease involved in the pathogenesis of PD. Mesenchymal stem cells (MSC)-derived extracellular vesicles (EVs) have been
Mesenchymal stem cell-derived extracellular established as an attractive therapeutic tool, since they can serve as biological nanoparticles with beneficial
vesicles
effects in PD. Herein, the study aimed to investigate the effects of EVs derived microRNA (miR)-106b on
microRNA-106b
autophagy of neurons in PD. Following the development of a mouse model of PD, we conducted behavior test,
CDKN2B
Autophagy TUNEL assay and HE staining to verify the success of modeling. Afterward, MSC-derived EVs were extracted and
Neuron identified. In hippocampal tissues and neurons of PD mice, miR-106b was poorly expressed, while CDKN2B was
highly expressed. miR-106b shuttled by MSC-derived EVs increased neuronal survival, autophagy, LC3II/LC3I
ratio and Bcl-2 protein expression, while inhibited neuronal apoptosis and Bax expression in PD mice. It was also
confirmed that CDKN2B is a downstream target of miR-106b. Overexpression of CDKN2B reversed the protective
effects of miR-106b-containing EVs on neurons in mice with PD. Collectively, miR-106b-containing EVs alleviate
neuronal apoptosis and enhance neuronal autophagy in PD by downregulating CDKN2B.

1. Introduction physiology of neuronal circuits by delivering proteins, RNA, lipids, and


metabolites, are involved in the development of neurodegenerative
Parkinson’s disease (PD), a neurological disorder, is characterized by diseases, including PD [7]. EVs, from specific cell types, can alleviate
the motor features of parkinsonism related to Lewy bodies and loss of some diseases by regulating autophagy [8]. Moreover, EVs can mediate
dopaminergic neurons in the substantia nigra [1]. PD is the second most the pathogenesis of PD, such as the dysfunction of autophagy and
prevalent neurodegenerative disease globally, disturbing more than 10 lysosomal degradation [9]. Mesenchymal stem cell (MSC)-derived EVs
million people, about 1% of the global population over 60 years old [2]. may be applied as a possible therapeutic approach or as an adjuvant
The exposure to pesticides, consumption of dairy products, history of therapy to halt the disease progression and improve PD symptoms [10].
melanoma, as well as traumatic brain injury pose the risk factors of PD In addition, the activation of autophagy stimulated by human umbilical
[3]. Despite decades of research dedicated to developing therapy op­ cord MSC (hUCMSC)-derived EVs can efficiently reduce the nephro­
tions, multiple agents have failed in clinic, and the task of improving PD toxicity of cisplatin [11]. Therefore, we assumed that hUCMSC-derived
seems increasingly daunting as the intricacies of the genotype, pheno­ EVs could be related to autophagy of neurons in PD as well. Interest­
type, and pathogenesis of PD continue to emerge [4]. Autophagy, a ingly, homo sapiens-microRNA (miR)-106b-3p was found as one of the
multifaceted intracellular process, exists to identify and capture a large five miRNAs that were downregulated in plasma EVs from PD patients
repertoire of intracellular components and bring them to the lysosomal than that in controls, indicating that it might be a possible biomarker for
compartment [5]. Autophagy, which is suppressed in aging cells, could the diagnosis of PD [12]. The close interaction between miR-106b and
protect the cells against aging and disease [6]. Therefore, detailed illu­ autophagy has been appreciated in inflammatory bowel disease [13].
mination of the mechanism underlying autophagy has great signifi­ Hence, we hypothesized that EVs released from hUCMSC, as carriers,
cances for exploring effective therapies against PD. could effectively deliver miR-106b into the neurons to regulate auto­
Extracellular vesicles (EVs), having the competence to modify the phagy in PD. All efforts made in the present study were to offer a new

* Corresponding author at: Department Five of Neurology, Cangzhou Central Hospital, No. 50, Xinhua West Road, Yunhe District, Cangzhou 061000, Hebei, PR
China.
E-mail address: baixue5191@126.com (X. Bai).

https://doi.org/10.1016/j.neulet.2021.136094
Received 12 May 2021; Received in revised form 24 June 2021; Accepted 26 June 2021
Available online 30 June 2021
0304-3940/© 2021 Elsevier B.V. All rights reserved.
X. Bai et al. Neuroscience Letters 760 (2021) 136094

idea for the discovery of new strategies for the treatment of PD. multiplicity of infection = 5, and the 6-well plate was incubated in an
incubator at 37 ◦ C with 5% CO2. After 24 h, the cells were then collected,
2. Materials and methods and the cell proliferation and apoptosis and the expression level of
autophagy-related protein LC3 were detected by cell counting kit-8
2.1. Ethics statement (CCK-8), flow cytometry, and immunofluorescence, respectively
(described in detail in the Supplementary File).
The experiments regarding animals were performed as per the pro­
tocols and guidelines and were approved by the Ethics Committee of 2.4. Isolation and identification of hUCMSC-derived EVs
Cangzhou Central Hospital. Best efforts were made to minimize the
suffering of the included animals. hUCMSCs (HUXUC-01001, Cyagen, Suzhou, Jiangsu, China) were
cultured with DMEM containing 10% FBS, 100 U/mL penicillin and 100
2.2. Establishment and treatment of the PD mouse model μg/mL streptomycin for 48 h in six-well plates at 37 ◦ C in 5% CO2. All
experiments were performed using MSC at passage 3–8.
Forty-five male healthy C57BL mice (8 weeks old, 18–22 g) were MSCs were cultured in FBS-free medium and the supernatant was
from the Laboratory Animal Center of Sun Yat-sen University centrifuged sequentially (CP100WX/CR22N Hitachi Tokyo Japan) at
(Guangzhou, Guangdong, China). The animals were kept on in a 300 g for 10 min at 2000 g for 20 min and at 10,000 g for 30 min. After
specific-pathogen-free environment at 24–26 ◦ C with 45–55% humidity. each centrifugation step the precipitate was removed. The EVs were then
The feed and drinking water were sterilized at high temperature before precipitated by ultracentrifugation of the supernatant (70 min at
use. 100,000 g). The precipitate was resuspended in PBS. The EVs were
Ten mice were randomly selected as the control group and 35 mice passed through a 0.22 μm filter and stored at − 20 ◦ C until use. The
were used for PD model generation using an intraperitoneal injection of identification method of the extracted EVs is shown in the Supplemen­
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP) [14]. MPTP tary File
(Sigma-Aldrich Chemical Company, St Louis, MO, USA) was dissolved in
sterile saline and injected intraperitoneally into the mice at a dose of 25 2.5. Reverse transcription-quantitative polymerase chain reaction (RT-
mg/kg once daily for 7 d. Control mice were injected with equal qPCR) and western blot
amounts of saline.
After 25 d of modeling, the mice were anesthetized by intraperito­ We used RT-qPCR and western blot to detect the expression of the
neal injection of 2% sodium pentobarbital (60 mg/kg), and the skull was relevant genes in cells and tissues (shown in the Supplementary File).
exposed by a longitudinal median incision after alcohol disinfection of
the skin on the top of the head. A small hole was drilled with a 5-gauge 2.6. Dual-luciferase reporter assay
needle (1 mm from bregma and 2 mm next to the midline). A micro­
injector was inserted vertically to a depth of 2.15 mm, and 10 μL len­ Starbase (http://starbase.sysu.edu.cn/) was utilized to predict miR-
tiviruses (titer 2 × 108 U/mL) or 5 μL phosphate-buffered saline (PBS) 106b binding sites to CDKN2B, and dual-luciferase assays were per­
containing 50 μg human umbilical cord MSC (hUCMSC)-EVs was slowly formed to verify whether CDKN2B is a target of miR-106b. The CDKN2B
injected. The needle was left in place for 5 min after injection. Ten PD 3′ UTR with binding sites were artificially synthesized and cloned into
mice were left untreated, and the remaining 25 PD mice were randomly pmirGLO vector (Promega, Madison, WI, USA) using endonuclease sites.
categorized into five groups (n = 5): PD + EVs group (PD mice injected The complementary sequence mutation (MUT) of the seed sequence was
with 50 μg hUCMSC-EVs suspended in 5 μL PBS), PD + EVs-NC group designed on the CDKN2B wild type (WT), and the target fragment was
(PD mice injected with 50 μg hUCMSC-EVs-mimic NC suspended in 5 μL subcloned into the pGL3-control vector by restriction endonuclease
PBS), PD + EVs-miR-106b group (PD mice injected with 50 μg hUCMSC- digestion using T4 DNA ligase. The 293 T cells were co-transfected with
EVs-miR-106b mimic suspended in 5 μL PBS), PD + EVs-miR-106b + oe- correctly sequenced luciferase reporter plasmids WT or MUT and miR-
NC group (PD mice injected with 50 μg hUCMSC-EVs-miR-106b sus­ 106b mimic, respectively. The cells were lysed 48 h after transfection,
pended in 5 μL PBS and control lentivirus), and PD + EVs-miR-106b + and luciferase activity was measured on a Luminometer TD-20/20
oe-CDKN2B group (PD mice injected with 50 μg hUCMSC-EVs-miR-106b analyzer (model: E5311, Promega) using the Dual-Luciferase Reporter
suspended in 5 μL PBS and CDKN2B overexpressing lentivirus). After 2 Assay System kit (Promega).
d, the pole test and swimming test were performed (as described in
Supplementary File). After the above experiments were completed, the 2.7. Data analysis
mice were euthanized by intraperitoneal injection of excessive sodium
pentobarbital, and the brain tissues were harvested for subsequent ex­ SPSS 21.0 software (IBM Corp. Armonk, N.Y., USA) was applied for
periments. Pathologically damaged structures in mouse hippocampal the analysis of the data. The results are expressed as the means ± SD. All
tissues and the proportion of apoptosis were evaluated using the experiments were repeated at least three times. Comparisons be­
hematoxylin-eosin (HE) staining and Terminal deoxynucleotidyl trans­ tween every two groups were analyzed using an unpaired t-test, while
ferase (TdT)-mediated 2′ -Deoxyuridine 5′ -Triphosphate (dUTP) nick comparisons among three groups were analyzed by one-way ANOVA.
end labeling (TUNEL) (as described in the Supplementary File). The post hoc test was conducted using the Tukey’s in multiple com­
parisons. Statistical significance was defined as p < 0.05.
2.3. Treatment of primary mouse hippocampal neurons
3. Results
Mouse primary hippocampal neurons were isolated as described in
the Supplementary File. The neurons were treated with 500 μg hUCMSC- 3.1. miR-106b expresses poorly in PD mice
derived EVs suspended in 500 μL PBS or 500 μg EVs derived from
hUCMSC transfected with miR-106b mimic in 500 μL PBS. The neurons PD miRNA dataset GSE16658 from the GEO database was inquired
were infected with GV287 (Genechem, Shanghai, China) overexpression for analysis, and we found 49 significantly upregulated miRNAs and 19
lentiviral vectors. Normal neurons were also infected with lentiviruses significantly downregulated miRNAs (Fig. 1A). The heat map shows the
harboring mimic NC (5′ -UUCUCCGAACGUGUCACGUTT-3′ ) or miR- top 15 differentially expressed miRNAs (Fig. 1B), of which miR-106 is
106b (5′ -TAAAGTGCTGACAGTGCAGAT-3′ ). An appropriate amount of the most pronounced one. For validation, we induced a PD mouse model
lentivirus was supplemented to the cell culture plate according to the using MPTP and conducted RT-qPCR to detect the miR-106b expression

2
X. Bai et al. Neuroscience Letters 760 (2021) 136094

Fig. 1. miR-106b is poorly expressed in PD mice. A, 68 differentially expressed miRNAs in GSE16658 dataset in the GEO database; B, the first 15 differentially
expressed miRNAs in GSE16658 dataset where horizontal coordinates indicate sample numbers, vertical coordinates indicate gene names, each small square in the
figure indicates the expression of a gene in a sample, and the histogram on the top right is the color scale; C, miR-106b expression in hippocampal tissues of control
and PD mice by RT-qPCR. *p < 0.05.

in the hippocampal tissues of PD mice. The results showed that miR- 3.3. EVs-miR-106b promotes autophagy and inhibits neuronal apoptosis
106b expression in the hippocampal tissues of PD mice was signifi­ in PD
cantly lower compared with the control mice (Fig. 1C).
Neurons from mice in the control and PD groups were extracted, and
3.2. Identification of EVs-miR-106b the expression of MAP-2 protein, a marker of primary neurons, was
detected by immunofluorescence. The results showed that the positive
We isolated MSC-derived EVs for identification, and TEM observa­ expression rate of MAP-2 in the primary neurons extracted from the
tion showed that the EVs released by MSC showed a cup-shaped or control and PD mice reached more than 95%, indicating that the
spherical morphology (Fig. 2A). NTA analysis exhibited that EVs were extracted neurons were of high purity and could be used for subsequent
40–120 nm in diameter (Fig. 2B), and Western blot demonstrated that all experiments (Fig. 3A). Neurons in PD mice were treated with MSC-
EVs from MSC expressed CD63, CD81 and TSG101 and did not express derived EVs or EVs-miR-106b. CCK8 assay and flow cytometry exhibi­
Calnexin (Fig. 2C). After transfection of miR-106b mimic in MSC ted that (Fig. 3B, C): compared with normal neurons, PD neuronal
(Fig. 2D), EVs released from MSC also showed high miR-106b expression viability was decreased and apoptosis was significantly increased;
(Fig. 2E). relative to the PD + EVs group, neuronal survival was increased and
apoptosis was significantly decreased in the PD + EVs-miR-106b group.
Immunofluorescence detection showed that neuronal autophagy was
reduced in the PD group compared with the normal group, and over­
expression of miR-106b in EVs facilitated the neuronal autophagy

3
X. Bai et al. Neuroscience Letters 760 (2021) 136094

Fig. 2. Identification of hUCMSC-EVs-miR-106b. A, observation of EVs morphology by TEM (bar = 100 nm); B, the size distribution of EVs determined by NTA; C,
detection of EVs-specific surface marker proteins determined by Western blot; D, miR-106b expression in MSCs after transfection with miR-106b mimic by RT-qPCR;
E, expression of miR-106b in MSC-EVs detected by RT-qPCR after transfection of MSCs with miR-106b mimic. All the experiments were repeated at least three times.
*#p < 0.05.

(Fig. 3D). Western blot results in Fig. 3E revealed that compared with 3.4. EVs-miR-106b promotes neuronal autophagy and alleviates neuronal
the normal neurons, the LC3II/LC3I ratio and Bcl-2 expression were damage in PD mice
reduced, while Bax protein expression was enhanced in PD neurons. By
contrast, compared with the EVs treatment, LC3II/LC3I ratio and Bcl-2 The mice were injected with EVs or EVs overexpressing miR-106b
expression were increased, whereas Bax protein expression was after successful PD modeling. As shown in Fig. 4A, B, mice in the PD
decreased in PD neurons treated with EVs overexpressing miR-106b. group spent significantly more time in the pole test and had significantly

4
X. Bai et al. Neuroscience Letters 760 (2021) 136094

Fig. 3. EVs-miR-106b promotes autophagy and inhibits apoptosis in PD neurons. A, immunofluorescence detection of MAP-2 protein expression in cells; B, neuronal
activity determined by CCK8 assay; C, detection of apoptosis rate of neurons tested by flow cytometry; D, LC3 protein expression in neurons examined by immu­
nofluorescence detection; E, the expression of autophagy-related proteins LC3II and LC3I, apoptosis-related proteins Bcl-2 and Bax in each group of neurons
examined by western blot. *#p < 0.05.

5
X. Bai et al. Neuroscience Letters 760 (2021) 136094

A Control B Control
PD PD
PD + EVs PD + EVs
30 PD + EVs-NC 4 PD + EVs-NC
PD + EVs-miR-106b PD + EVs-miR-106b
The total time (s)

20
* 3

Score
# #
2

10
1
*

0 0
C

50 μm 50 μm 50 μm

Control PD PD + EVs

50 μm 50 μm

PD + EVs-NC PD + EVs-miR-106b
D Control
PD
PD + EVs
50 PD + EVs-NC
PD + EVs-miR-106b
Apoptosis rate (%)

40

30
*
20 #

10

0
Fig. 4. EVs-miR-106b affects behavioral changes and alleviates neuronal damage in PD mice. A, the time spent on pole climbing by each group of mice; B, the score
of each group of mice examined by swimming test; C, histopathological changes of hippocampal neurons in the brain of mice were observed by HE staining; D,
apoptosis of hippocampal neurons in mice examined by TUNEL assay. *#p < 0.05.

6
X. Bai et al. Neuroscience Letters 760 (2021) 136094

lower scores in the swimming test versus control mice. Compared with of control mice, while there was an increase in hippocampal tan positive
the PD + EVs group, mice in the PD + EVs-miR-106b group spent cells in PD group, PD + EVs group, and PD + EVs-miR-106b group.
significantly less time in the pole test and had higher scores in the
swimming test. The results of HE staining (Fig. 4C) showed that the
hippocampal neurons in control mice were evenly distributed with clear 3.5. CDKN2B is a target gene of miR-106b
nuclei structure; some neurons in PD and PD + EVs groups were swollen
with interstitial edema and shriveled nucleus. However, the hippo­ To investigate the mechanism of miR-106b affecting PD, Starbase
campal tissues in the PD + EVs-miR-106b group had a more intact website demonstrated that CDKN2B shares a binding relation with miR-
structure and clearer nuclei results. The TUNEL assay in Fig. 4D showed 106b (Fig. 5A). Therefore, we speculated that miR-106b may influence
that few tan positive granular cells were seen in the hippocampal tissues PD progression by regulating the CDKN2B expression.
Dual-luciferase assay revealed that the fluorescence intensity of cells

Fig. 5. CDKN2B is a target gene of miR-106b. A, CDKN2B shares a specific binding relation to miR-106b predicted by starbase website; B, the target binding
relationship between miR-106b and CDKN2B verified by dual-luciferase assay; C, miR-106b and CDKN2B mRNA expression in each group of neurons by RT-qPCR; D,
CDKN2B protein expression in neurons in response to miR-106b mimic or mimic NC examined by western blot; E, CDKN2B protein expression in PD neurons co-
cultured with EVs-miR-106b mimic by western blot. *#p < 0.05.

7
X. Bai et al. Neuroscience Letters 760 (2021) 136094

transfected with miR-106b mimic + CDKN2B-WT was significantly downregulated after miR-106b mimic treatment. Western blot was
decreased compared with those transfected with mimic-NC + CDKN2B- implemented to detect CDKN2B expression in neurons after PD
WT (Fig. 5B). In addition, miR-106b mimic was transfected into modeling, and the results showed that CDKN2B expression was highly
extracted normal neurons, and miR-106b and CDKN2B mRNA expres­ expressed in neurons upon PD modeling, and was significantly reduced
sion in cells were detected using RT-qPCR and CDKN2B levels were after EVs-miR-106b treatment (Fig. 5E).
detected by Western blot, and the results showed (Fig. 5C, D): compared
with the mimic NC treatment, miR-106b expression was upregulated
and CDKN2B mRNA and protein levels were significantly

Fig. 6. EVs-miR-106b inhibits CDKN2B to promote neuronal autophagy and suppress neuronal apoptosis in PD mice. Neurons extracted from PD mice treated with
EVs-miR-106b + oe-NC or EVs-miR-106b + oe-CDKN2B. A, miR-106b and CDKN2B mRNA expression in neurons by RT-qPCR; B, CDKN2B protein expression in PD
neurons by western blot; C, neuronal activity determined by CCK8 assay; D, detection of apoptosis rate of neurons tested by flow cytometry; E, LC3 protein expression
in neurons examined by immunofluorescence detection; F, the expression of autophagy-related proteins LC3II and LC3I, apoptosis-related proteins Bcl-2 and Bax in
each group of neurons examined by western blot. *p < 0.05.

8
X. Bai et al. Neuroscience Letters 760 (2021) 136094

3.6. EVs-miR-106b inhibits CDKN2B to promote neuronal autophagy and ratio and Bcl-2 protein were decreased and Bax protein expression was
suppress neuronal apoptosis in PD mice increased in the PD + EVs-miR-106b + oe-CDKN2B group. In summary,
the EVs-miR-106b promotes autophagy and inhibits neuronal apoptosis
CDKN2B was then overexpressed in PD neurons treated with EVs- in PD neurons by targeting and negatively regulating CDKN2B.
miR-106b. RT-qPCR and western were carried out to detect expression
pattern changes, and the results showed that (Fig. 6A, B): compared with 3.7. EVs-miR-106b inhibits CDKN2B to alleviate neuronal damage in PD
the PD + EVs-miR-106b + oe-NC group, there was no significant dif­ mice
ference in miR-106b expression in neurons of PD + EVs-miR-106b + oe-
CDKN2B group, while CDKN2B mRNA and protein expression were CDKN2B overexpression treatment also was performed on EVs-miR-
significantly increased. 106b-treated mice after successful PD modeling to observe the behav­
CCK8, immunofluorescence and flow cytometry were performed to ioral changes in mice. As shown in Fig. 7A, B, compared with the PD +
detect neuronal survival, autophagy, and apoptosis, respectively. EVs-miR-106b + oe-NC group, mice in the PD + EVs-miR-106b + oe-
Compared with the PD + EVs-miR-106b + oe-NC group, the neuronal CDKN2B group spent significantly more time in the pole test, and had
survival rate of PD + EVs-miR-106b + oe-CDKN2B group decreased, significantly lower scores in the swimming test. The results of HE
autophagy was alleviated, and the proportion of apoptotic cells was staining (Fig. 7C) indicated that the neurons in the hippocampus of mice
significantly increased (Fig. 6C-E). Next, western blot was performed to with PD + EVs-miR-106b + oe-NC were more uniformly distributed, and
measure the expression of autophagy-related proteins LC3II and LC3I, the nuclei were more clearly structured. However, some neurons in the
apoptosis-related proteins Bcl-2 and Bax. Fig. 6F exhibited that hippocampus of mice overexpressing CDKN2B were swollen, with
compared with the PD + EVs-miR-106b + oe-NC group, the LC3II/LC3I interstitial edema and shriveled nucleus. TUNEL assay results showed

A PD + EVs-miR-106b + oe-NC
B PD + EVs-miR-106b + oe-NC
PD + EVs-miR-106b + oe-CDKN2B PD + EVs-miR-106b + oe-CDKN2B

*
The total time (s)

25 2.5

20 2.0
Score

15 1.5
*
10 1.0

5 0.5

0 0.0
C

50 μm 50 μm

PD + EVs-miR-106b + oe-NC PD + EVs-miR-106b + oe-CDKN2B

D PD + EVs-miR-106b + oe-NC
PD + EVs-miR-106b + oe-CDKN2B
50
Apoptosis rate (%)

40 *
30

20

10

0
Fig. 7. EVs-miR-106b inhibits CDKN2B to alleviate neuronal damage in PD mice. PD mice were treated with EVs-miR-106b + oe-NC or EVs-miR-106b + oe-CDKN2B.
A, the time spent on pole climbing by each group of mice; B, the score of each group of mice examined by swimming test; C, histopathological changes of hip­
pocampal neurons in the brain of mice were observed by HE staining; D, apoptosis of hippocampal neurons in mice examined by TUNEL assay. *p < 0.05.

9
X. Bai et al. Neuroscience Letters 760 (2021) 136094

(Fig. 7D) that the level of apoptosis was significantly enhanced in the target during nerve repair in post-peripheral nerve injury [30]. Over­
hippocampal tissues of mice in the PD + EVs-miR-106b + oe-CDKN2B expression of CDKN2B resulted in a reduction in retinal ganglion cell
group relative to the PD + EVs-miR-106b + oe-NC group. The above viability in N-methyl-d-aspartate-injected retinas, and adeno-associated
finding indicates that the EVs-miR-106b ameliorates behavioral deficits virus type 2-CRISPR-Cas9-mediated knockout of CDKN2B attenuated N-
and pathological changes in PD mice by targeting and negatively regu­ methyl-d-aspartate-induced retinal ganglion cell death [31]. However,
lating the CDKN2B. whether CDKN2B is also involved in the modulation of neuronal auto­
phagy remains largely unknown. The rescue experiments in vitro and in
4. Discussion vivo exhibited that CDKN2B repressed the neuronal autophagy and
augmented apoptosis.
In recent years, EVs derived from all cell-types have been under­
scored as mediators of PD which remains the second most frequent 5. Conclusion
neurodegenerative diseases worldwide [15]. Moreover, the regulation
of dopaminergic neuronal loss and autophagy is of great importance to In conclusion, our findings reveal that EV-miR-106b released from
reveal the pathogenesis of PD [16,17]. Moreover, therapeutic strategies MSC may relieve PD symptoms via inhibiting neuronal apoptosis and
that boost autophagy may be theoretically beneficial for PD [18]. The enhancing autophagy. The downregulation of CDKN2B may be a key
current study aimed to investigate the effect of MSC-secreted EVs con­ downstream mechanism to mediate the role of EV-miR-106b in PD
taining miR-106b on autophagy and apoptosis of neurons in PD. MPTP, a development. Our study provided a possible novel strategy for the
neurotoxin which recapitulates the neuropathology of PD, could be treatment of PD. Further studies are still essential to verify our findings.
specifically uptaken by dopaminergic neurons and targets the mito­
chondria of neurons, thus triggering loss of dopaminergic neurons in Funding
animals [19]. Taken together, our findings revealed that MSC-derived
EV-miR-106b can promote autophagy and alleviate apoptosis of neu­ None.
rons from MPTP-induced mice by downregulating CDKN2B to relieve
PD.
Competing interests
The capability of EVs to deliver proteins and genetic material be­
tween cells, including miRNAs, provides vital insights as to how EVs
The authors declare that there are no competing interests associated
may involve in pathological progression in PD [20–22]. More relevantly,
with the manuscript.
MSCs-released EVs have been proposed as a capable therapeutic tool
because it has been established that they can serve as biological nano­
particles with advantageous effects on various pathological conditions, CRediT authorship contribution statement
including PD [23]. Firstly, we discovered via a miRNA dataset
GSE16658 that miR-106b was downregulated in PD, which was further Xue Bai: Data curation, Conceptualization, Methodology, Visuali­
proved in the hippocampal tissues of PD mice. In line with our study, the zation, Writing - original draft, Validation. Qiaoyun Dong: Writing -
expression of miR-106b was decreased in the temporal cortex of patients original draft, Writing - review & editing, Formal analysis, Data cura­
with Alzheimer’s disease [24]. MAP-2 has been indicated to be specif­ tion, Validation. Li Zhao: Writing - original draft, Writing - review &
ically expressed in neuronally differentiated cells, and has been formerly editing, Formal analysis, Data curation, Validation. Yan Yao: Data
applied as a sensitive and specific marker for neurons [25]. Therefore, curation, Supervision, Resources. Bo Wang: Visualization, Investigat,
we characterized the extracted neurons from PD mice with MAP-2 ion, Resources, Formal analysis.
expression using immunofluorescence staining. Subsequently, EVs
derived from MSC transfected with miR-106b were used for neuron co- Appendix A. Supplementary data
culture or mouse treatment. It was found that MSC-EVs rescued the
neuronal loss induced by MPTP and enhanced autophagy, while more Supplementary data to this article can be found online at https://doi.
pronounced trends were observed in neurons treated with EV-miR-106b. org/10.1016/j.neulet.2021.136094.
Pretreatment with EVs augmented 6-hydroxydopamine-stimulated SH-
SY5Y cells to proliferate and inhibit apoptosis by inducing autophagy References
[26], which were largely in agreement with our finding. Next, we
revealed the neuroprotective role of EV-miR-106b in vivo, as evidenced [1] L.V. Kalia, A.E. Lang, Parkinson’s disease, Lancet 386 (2015) 896–912.
by increased LC3II/LC3I ratio and Bcl-2 expression, as well as decreased [2] T. Pringsheim, N. Jette, A. Frolkis, T.D. Steeves, The prevalence of Parkinson’s
disease: a systematic review and meta-analysis, Mov. Disord. 29 (2014)
Bax protein expression. 1583–1590.
Furthermore, the current study also indicated that EV-miR-106b [3] A. Ascherio, M.A. Schwarzschild, The epidemiology of Parkinson’s disease: risk
promoted autophagy and inhibited apoptosis of neurons via down­ factors and prevention, Lancet Neurol. 15 (2016) 1257–1272.
[4] L.V. Kalia, S.K. Kalia, A.E. Lang, Disease-modifying strategies for Parkinson’s
regulation of CDKN2B. CDKN2B was first predicted by Starbase to be a disease, Mov. Disord. 30 (2015) 1442–1450.
putative target of miR-106b and validated through dual-luciferase re­ [5] C. Giorgi, E. Bouhamida, A. Danese, M. Previati, P. Pinton, S. Patergnani,
porter assay. CDKN2B was then identified to be overexpressed in neu­ Relevance of autophagy and mitophagy dynamics and markers in
neurodegenerative diseases, Biomedicines 9 (2021).
rons extracted from PD mice, which was downregulated by miR-106b [6] H. Rottenberg, J.B. Hoek, The mitochondrial permeability transition: nexus of
overexpression. CDKN2B (also known as INK4B, p15), lies in band 9p21 aging, disease and longevity, Cells 10 (2021).
and encodes a protein that encourages a G1-phase cell cycle arrest via [7] R. Quiroz-Baez, K. Hernandez-Ortega, E. Martinez-Martinez, Insights into the
proteomic profiling of extracellular vesicles for the identification of early
the suppression of cyclin-dependent kinase 4/6 [27]. CDKN2B was biomarkers of neurodegeneration, Front. Neurol. 11 (2020), 580030.
identified by Chung et al. to share an association with the risk of PD or [8] H. Xing, J. Tan, Y. Miao, Y. Lv, Q. Zhang, Crosstalk between exosomes and
Alzheimer’s disease and the severity of cognitive impairment in both autophagy: a review of molecular mechanisms and therapies, J. Cell Mol. Med. 25
(2021) 2297–2308.
Caucasian and Asian populations [28]. Folch et al. also found that acute
[9] Y. Zhao, G. Yang, Potential of extracellular vesicles in the Parkinson’s disease -
exposure of rat cerebellar granule neurons to micromolar concentrations pathological mediators and biomarkers, Neurochem. Int. 144 (2021), 104974.
of pentachlorophenol enhanced the transcriptional activity of genes [10] M. d’Angelo, A. Cimini, V. Castelli, Insights into the effects of mesenchymal stem
related to mitogenic response, including CDKN2B [29]. CDKN2B cell-derived secretome in Parkinson’s disease, Int. J. Mol. Sci. 21 (2020).
[11] B. Wang, H. Jia, B. Zhang, J. Wang, C. Ji, X. Zhu, Y. Yan, L. Yin, J. Yu, H. Qian,
knockdown has been reported to noticeably increase the axon outgrowth W. Xu, Pre-incubation with hucMSC-exosomes prevents cisplatin-induced
in dorsal root ganglion neurons, indicating it might be a novel potential nephrotoxicity by activating autophagy, Stem Cell Res. Ther. 8 (2017) 75.

10
X. Bai et al. Neuroscience Letters 760 (2021) 136094

[12] S. Xie, W. Niu, F. Xu, Y. Wang, S. Hu, C. Niu, Differential expression and [23] H. Vilaça-Faria, A.J. Salgado, F.G. Teixeira, Mesenchymal stem cells-derived
significance of miRNAs in plasma extracellular vesicles of patients with Parkinson’s exosomes: a new possible therapeutic strategy for Parkinson’s disease? Cells 8
disease, Int. J. Neurosci. (2020) 1–16. (2019).
[13] S. Wang, Y. Huang, C. Zhou, H. Wu, J. Zhao, L. Wu, M. Zhao, F. Zhang, H. Liu, The [24] W. Liu, J. Zhao, G. Lu, miR-106b inhibits tau phosphorylation at Tyr18 by targeting
role of autophagy and related microRNAs in inflammatory bowel disease, Fyn in a model of Alzheimer’s disease, Biochem. Biophys. Res. Commun. 478
Gastroenterol. Res. Pract. 2018 (2018) 7565076. (2016) 852–857.
[14] V. Jackson-Lewis, S. Przedborski, Protocol for the MPTP mouse model of [25] Y. Liu, R.S. Saad, S.S. Shen, J.F. Silverman, Diagnostic value of microtubule-
Parkinson’s disease, Nat. Protoc. 2 (2007) 141–151. associated protein-2 (MAP-2) for neuroendocrine neoplasms, Adv. Anat. Pathol. 10
[15] S. Rastogi, V. Sharma, P.S. Bharti, K. Rani, G.P. Modi, F. Nikolajeff, S. Kumar, The (2003) 101–106.
evolving landscape of exosomes in neurodegenerative diseases: exosomes [26] H.X. Chen, F.C. Liang, P. Gu, B.L. Xu, H.J. Xu, W.T. Wang, J.Y. Hou, D.X. Xie, X.
characteristics and a promising role in early diagnosis, Int. J. Mol. Sci. 22 (2021). Q. Chai, S.J. An, Exosomes derived from mesenchymal stem cells repair a
[16] S. Ghavami, S. Shojaei, B. Yeganeh, S.R. Ande, J.R. Jangamreddy, M. Mehrpour, Parkinson’s disease model by inducing autophagy, Cell Death Dis. 11 (2020) 288.
J. Christoffersson, W. Chaabane, A.R. Moghadam, H.H. Kashani, M. Hashemi, A. [27] M. De Braekeleer, N. Douet-Guilbert, E. De Braekeleer, Prognostic impact of p15
A. Owji, M.J. Łos, Autophagy and apoptosis dysfunction in neurodegenerative gene aberrations in acute leukemia, Leuk. Lymphoma 58 (2017) 257–265.
disorders, Prog. Neurobiol. 112 (2014) 24–49. [28] S.J. Chung, M.-J. Kim, J. Kim, H.-S. Ryu, Y.J. Kim, S.Y. Kim, J.-H. Lee, Association
[17] D.J. Surmeier, Determinants of dopaminergic neuron loss in Parkinson’s disease, of type 2 diabetes GWAS loci and the risk of Parkinson’s and Alzheimer’s diseases,
FEBS J. 285 (2018) 3657–3668. Parkinsonism Relat. Disord. 21 (2015) 1435–1440.
[18] S. Cerri, F. Blandini, Role of autophagy in Parkinson’s disease, Curr. Med. Chem. [29] J. Folch, M. Yeste-Velasco, D. Alvira, A.V. de la Torre, M. Bordas, M. López, F.
26 (2019) 3702–3718. X. Sureda, V. Rimbau, A. Camins, M. Pallàs, Evaluation of pathways involved in
[19] E. Lee, I. Hwang, S. Park, S. Hong, B. Hwang, Y. Cho, J. Son, J.W. Yu, MPTP-driven pentachlorophenol-induced apoptosis in rat neurons, Neurotoxicology 30 (2009)
NLRP3 inflammasome activation in microglia plays a central role in dopaminergic 451–458.
neurodegeneration, Cell Death Differ. 26 (2019) 213–228. [30] D. Wang, Y. Chen, M. Liu, Q. Cao, Q. Wang, S. Zhou, Y. Wang, S. Mao, X. Gu,
[20] Y.H. Chang, K.C. Wu, H.J. Harn, S.Z. Lin, D.C. Ding, Exosomes and stem cells in Z. Luo, B. Yu, The long noncoding RNA Arrl1 inhibits neurite outgrowth by
degenerative disease diagnosis and therapy, Cell Transplant. 27 (2018) 349–363. functioning as a competing endogenous RNA during neuronal regeneration in rats,
[21] D.J. Park, W.S. Yun, W.C. Kim, J.E. Park, S.H. Lee, S. Ha, J.S. Choi, J. Key, Y.J. Seo, J. Biol. Chem. 295 (2020) 8374–8386.
Improvement of stem cell-derived exosome release efficiency by surface-modified [31] H. Tawarayama, Q. Feng, N. Murayama, N. Suzuki, T. Nakazawa, Cyclin-dependent
nanoparticles, J. Nanobiotechnol. 18 (2020) 178. kinase inhibitor 2b mediates excitotoxicity-induced death of retinal ganglion cells,
[22] J.R. Pinnell, M. Cui, K. Tieu, Exosomes in Parkinson disease, J. Neurochem. 157 Invest. Ophthalmol. Vis. Sci. 60 (2019) 4479–4488.
(2021) 413–428.

11

You might also like