Akt activation by targeting PDCD4

Akt activation by targeting PDCD4

Journal Pre-proof MiR-340-5p alleviates oxygen-glucose deprivation/reoxygenation-induced neuronal injury via PI3K/Akt activation by targeting PDCD4 Ya...

2MB Sizes 0 Downloads 36 Views

Journal Pre-proof MiR-340-5p alleviates oxygen-glucose deprivation/reoxygenation-induced neuronal injury via PI3K/Akt activation by targeting PDCD4 Yake Zheng, Peng Zhao, Yajun Lian, Shuang Li, Yuan Chen, Lihao Li PII:

S0197-0186(19)30441-3

DOI:

https://doi.org/10.1016/j.neuint.2019.104650

Reference:

NCI 104650

To appear in:

Neurochemistry International

Received Date: 8 August 2019 Revised Date:

28 November 2019

Accepted Date: 19 December 2019

Please cite this article as: Zheng, Y., Zhao, P., Lian, Y., Li, S., Chen, Y., Li, L., MiR-340-5p alleviates oxygen-glucose deprivation/reoxygenation-induced neuronal injury via PI3K/Akt activation by targeting PDCD4, Neurochemistry International (2020), doi: https://doi.org/10.1016/j.neuint.2019.104650. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier Ltd.

1

MiR-340-5p

alleviates

oxygen-glucose

deprivation/reoxygenation-induced

2

neuronal injury via PI3K/Akt activation by targeting PDCD4

3

Yake Zheng1, Peng Zhao2, Yajun Lian1,*, Shuang Li1, Yuan Chen1, Lihao Li1

4

1

5

Zhengzhou, China, 450052;

6

2

7

China, 450004;

8

* Corresponding author: Yajun Lian

9

Jianshe East Road, The First Affiliated Hospital of Zhengzhou University, Zhengzhou,

Department of Neurology, The First Affiliated Hospital of Zhengzhou University,

Department of Neurology, The First People’s Hospital of Zhengzhou, Zhengzhou,

10

China, 450052;

11

Email: [email protected]

12

Tel: 13838367143

13

1

1

Abstract

2

MicroRNA-340-5p (miR-340-5p), a suppressor of certain target genes in brain,

3

reportedly is decreased in peripheral circulation of acute stroke patients. However,

4

little is known regarding its role in regulating cerebral ischemia/reperfusion injury.

5

This study explores the effect of miR-340-5p on ischemia/reperfusion insults by

6

exposing rat hippocampal neurons to oxygen-glucose deprivation/reoxygenation

7

(OGDR) in vitro. We found miR-340-5p to be poorly expressed in these neurons after

8

OGDR stimulation. OGDR stimulation decreased cell viability, increased lactate

9

dehydrogenase (LDH) activity and cell apoptosis, all of which were significantly

10

inhibited by miR-340-5p overexpression and enhanced by miR-340-5p inhibition.

11

Using bioinformatics analysis, we identified mRNA encoding the pro-apoptotic factor,

12

programmed cell death 4 (PDCD4) as a putative target of miR-340-5p. A

13

dual-luciferase reporter assay suggested that miR-340-5p targeted the 3’-UTR of

14

PDCD4. PDCD4 was upregulated in cells exposed to OGDR, and miR-340-5p

15

negatively modulated expression of PDCD4. PDCD4 overexpression partly reversed

16

the neuroprotective effect of miR-340-5p during OGDR-induced injury. MiR-340-5p

17

overexpression significantly promoted the activation of PI3K/Akt signaling pathway

18

(P<0.05) in OGDR-exposed cells, and PDCD4 overexpression attenuated this effect

19

(P<0.05). Collectively, our results indicate that miR-340-5p might exerted

20

neuroprotective effects during OGDR injury by targeting PDCD4 and then activating

21

the PI3K/Akt pathway. These results indicated a novel target for treating cerebral

22

ischemic injury.

23

Keywords: miR-340-5p, oxygen glucose deprivation/reoxygenation (OGDR),

24

neuroprotection, PDCD4, PI3K/Akt

25 26

Abbreviations

27

miRNAs, microRNAs; IR, ischemia/reperfusion; PDCD4, programmed cell death 4;

28

OGDR, oxygen-glucose deprivation/reoxygenation; FBS, fetal bovine serum;

29

HEK293, human embryonic kidney 293 cells; LDH, lactate dehydrogenase; PI,

30

propidium iodide; PVDF, polyvinylidene difluoride 2

1

Introduction

2

Ischemic stroke is a brain disease caused by temporary cessation or insufficient

3

blood flow to brain. It is associated with high disability and mortality rates(Stegner et

4

al., 2019). The subsequent reperfusion and reoxygenation of blood can further

5

aggravate tissue injury and cell death(Galkin, 2019). Due to its high disability and

6

mortality, ischemic stroke brings heavy burden to the patients and the society(Patel

7

and McMullen, 2017). Understanding the complex pathophysiological mechanism

8

underlying stroke-related neuron damage may help identify effective treatments for

9

ischemic stroke.

10

MicroRNAs (miRNAs) are small (18–28 nucleotides), noncoding RNAs that

11

negatively modulate expression of their target genes by binding to the 3’-UTRs of

12

target mRNA. MiRNAs help modulate pathophysiological processes in cerebral

13

ischemia/reperfusion (IR) injury. For example, miR-34b targets Keap1 to protect

14

against focal cerebral IR injury in rats with middle cerebral artery occlusion (Huang et

15

al., 2019). MiR-424 inhibits oxidative stress to ameliorate focal cerebral IR injury in

16

mice (Liu et al., 2015). MiR-99a reduces cerebral IR-induced neuronal damage by

17

regulating the cell cycle and apoptosis (Tao et al., 2015). MiR-200a silencing protects

18

neural stem cells during cerebral IR injury (Ma et al., 2017). Ou et al. reports that

19

miR-375 exhibits protective effects against cerebral IR injury by targeting Ctgf (Ou et

20

al., 2017). Additionally, miR-182-5p mitigates neuronal injury after cerebral IR injury

21

by targeting Toll-like receptor 4 (Wang et al., 2018). In rats with sciatic nerve crush

22

injury, miR-340 dysregulation occurs in the injured nerves, which affects the removal

23

of cell debris and axon regeneration (Li et al., 2017). Furthermore, miR-340-5p is

24

downregulated in the peripheral blood of patients with acute stroke (Yoo et al., 2018).

25

These findings suggest that miRNAs are potential targets for cerebral IR injury.

26

However, the specific function of miR-340-5p in cerebral IR-induced neural injury

27

remains unknown.

28

Apoptosis is a critical mechanism in cerebral IR injury, and the programmed cell

29

death 4 (PDCD4) protein mediates this process. PDCD4 knockdown attenuates mouse

30

myocardial injury by inhibiting apoptosis(Xu et al., 2017). It also inhibits protein 3

1

translation and accelerates apoptosis (Colburn et al., 2006). A study reveals that

2

miR-499-5p protects against cardiomyocyte ischemic injury by directly targeting

3

PDCD4 (Li et al., 2016). Other studies shows that PDCD4 participates in regulation

4

of acute myocardial infarction (Gu et al., 2018). PDCD4 also directly binds to miR-21

5

and helps mediate ischemic neuron death (Yan et al., 2017). Therefore, targeting

6

PDCD4 may be a novel therapeutic strategy for cerebral IR injury.

7

In

this

study,

we

used

an

in

vitro

model

of

oxygen-glucose

8

deprivation/reperfusion (OGDR) injury in primary rat hippocampal neurons to

9

simulate cerebral IR injury. We explored the expression, effect, and potential

10

mechanism of miR-340-5p in OGDR-injured hippocampal neurons.

11

Materials and methods

12

Cell culture

13

Primary hippocampal neurons were isolated, as previously described (Krohn et al.,

14

1998) (provided by the Committee of Animal Care and Use at Zhengzhou University).

15

Briefly, the hippocampal tissues were dissected from neonatal SD rats and dissociated

16

using trypsin-EDTA (0.25%). The isolated cells were seeded on neurobasal media

17

(Life Technologies, Carlsbad, CA, USA) using GlutaMAX, B27, and glucose (4.5

18

mg/ml) for 7 days. Cells then were cultured in neurobasal medium with 5% fetal

19

bovine serum (FBS) and glucose (4.5 mg/ml) for 14 days. Animal experiments were

20

approved by The First Affiliated Hospital of Zhengzhou University. Human

21

embryonic kidney 293 (HEK293) cells (ATCC, Manassas, VA, USA) were grown in

22

DMEM (Life Technologies, Carlsbad, CA, USA). Upon reaching 70% to 80%

23

confluency, the HEK293 cells were collected for experiments. All cells were cultured

24

in an incubator containing 5% CO2 at 37 °C.

25

Oxygen-glucose deprivation/reperfusion model

26

Hippocampal neurons were cultured in OGDR conditions to mimic cerebral IR in

27

vitro. The OGDR model was prepared as previously described (Yang et al., 2015).

28

Briefly, hippocampal neurons were subjected to glucose-free DMEM (Gibco, Grand

29

Island, NY, USA) and incubated at 37 °C in hypoxic conditions (1% O2, 94% N2, 5%

30

CO2) for 2 h. Then, the cells were cultured in normal DMEM containing glucose in a 4

1

normoxic atmosphere (95% air, 5% CO2) for 24 h.

2

Cell transfection

3

Hippocampal neurons were transfected with a miR-340-5p mimic, miR-340-5p

4

inhibitor, and their respective negative controls, mimic Ctrl and inhibitor Ctrl

5

(GenePharma, Shanghai, China), using Lipofectamine 2000 (Invitrogen, Carlsbad,

6

CA, USA). MiR-340-5p mimic sequences: 5’-UUA UAA AGC AAU GAG ACU GA

7

UU-3’; mimic control sequences are: 5’- UUC UCC GAA CGU GUC ACG UTT-3’;

8

miR-340-5p inhibitor sequences: 5’-AAU CAG UCU CAU UGC UUU AU AA-3’;

9

inhibitor control sequences: 5’-CAC UAC UUU UGU GU AGUA CAA-3’. The

10

full-length of PDCD4 was amplified and cloned into the pcDNA3.1 plasmid to

11

construct pcDNA3.1-PDCD4. The pcDNA3.1-PDCD4 or pcDNA3.1 plasmid was

12

transfected into hippocampal neurons using Lipofectamine 2000.

13

Cell viability

14

MiRNA-transfected hippocampal neurons were seeded in 96-well plates (5×104

15

cells/well).

After

OGDR

exposure,

cells

were

incubated

with

3-(4,

16

5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT, 5 mg/ml; Sigma St.

17

Louis, MO, USA) for 4 h. Dimethyl sulfoxide (Sigma) was used to dissolve the

18

formazan crystals. Absorbance at 490 nm was detected to determine cell viability.

19

Lactate dehydrogenase assay

20

Neuronal cells in each group were cultured into 96-well plates, and the collected

21

cell medium were mixed with the lactate dehydrogenase (LDH) assay reagent

22

(Nanjing Jiancheng Bioengineering Institute, Nanjing, China) for 1 h. LDH activity

23

was determined via detection of absorbance at 450 nm.

24

Flow cytometry

25

An annexin V/propidium iodide (PI) kit (BD Bioscience, Franklin Lakes, NJ, USA)

26

was used to determine apoptosis. After incubation at different experimental conditions,

27

the neurons were plated in 6-well plates (5×105 cells/well) and suspended in binding

28

buffer with 10 µL of Annexin V-FITC and 10 µL of PI in the dark for 15 minutes. 5

1

Afterwards, flow cytometry was conducted to determine apoptotic cells.

2

qRT-PCR

3

Total RNA was extracted from TRIzol reagent (Invitrogen), and cDNA synthesis

4

was conducted using a miRNA reverse transcription kit (Applied Biosystems, Foster

5

City, CA, USA) for miR-340-5p detection. Real-time PCR was performed using a

6

SYBR Green qPCR Master Mix Kit (Applied Biosystems). Real-time PCR was

7

performed using the ABI PRISM 7900 system (Applied Biosystems). The primers

8

were as follows: miR-340-5p forward 5’-GCG GTT ATA AAG CAA TG AGA-3’ and

9

reverse 5’-GTG CGT GTC GTG GAG TCG-3’; U6 forward 5’-CTC GCT TCG GCA

10

GCA CA-3’ and reverse 5’-AAC GCT TCA CGA ATT TG CGT-3’. The relative

11

abundance of miR-340-5p was normalized to U6. Data were quantified using the

12

2−∆∆Ct method.

13

Western blot

14

Hippocampal neurons were lysed in lysis buffer for 0.5 h. After cells were

15

centrifuged, the protein concentrations in lysates were measured using a bicinchoninic

16

acid (BCA) assay (Beyotime Biotechnology, Haimen, China). Equivalent amounts of

17

protein were separated using 10%–15% SDS-PAGE and transferred to polyvinylidene

18

difluoride (PVDF) membranes (Millipore, Boston, MA, USA), which were blocked

19

and incubated at 4 °C overnight with primary antibodies, including PDCD4 (1:1000),

20

Bcl-2 (1:1000), Bax (1:1000), p-PI3K (1:1000), PI3K (1:1000), p-Akt phospho

21

Ser473 (1:5000), Akt (1:500), and internal control β-actin (1:1000), all from Abcam

22

(Cambridge, MA, USA). After washing with TBST, the membranes were probed with

23

secondary antibody IgG (1:5000; ZSGB-BIO, Beijing, China) for 2 h. Blots were

24

processed using an enhanced chemiluminescence reaction system (Amersham, Little

25

Chalfont, UK). Gray scale was quantified using Quantity One Software (Bio-Rad,

26

Hercules, CA, USA).

27

Dual luciferase activity assay

28

A 350 bp sequence from the 3’-UTR of PDCD4 containing the miR-340-5p binding 6

1

site (Wt) was amplified by PCR. The sequence for mutant PDCD4 (Mut) was

2

amplified by point mutation method using KOD-Plus mutagenesis Kit (TOYOBO,

3

Osaka, Japan). The fragments from the Wt or Mut PDCD4 3’-UTR were then

4

subcloned into the pGL3 luciferase promoter vector (Promega, Madison, WI, USA).

5

HEK 293 cells or OGDR induced hippocampal neurons were co-transfected with the

6

constructs and miR-340-5p mimic or mimic control using Lipofectamine 2000

7

(Invitrogen) for 48 h. Luciferase activities were detected using the dual-luciferase

8

assay system (Promega).

9

Statistical analysis

10

Data are expressed as mean ± standard derivations (SD) and analyzed with SPSS

11

22.0 (SPSS Inc., Chicago, IL, USA). Differences between two groups were compared

12

via t test and differences among multiple groups were examined using ANOVA

13

followed by Bonferroni test.

14

Results

15

Low abundance of miR-340-5p in hippocampal neurons exposed to OGDR

16

To assess the effect of miR-340-5p in OGDR-injured hippocampal neurons, we first

17

used qRT-PCR to detect the level of miR-340-5p. The results showed significantly

18

downregulated abundance of miR-340-5p in OGDR-injured hippocampal neurons

19

(0.34±0.07), compared with the control (1.00±0.06) (t=12.61, P<0.05, Fig.1A),

20

implying that miR-340-5p may play an important role against OGDR injury of

21

hippocampal neurons.

22

MiR-340-5p had protective effect against neuronal injury caused by OGDR

23

To investigate the contribution of miR-340-5p in OGDR-induced hippocampal

24

neuron injury, neurons were transfected with a miR-340-5p mimic or miR-340-5p

25

inhibitor for 24 h and then exposed to OGDR. The miR-340-5p mimic upregulated

26

miR-340-5p abundance, and the miR-340-5p inhibitor significantly downregulated

27

miR-340-5p abundance in the injured neurons (F=170.1, P<0.05, Fig. 1B). 7

1

MiR-340-5p overexpression resulted in increased cell viability (F=54.94, P<0.05, Fig.

2

2A), and significantly decreased LDH activity (F=78.45, P<0.05, Fig. 2B) and

3

apoptosis (F=40.87, P<0.05, Fig. 2C) in OGDR treated cells. In contrast, miR-340-5p

4

suppression showed the opposite effect. Moreover, the upregulation of Bax (F=62.36,

5

P<0.05, Fig. 2D), the downregulation of Bcl-2 (F=41.63, P<0.05, Fig. 2D), and the

6

increased ratio of Bax/Bcl-2 (F=37.34, P<0.05, Fig. 2E), which induced by OGDR,

7

were weakened by miR-340-5p overexpression, whereas miR-340-5p suppression

8

produced the opposite effects. Overall, these results suggest that miR-340-5p had a

9

neuroprotective effect in OGDR-induced neuronal injury.

10

MiR-340-5p directly regulated PDCD4 expression

11

To explore the mechanism underlying the neuroprotective role of miR-340-5p, we

12

conducted a bioinformatic analysis of its targets. We identified PDCD4, a

13

pro-apoptotic gene, as a putative target of miR-340-5p (Fig. 3C). We also found that

14

co-transfection with the miR-340-5p mimic and the PDCD4-wt 3’-UTR significantly

15

decreased luciferase activity as compared with cells co-tranfected with miR-340-5p

16

mimic control and PDCD4-wt (t=7.933, P<0.05). However, co-transfection of the

17

miR-340-5p mimic and PDCD4-mut 3’-UTR showed no obvious effect on luciferase

18

activity in HEK 293 cells (Fig. 3A). Similar results was also detected in hippocampal

19

neurons exposed to OGDR (Fig. 3B). Furthermore, miR-340-5p inhibition markedly

20

upregulated the protein content of PDCD4 during OGDR, whereas its overexpression

21

restrained PDCD4 content (F=187.5, P<0.05, Fig. 3D and E). These results

22

demonstrate that miR-340-5p directly targeted PDCD4.

23

MiR-340-5p downregulated PDCD4 to protect hippocampal neurons against

24

injury caused by OGDR

25

To further investigate the contribution of PDCD4 to the protective effects of

26

miR-340-5p in OGDR-induced neuron injury, neurons were co-transfected with a

27

PDCD4 vector and miR-340-5p mimic. The data showed that PDCD4 overexpression

28

impeded the ability of miR-340-5p to repress PDCD4’s protein content. (F=33.73, Fig. 8

1

4A). In neurons exposed to OGDR, the miR-340-5p mimic improved cell viability

2

(F=19.48, Fig. 4B), reduced LDH (F=14.58, Fig. 4C) and apoptosis (F=23.96, Fig.

3

4D) levels. PDCD4 overexpression inhibited these effects (P<0.05). Additionally, The

4

miR-340-5p mimic led to upregulation of Bcl-2 (F=31.04, P<0.05, Fig. 4E),

5

downregulation of Bax

6

ratio in OGDR-injured neuron (F=24.04, P<0.05, Fig. 4F)s. PDCD4 overexpression

7

blocked these effects (P<0.05, Fig. 4E and 4F). These data indicate that PDCD4

8

overexpression abrogated the neuroprotective role of miR-340-5p in OGDR-injured

9

neurons.

(F=17.64, P<0.05, Fig. 4E), and an increased Bax/Bcl-2

10

MiR-340-5p overexpression activated the PI3K/Akt signaling pathway in

11

neurons exposed to OGDR

12

The PI3K/Akt pathway is an important cell survival signaling pathway that protects

13

against cerebral IR injury(Breivik et al., 2015). We explored its involvement in

14

modulating miR-340-5p in OGDR-injured neurons. Western blot results showed that

15

the phosphorylation of p-PI3K (F=61.95, Fig. 5B) and p-Akt (F=54.35, Fig. 5C),

16

which was inhibited by OGDR, was remarkably increased by miR-340-5p

17

overexpression, whereas it was significantly reduced by miR-340-5p suppression

18

(P<0.05, Fig.5).

19

MiR-340-5p targeted PDCD4 to influence PI3K/Akt signaling

20

Finally, we tested whether the PI3K/Akt pathway helped regulate miR-340-5p and

21

PDCD4 in OGDR-injured hippocampal neurons. Cells were co-transfected with

22

miR-340-5p mimic and PDCD4 vector and then exposed to OGDR. As shown in Fig.

23

6, miR-340-5p overexpression upregulated the phosphorylation of p-PI3K (F=61.95,

24

Fig. 6B) and p-Akt (F=61.95, Fig. 6C), whereas this upregulation was partly reversed

25

by PDCD4 overexpression (P<0.05). These results indicate that miR-340-5p could

26

activate the PI3K/Akt pathways by downregulating PDCD4.

27

Discussion

28

After cerebral IR injury, nerve cells undergo complex pathophysiological 9

1

processes. Treating cerebral IR injury requires alleviating this damage to save dying

2

nerve cells. Therefore, it is important to identify the targets in IR injury. MiRNAs

3

serve a crucial role in cerebral IR injury. For example, miR-340 has a role in nerve

4

injury(Cosset et al., 2016), and miR-340-5p is downregulated in peripheral blood

5

from patients with acute stroke (Yoo et al., 2018). However, the function of

6

miR-340-5p in IR-induced cerebral neural injury is unknown.

7

In the current study, we used primary rat hippocampal neurons to establish an in

8

vitro OGDR injury model that mimics cerebral IR injury. We found miR-340-5p to be

9

downregulated in primary hippocampal neurons that were injured by OGDR.

10

MiR-340-5p overexpression targeted PDCD4 to enhance cell viability and reduce

11

LDH activity and apoptosis, whereas miR-340-5p inhibition showed the opposite

12

effects. Our study confirmed the contribution of miR-340-5p in regulating OGDR

13

injury, revealing a potential target for treating cerebral ischemic stroke. In future

14

researches, it is needed to explore the function of miR-340-5p in other brain cells,

15

such as glial cells, to further understand the role of miR-340-5p in cerebral IR injury

16

and in cerebral diseases.

17

PDCD4 is a crucial regulator of apoptosis(Singh et al., 2019). It reportedly

18

participates in translation, transcription, and cell growth regulation(Matsuhashi et al.,

19

2019). Numerous studies have found low expression of PDCD4 in various cancers,

20

and it is associated with the progression of prostate cancer (Zennami et al., 2019),

21

colon carcinoma (Wang et al., 2017), and glioma(Monfared et al., 2019). Recently,

22

PDCD4 was identified as a crucial gene in the regulation of IR injury. A study has

23

shown that by regulating the miR-21/PDCD4 signaling pathway, serum-derived

24

extracellular vesicles protect mouse hearts against acute myocardial infarction (Gu et

25

al., 2018). Moreover, the lncRNA MEG3 regulates ischemic neuronal death by

26

targeting the miR-21/PDCD4 signaling pathway (Yan et al., 2017). In rat cardiac cells

27

exposed to IR injury, miR-21 regulates PDCD4, which exerts an anti-apoptotic effect

28

(Cheng et al., 2010). By targeting PDCD4, miR-499 inhibits cardiomyocyte apoptosis

29

and is involved in ischemic postconditioning-mediated cardiac protection against IR 10

1

injury (Zhu et al., 2016). In our study, it was revealed that miR-340-5p targeted and

2

negatively modulated the PDCD4 gene. PDCD4 protein levels markedly increased in

3

rat primary hippocampal neurons that were exposed to OGDR. Furthermore, we

4

confirmed that PDCD4 participated in the neuroprotective effect of miR-340-5p on

5

hippocampal neurons that were injured by OGDR.

6

The PI3K/Akt signaling pathway is an important survival signal in various nerve

7

cells. This pathway mainly involves PI3K, Akt, and their downstream molecules. It is

8

an important pathway for intracellular transduction of membrane receptor signals and

9

for various cellular processes, such as proliferation, differentiation, and apoptosis.

10

PI3K/Akt signaling plays a crucial role in cerebral IR injury(Kamel et al., 2019).

11

MiR-375 and Ctgf protect against cerebral IR injury by regulating the p21/PI3K/Akt

12

signaling pathways (Ou et al., 2017). MiR-19b exerts neuronal protective effects

13

during hypoxia/reoxygenation-induced injury by targeting PTEN via the PI3K/Akt

14

pathway (Liu et al., 2019). MiR-340 may impede laryngeal squamous cell carcinoma

15

progression by targeting EZH2 via p27 expression enhancement and PI3K/Akt

16

inactivation (Yu et al., 2016). MiR-340 suppression also has been shown to induce

17

activation of PI3K and Akt in ovarian cancer SKOV3 cells (Qu and Wang, 2017).

18

In this study, we found that OGDR injury blocked PI3K/Akt signaling and that

19

miR-340-5p mimics could activate this signaling. This result is contrary to the effects

20

of miR-340-5p on PI3K and Akt that have been previously described by (Yu et al.,

21

2016) and (Qu and Wang, 2017). These contradictory findings indicate that the effects

22

of miR-340-5p on the PI3K/Akt pathway might depend on the experimental

23

conditions. Moreover, there may be more than one target gene for miR-340-5p.

24

Therefore, miR-340a-5p may influence different target gene and then have

25

contradictory effect on PI3K/Akt signaling in different disease. Previous research

26

indicates that PDCD4 may impede Akt activation(Cristofoletti et al., 2019). Our data

27

reveal that miR-340-5p activated suppression of the PI3K/Akt pathway in

28

OGDR-injured hippocampal neurons and that PDCD4 overexpression partly reversed

29

this effect, which suggested miR-340-5p regulated OGDR-induced neurons injury 11

1

partially by modulating PI3K/Akt pathway. In future research, we should explore

2

other target gene of miR-340-5p apart from in ischemic stroke, and further understand

3

the underlying mechanism of miR-340-5P in brain injury. Additionally, the

4

down-stream of PDCD4 should also be investigated, thus other I/R related signaling

5

pathways regulated by PDCD4 should be explored.

6

In summary, our study suggests that miR-340-5p alleviated OGDR-induced

7

injury to neurons by targeting PDCD4 to activate the PI3K/Akt pathway. We

8

identified miR-340 as a promising target for the prevention of cerebral ischemic injury.

9

Our in vitro experiment examined the role of miR-340-5p in hippocampal neurons

10

that were exposed to OGDR injury. Its role in cerebral IR injury needs to be further

11

investigated in vivo.

12

Conflicts of interest

13

No conflict of interest.

14

12

1

References

2

Breivik, L., Jensen, A., Guvag, S., Aarnes, E.K., Aspevik, A., Helgeland, E., Hovland, S.,

3

Brattelid, T., Jonassen, A.K., 2015. B-type natriuretic peptide expression and cardioprotection

4

is regulated by Akt dependent signaling at early reperfusion. Peptides 66, 43-50.

5

Cheng, Y., Zhu, P., Yang, J., Liu, X., Dong, S., Wang, X., Chun, B., Zhuang, J., Zhang, C.,

6

2010.

7

ischaemia/reperfusion injury via anti-apoptosis through its target PDCD4. Cardiovasc Res 87,

8

431-439.

9

Colburn, N.H., Yang, H.-S., Jansen, A., 2006. Pdcd4 targets eIF4A to inhibit translation,

Ischaemic

preconditioning-regulated

miR-21

protects

heart

against

10

transcription, tumorigenesis, and invasion. EJC Supplements 4, 23.

11

Cosset, E., Petty, T., Dutoit, V., Tirefort, D., Otten-Hernandez, P., Farinelli, L., Dietrich, P.Y.,

12

Preynat-Seauve, O., 2016. Human tissue engineering allows the identification of active

13

miRNA regulators of glioblastoma aggressiveness. Biomaterials 107, 74-87.

14

Cristofoletti, C., Bresin, A., Picozza, M., Picchio, M.C., Monzo, F., Helmer Citterich, M.,

15

Passarelli, F., Frezzolini, A., Scala, E., Monopoli, A., Cantonetti, M., Benucci, R., D'Atri, S.,

16

Caprini, E., Russo, G., Narducci, M.G., 2019. Blood and skin-derived Sezary cells:

17

differences in proliferation-index, activation of PI3K/AKT/mTORC1 pathway and its

18

prognostic relevance. Leukemia 33, 1231-1242.

19

Galkin, A., 2019. Brain Ischemia/Reperfusion Injury and Mitochondrial Complex I Damage.

20

Biochemistry (Mosc) 84, 1411-1423.

21

Gu, H., Liu, Z., Li, Y., Xie, Y., Yao, J., Zhu, Y., Xu, J., Dai, Q., Zhong, C., Zhu, H., Ding, S.,

22

Zhou, L., 2018. Serum-Derived Extracellular Vesicles Protect Against Acute Myocardial 13

1

Infarction by Regulating miR-21/PDCD4 Signaling Pathway. Front Physiol 9, 348.

2

Huang, R., Ma, J., Niu, B., Li, J., Chang, J., Zhang, Y., Liu, P., Luan, X., 2019. MiR-34b

3

Protects Against Focal Cerebral Ischemia-Reperfusion (I/R) Injury in Rat by Targeting Keap1.

4

J Stroke Cerebrovasc Dis 28, 1-9.

5

Kamel, E.O., Hassanein, E.H.M., Ahmed, M.A., Ali, F.E.M., 2019. Perindopril Ameliorates

6

Hepatic

7

JAK1/STAT-3, Nrf-2, and PI3K/Akt/mTOR Signaling Pathways. Anat Rec (Hoboken).

8

Krohn, A.J., Preis, E., Prehn, J.H., 1998. Staurosporine-induced apoptosis of cultured rat

9

hippocampal neurons involves caspase-1-like proteases as upstream initiators and increased

Ischemia

Reperfusion

Injury

Via

Regulation

of

NF-kappaB-p65/TLR-4,

10

production of superoxide as a main downstream effector. J Neurosci 18, 8186-8197.

11

Li, S., Zhang, R., Yuan, Y., Yi, S., Chen, Q., Gong, L., Liu, J., Ding, F., Cao, Z., Gu, X., 2017.

12

MiR-340 Regulates Fibrinolysis and Axon Regrowth Following Sciatic Nerve Injury. Mol

13

Neurobiol 54, 4379-4389.

14

Li, Y., Lu, J., Bao, X., Wang, X., Wu, J., Li, X., Hong, W., 2016. MiR-499-5p protects

15

cardiomyocytes against ischaemic injury via anti-apoptosis by targeting PDCD4. Oncotarget

16

7, 35607-35617.

17

Liu, P., Zhao, H., Wang, R., Wang, P., Tao, Z., Gao, L., Yan, F., Liu, X., Yu, S., Ji, X., Luo, Y.,

18

2015. MicroRNA-424 protects against focal cerebral ischemia and reperfusion injury in mice

19

by suppressing oxidative stress. Stroke 46, 513-519.

20

Liu,

21

hypoxia/reoxygenation-induced injury by targeting PTEN. J Cell Physiol.

22

Ma, J., Shui, S., Han, X., Guo, D., Li, T., Yan, L., 2017. microRNA-200a silencing protects

W.G.,

Han,

L.L.,

Xiang,

R.,

14

2019.

Protection

of

miR-19b

in

1

neural stem cells against cerebral ischemia/reperfusion injury. PLoS One 12, e0172178.

2

Matsuhashi, S., Manirujjaman, M., Hamajima, H., Ozaki, I., 2019. Control Mechanisms of the

3

Tumor Suppressor PDCD4: Expression and Functions. Int J Mol Sci 20.

4

Monfared, H., Jahangard, Y., Nikkhah, M., Mirnajafi-Zadeh, J., Mowla, S.J., 2019. Potential

5

Therapeutic Effects of Exosomes Packed With a miR-21-Sponge Construct in a Rat Model of

6

Glioblastoma. Front Oncol 9, 782.

7

Ou, J., Kou, L., Liang, L., Tang, C., 2017. MiR-375 attenuates injury of cerebral

8

ischemia/reperfusion via targetting Ctgf. Biosci Rep 37.

9

Patel, R.A.G., McMullen, P.W., 2017. Neuroprotection in the Treatment of Acute Ischemic

10

Stroke. Prog Cardiovasc Dis 59, 542-548.

11

Qu, F., Wang, X., 2017. microRNA-340 induces apoptosis by downregulation of BAG3 in

12

ovarian cancer SKOV3 cells. Pharmazie 72, 482-486.

13

Singh, P., Singh, A.P., Tripathi, S.K., Kumar, V., Sane, A.P., 2019. Petal abscission in roses is

14

associated with the activation of a truncated version of the animal PDCD4 homologue,

15

RbPCD1. Plant Sci 288, 110242.

16

Stegner, D., Klaus, V., Nieswandt, B., 2019. Platelets as Modulators of Cerebral

17

Ischemia/Reperfusion Injury. Front Immunol 10, 2505.

18

Tao, Z., Zhao, H., Wang, R., Liu, P., Yan, F., Zhang, C., Ji, X., Luo, Y., 2015. Neuroprotective

19

effect of microRNA-99a against focal cerebral ischemia-reperfusion injury in mice. J Neurol

20

Sci 355, 113-119.

21

Wang, J., Xu, Z., Chen, X., Li, Y., Chen, C., Wang, C., Zhu, J., Wang, Z., Chen, W., Xiao, Z.,

22

Xu, R., 2018. MicroRNA-182-5p attenuates cerebral ischemia-reperfusion injury by targeting 15

1

Toll-like receptor 4. Biochem Biophys Res Commun 505, 677-684.

2

Wang, Q., Zhu, J., Wang, Y.W., Dai, Y., Wang, Y.L., Wang, C., Liu, J., Baker, A., Colburn,

3

N.H., Yang, H.S., 2017. Tumor suppressor Pdcd4 attenuates Sin1 translation to inhibit

4

invasion in colon carcinoma. Oncogene 36, 6225-6234.

5

Xu, H., Cao, H., Zhu, G., Liu, S., Li, H., 2017. Overexpression of microRNA-145 protects

6

against rat myocardial infarction through targeting PDCD4. Am J Transl Res 9, 5003-5011.

7

Yan, H., Rao, J., Yuan, J., Gao, L., Huang, W., Zhao, L., Ren, J., 2017. Long non-coding RNA

8

MEG3 functions as a competing endogenous RNA to regulate ischemic neuronal death by

9

targeting miR-21/PDCD4 signaling pathway. Cell Death Dis 8, 3211.

10

Yang, L., Xiong, Y., Hu, X.F., Du, Y.H., 2015. MicroRNA-323 regulates ischemia/reperfusion

11

injury-induced neuronal cell death by targeting BRI3. Int J Clin Exp Pathol 8, 10725-10733.

12

Yoo, H., Kim, J., Lee, A.R., Lee, J.M., Kim, O.J., Kim, J.K., Oh, S.H., 2018. Alteration of

13

microRNA 340-5p and Arginase-1 Expression in Peripheral Blood Cells during Acute

14

Ischemic Stroke. Mol Neurobiol.

15

Yu, W., Zhang, G., Lu, B., Li, J., Wu, Z., Ma, H., Wang, H., Lian, R., 2016. MiR-340 impedes

16

the progression of laryngeal squamous cell carcinoma by targeting EZH2. Gene 577, 193-201.

17

Zennami, K., Choi, S.M., Liao, R., Li, Y., Dinalankara, W., Marchionni, L., Rafiqi, F.H.,

18

Kurozumi, A., Hatano, K., Lupold, S.E., 2019. PDCD4 Is an Androgen-Repressed Tumor

19

Suppressor that Regulates Prostate Cancer Growth and Castration Resistance. Mol Cancer

20

Res 17, 618-627.

21

Zhu, J., Yao, K., Wang, Q., Guo, J., Shi, H., Ma, L., Liu, H., Gao, W., Zou, Y., Ge, J., 2016.

22

Ischemic

Postconditioning-Regulated

miR-499 16

Protects

the

Rat

Heart

Against

1

Ischemia/Reperfusion Injury by Inhibiting Apoptosis through PDCD4. Cell Physiol Biochem

2

39, 2364-2380.

3 4

17

1

Figure legends

2

Figure 1. Downregulation of miR-340-5p in hippocampal neurons after

3

oxygen-glucose deprivation/reoxygenation (OGDR). (A) The qRT-PCR results of

4

miR-340-5p in primary hippocampal neurons after exposure to OGDR. (B) The

5

relative miR-340-5p abundance in OGDR-injured hippocampal neurons after

6

transfection with miR-340-5p mimics or miR-340-5p inhibitors for 24 h. N=3,

7

#

8

Figure 2. MiR-340-5p relieved OGDR-induced neuronal injury. (A) Cell viability

9

was detected using a CCK-8 assay. (B) LDH release in culture medium. (C) Apoptosis

10

analyzed by flow cytometry, apoptosis cell rate were represented in right upper

11

quadrant. (D) Western blot analysis of protein content of Bax and Bcl-2 in

12

hippocampal neurons. (E) Ratio of Bax/Bcl-2. N=3, #P<0.05 vs. control; *P<0.05 vs.

13

OGDR.

14

Figure 3. PDCD4 was a target of miR-340-5p. (A) Dual-Luciferase activity assay

15

results of the interaction between miR-340-5p and the 3’-UTR of PDCD4 in HEK293

16

cells. (B) Dual-Luciferase activity assay results in OGDR induced hippocampal

17

neurons. (C) The putative binding sequences of the 3’-UTR of PDCD4 with

18

miR-340-5p. (D and E) The protein content of PDCD4, tested by western blot. N=3,

19

#

20

Figure 4. MiR-340-5p alleviated injury to hippocampal neurons exposed to OGDR by

21

downregulating PDCD4. OGDR stimulated Hippocampal neurons were co-transfected

22

with miR-340-5p mimics and a PDCD4 vector. (A) Relative PDCD4 protein content.

23

(B) Cell viability. (C) LDH activity. (D) Apoptosis. (E) Relative protein content of

24

Bax and Bcl-2. (F) Ratio of Bax and Bcl-2. N=3, #P<0.05 vs. OGDR; *P<0.05 vs.

25

OGDR+miR-340-5p mimic.

26

Figure 5. MiR-340-5p alleviated injury to hippocampal neurons exposed to OGDR by

27

activating PI3K/Akt signaling pathway. (A) Western blot analysis of p-PI3K, PI3K,

28

p-Akt, and Akt protein content. (B) Quantitative analysis of relative protein content of

P<0.05 vs. control; *P<0.05 vs. OGDR.

P<0.05 vs. mimic control.

18

1

p-PI3K and PI3K. (C) Quantitative analyses of relative protein content of p-Akt and

2

Akt. N=3, #P<0.05 vs. control; *P<0.05 vs. OGDR.

3

Figure 6. MiR-340-5p activated PI3K/Akt signaling pathway by targeting PDCD4 in

4

OGDR induced neurons. (A) Western blot analysis of p-PI3K, PI3K, p-Akt, and Akt

5

protein content. (B) Quantitative analysis of relative protein content of p-PI3K and

6

PI3K. (C) Quantitative analysis of relative protein levels of p-Akt and Akt. N=3,

7

#

P<0.05 vs. control; *P<0.05 vs. OGDR. (D) Schematic diagram.

19

Highlights 1.

Low expression of miR-340-5p in hippocampal neurons exposed to OGDR

2.

MiR-340-5p had protective effect against neuronal injury caused by OGDR

3.

MiR-340-5p directly regulated PDCD4 expression

4.

MiR-340-5p inhibited PDCD4 to protect hippocampal neurons against OGDR injury

5.

MiR-340-5p targeted PDCD4 to influence PI3K/Akt signaling

Yake Zheng: Design, Experiment perform, Writing original draft. Peng Zhao: Experiment perform, Data analyze Yajun Lian: Design, Supervision, Paper revise Shuang Li: Experiment perform, Data analyze Yuan Chen: Experiment perform, Paper revise Lihao Li: Experiment perform,