Open Access

Electro-acupuncture exerts beneficial effects against cerebral ischemia and promotes the proliferation of neural progenitor cells in the cortical peri-infarct area through the Wnt/β-catenin signaling pathway

  • Authors:
    • Bin Chen
    • Jing Tao
    • Yukun Lin
    • Ruhui Lin
    • Weilin Liu
    • Lidian Chen
  • View Affiliations

  • Published online on: September 2, 2015     https://doi.org/10.3892/ijmm.2015.2334
  • Pages: 1215-1222
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Electro-acupuncture (EA) is a novel therapy based on combining traditional acupuncture with modern electrotherapy, and it is currently being investigated as a treatment for ischemic stroke. In the present study, we aimed to investigate the mechanisms through which EA regulates the proliferation of neural progenitor cells (NPCs) in the cortical peri‑infarct area after stroke. The neuroprotective effects of EA on ischemic rats were evaluated by determining the neurological deficit scores and cerebral infarct volumes. The proliferation of the NPCs and the activation of the Wnt/β‑catenin signaling pathway in the cortical peri‑infarct area were examined. Our results revealed that EA significantly alleviated neurological deficits, reduced the infarct volume and enhanced NPC proliferation [nestin/glial fibrillary acidic protein (GFAP)‑double positive] in the cortex of rats subjected to middle cerebral artery occlusion (MCAO). Moreover, the Wnt1 and β‑catenin mRNA and protein levels were increased, while glycogen synthase kinase‑3 (GSK3) transcription was suppressed by EA. These results suggest that the upregulatory effects of EA on the Wnt/β‑catenin signaling pathway may promote NPC proliferation in the cortical peri-infarct area after stroke, consequently providing a therapeutic effect against cerebral ischemia.

Introduction

Stroke is the leading cause of adult disability, as well as the second most common cause of mortality worldwide (1). Ischemic stroke accounts for approximately 80% of all strokes (1,2). Over the past decade, thrombolysis has been established as an effective treatment in the most acute phase of ischemic stroke. However, many patients develop lifelong disabilities following ischemic stroke as they do not receive the necessary treatment within the therapeutic time window. Therefore, increasing the window of therapeutic efficacy of established treatments or identifying other therapies with alternative targets is necessary to improve the neurological outcomes in ischemic stroke victims. Targeting key cellular survival/proliferation mechanisms may improve prognosis. Recently, it was reported that the signaling molecule, β-catenin, is degraded in the peri-infarct area of the brain following focal cerebral ischemia (37). In a previous study, it was demonstrated that in doubleridge mice, which have a reduced expression of Dkk-1, an antagonist of Wnt/β-catenin signaling, the reduction of β-catenin was attenuated and the infarct volume was reduced following middle cerebral artery occlusion (MCAO) (5). This suggests that prevenint the decrease in Wnt/β-catenin signaling in cerebral ischemia may prove to be a potential novel therapeutic modality.

In addition, the conditional expression of stabilized β-catenin in neural progenitor cells (NPCs) enlarges the cortical surface area through the expansion of the progenitor cell population (8). The overexpression of Wnt3 has been shown to increase neurogenesis in adult hippocampal precursor cells, while the blockade of Wnt signaling reduces neurogen-esis both in vitro and in vivo (9). These studies also implicate the canonical Wnt/β-catenin pathway in the proliferation and self-renewal of NPCs (3,8,9).

Traditional Chinese medicine (TCM) has long been an important component of complementary and alternative medicine in several Asian countries, and recently in Western society. Modern research has revealed the potential therapeutic effects of TCM in the treatment of various diseases, including cerebrovascular diseases and cancer (10,11). Its unique functions in gene therapy have also been discussed (12,13). Electro-acupuncture (EA) is a traditional therapeutic method used in China, widely used for both the prevention and treatment/rehabilitation of cerebral ischemia. Nevertheless, the mechanisms responsible for its effects are not yet fully understood. Previous studies have indicated that EA significantly attenuates neurological deficits, and reduces infarct volume and mortality in both animal models of stroke and in patients suffering from stroke when administered at appropriate acupoints with suitable stimulation parameters (1419). Two specific acupoints, Quchi (LI11) and Zusanli (ST36), are one of the most effective prescriptions commonly used in EA treatment of ischemic stroke (17,20). Preliminary data have demonstrated that EA at these two acupoints significantly promotes NPC proliferation following cerebral ischemia in the subventricular zone (SVZ) of the lateral ventricle, and in the subgranular zone (SGZ) of the hippocampus (17,20). A growing body of evidence suggests that cortex-derived neural stem/progenitor cells may contribute to the repair of ischemic lesions of the cerebral cortex (2123). Based on these and other findings, the elucidation of the Wnt signaling mechanisms underlying the promoting effects of EA on NPC proliferation in the cortical peri-infarct area after stroke, is an important step toward validating the clinical application and benefits of this treatment modality in the treatment of ischemic stroke.

Materials and methods

Materials and reagents

TRIzol reagent was purchased from Life Technologies (Carlsbad, CA, USA). The RevertAid™ First Strand cDNA Synthesis kit and Taq DNA Polymerase were purchased from Fermentas (Hanover, MD, USA). Primary antibodies against glial fibrillary acidic protein (GFAP, a marker for reactive astrocytes; #3670), glycogen synthase kinase-3 (GSK3; #5676) and β-actin (#4970), and horseradish peroxidase (HRP)-conjugated secondary antibodies (anti-mouse, #7076; anti-rabbit, #4970) were all obtained from Cell Signaling Technology, Inc. (Beverly, MA, USA). Anti-microtubule-associated protein 2 (MAP2, a marker of neurons; ab32454), anti-nestin (a marker of progenitor cells and astrocytes; 2Q178) and anti-β-catenin (ab22656) primary antibodies were all obtained from Abcam (Cambridge, MA, USA). All other chemicals, unless otherwise stated, were obtained from Sigma-Aldrich (St. Louis, MO, USA).

Animals and groups

Adult male Sprague-Dawley rats (weighing 250–280 g) were obtained from Shanghai SLAC Laboratory Animal Co., Ltd. (Shanghai, China). All experiments were performed strictly in accordance with the International Ethical Guidelines and the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

A total of 72 rats were randomly divided into 4 groups (18 rats in each group) as follows: i) sham-operated (sham) group; ii) MCAO group; iii) MCAO + EA group: ischemic rats treated with EA at the Quchi (LI11) and Zusanli (ST36) acupoints; and ⅳ) sham + EA group: sham-operated rats treated with EA.

Induction of focal cerebral ischemia

A rat model of focal cerebral ischemia/reperfusion (I/R) was utilized in this study. The left middle cerebral artery (MCA) was occluded by the placement of an embolus at the origin of the MCA, as previously described (24). Following anesthetization with 10% chloral hydrate (300 mg/kg), each rat was placed in the prone position. A midline incision was made on the dorsal surface of the skull, and the skull was thinned with a burr hole over the left parietal cortex (5 mm lateral and 1 mm posterior to the bregma) without injury to the dura mater. The laser Doppler perfusion monitor (LDF100C; Biopac Systems, Inc., Goleta, CA, USA) was attached to the skull with dental cement. With the rat in a supine position, MCAO was performed via ligation of the left common carotid artery (CCA) and external carotid artery (ECA) and closure of the internal carotid artery (ICA). The embolus was gently advanced within the left ICA to the origin of the MCA, until a slight resistance was encountered (20±2 mm). Cerebral blood flow was measured beginning 5 min prior to the induction of occlusion. Ischemic rats that showed a stable drop of 80% in blood perfusion units (BPU) compared with the baseline level (before MCAO), were used in the subsequent experiments. Reperfusion was achieved by removing the intraluminal occlusive embolus to restore blood supply to the MCA area 2 h later. Animals subjected to sham operation were treated in a similar manner, but without ligations and occlusions.

Neurological assessment

Neurological deficits were assessed to confirm successful MCAO. A neurological score was assigned to each animal 2 h following I/R, in a blinded manner, according to a well-established 5-point neurological scale (24): score 0, no apparent deficits; 1, failure to fully extend the right forepaw; 2, circling to the right; 3, falling or leaning over to the right; 4, no spontaneous walking and a depressed level of consciousness; and 5, dead. Rats subjected to MCAO with neurological deficit scores of 1–3 were used in the subsequent experiments.

Treatment wtih EA

EA was applied at the LI11 (Quchi, in the depression lateral to the anterior aspect of the radius joint of the forelimb) and ST36 (Zusanli, 5 mm below the head of fibula under the knee joint and 2 mm lateral to the anterior tibial tubercle) acupoints on the right paralyzed limb using an EA stimulation instrument [Model G6805; Shanghai Marine Instrument General Factory (SMIF), Shanghai, China]. Two stainless steel acupuncture needles, 0.3 mm in diameter, connected to the output terminals of the EA stimulation instrument, were inserted at a depth of 2–3 mm at the LI11 and ST36 acupoints. The acupoints were stimulated with disperse-dense waves of 1 or 20 Hz frequencies for 30 min, once a day, and the current intensity was maintained slightly below the level that induced visible muscle contraction. Treatment commenced on the day following the operation and continued daily until the animals were sacrificed.

Measurement of cerebral infarct volume

Three days following cerebral I/R injury, the rats were euthanized under deep anesthesia using 10% chloral hydrate and perfused transcardiacally with 0.9% NaCl. The brains of all the rats were rapidly removed and sliced into 5 coronal blocks at a thickness of 2 mm per section. The fresh slices were incubated in 2% (w/v) 2,3,5-triphenyltetrazolium chloride (TTC; Sigma-Aldrich) solution in phosphate-buffered saline (PBS; HyClone, Beijing, China) for 20 min, at 37°C in the dark. Images of the 5 sections were captured using a high-resolution digital camera (PowerShot SX20 IS; Canon) and examined by a blinded observer to determine the infarct size using computerized image analysis software (Motic Med 6.0 system; Motic China Group Co.,. Ltd., Shenzhen, China). The infarct volume data are expressed as a percentage of the total brain volume. Lesion volume was estimated using an indirect method to avoid the effects of tissue swelling or shrinkage: 100× (contralateral hemisphere volume - non-infarct ipsilateral hemisphere volume)/contralateral hemisphere volume, as previously described (25).

Tissue preparation

The rats were anesthetized with 10% chloral hydrate and intracardially perfused with chilled saline followed by 0.01 M PBS containing 4% paraformaldehyde through the left ventricular lumen of the heart. The brains were collected and post-fixed in 4% paraformaldehyde at 4°C overnight, and then embedded in paraffin. Coronal sections were cut into 5-µm-thick sections, and used for immunofluoresence staining. For western blot analysis RT-PCR, the ischemic boundary zones were extracted from the ischemic brains and prepared accordingly.

Immunofluorescence staining

The brain sections were processed by immunofluorescence staining using several specific cell markers (MAP2, neurons; GFAP, astrocytes; nestin, NPC and astrocytes). Coronal sections (5-µm-thick) were de-paraffinized in dimethylbenzene, hydrated successively in gradient ethanols, and antigens were retrieved twice in 0.1 M citrate buffer (pH 6.0). The sections were blocked in blocking buffer (10% normal goat serum, 0.3% Triton X in PBS) for 1 h at room temperature, then incubated with primary antibodies at 4°C overnight. After washing in 0.01 M PBS, the sections were incubated for 2 h at room temperature with a combination of goat anti-mouse IgG H&L (FITC; ab6785; Abcam) and goat anti-rabbit IgG H&L (TRITC; ab6718; Abcam) secondary antibodies. The sections were stained with DAPI (Vector Laboratories, Burlingame, CA, USA) to localize the nuclei and were coverslipped for observation. The labelled sections were visualized and imaged using a confocal microscope (LSM710 META NLO; Carl Zeiss, Oberkochen, Germany).

Western blot analysis

The left cerebral tissues were dissected out and homogenized in RIPA buffer containing Protease Inhibitor Cocktail (Roche Applied Science, Mannheim, Germany) and PMSF. The samples were kept on ice for 30 min and the insoluble material was removed by centrifugation at 14,000 × g for 15 min. The protein concentration was quantified by BCA assay (Pierce Biotechnology, Inc., Rockford, IL, USA). Brain homogenates (50 µg) were separated by SDS-PAGE and transferred onto PVDF membranes. The membranes were subsequently blocked for 2 h with 5% non-fat powdered milk in Tris-buffered saline containing 0.1% Tween-20 (TBST) and then incubated overnight at 4°C with appropriate primary antibodies: GFAP, Wnt1 (SAB2102711; Sigma-Aldrich), GSK3, β-catenin and β-actin (at a dilution of 1:1,000). The membranes were then washed with TBST followed by incubation with the appropriate HRP-conjugated secondary antibody for 1–2 h at room temperature. Normalization of the results was ensured by running parallel western blot analyses with β-actin antibody. The optical density was quantified using a Bio-Image Analysis System (Bio-Rad, Hercules, CA, USA), with the value of the sham-operated group designated as 1.0.

RNA extraction and RT-PCR

Total RNA was extracted using TRIzol Reagent (Life Technologies). The RNA concentrations were determined by OD260/280 readings using a GeneQuant spectrophotometer (Amersham Biosciences, Amersham, UK). The oligo(dT)-primed RNA (3 µg) was reverse-transcribed using the RevertAid™ First Strand cDNA Synthesis kit (Fermentas, Chicago, IL, USA) according to the manufacturer's instructions. Semi-quantitative PCR was performed to measure the Wnt1, GSK3, β-catenin and β-actin mRNA expression levels. The primer sequences used for each gene are listed in Table I. The samples were analyzed by gel electrophoresis (1.5% agarose). The DNA bands were examined using a Gel Documentation System (Model Gel Doc 2000; Bio-Rad), with the value of the sham-operated group designated as 1.0.

Table I

Primer sequences used for PCR.

Table I

Primer sequences used for PCR.

Gene namesForwardReverse
Wnt15′-CAG TGG AGC AAC GGT ATG AG-3′5′-TTC TTC CCT GCC TTG ATG T-3′
GSK35′-AGA CCA AAA TCA TCT ACC AC-3′5′-ACT CTG TGC CTG TCT CAT-3
β-catenin5′-CAT CCT TAT CCC TCC TCA CGC-3′5′-TTA TTG GTC TGT CCA CGG TCT-3″
β-actin5′-CGG GAG AAC AGG GTA TGA-3′5′-CAG GCT GGA AGG AGA AGA T-3′

[i] GSK3, glycogen synthase kinase-3.

Cell quantification and statistical analysis

The infarct area was defined by tissue autofluorescence, while the peri-infarct area was defined by the presence of MAP2-positive immunofluorescent cells. Cell quantification in the cortical peri-infarct area was performed by observers blinded to the sample identity using Image-Pro Plus 6.0 software. The results are expressed as the number of MAP2-positive cells/cm2. All data were analyzed using the SPSS package for Windows (version 16.0) and are presented as the means ± standard error of the mean (SEM). Statistical data analysis was performed with the unpaired Student's t-test, the Mann-Whitney U test or ANOVA. Differences with P<0.05 were considered statistically significant.

Results

EA alleviates neurological deficits and reduces infarct volume after stroke

Compared with the rats in the sham-operated and sham + EA groups, which did not present with any signs of cerebral injury, all rats in both the MCAO and MCAO + EA groups demonstrated obvious manifestations of neurological deficits and cerebral infarction (Fig. 1 and Table II). There were no statistically significant differences observed between the MCAO and MCAO + EA groups at 2 h after cerebral I/R injury. However, EA administered at the Zusanli and Quchi acupoints for 3 days significantly improved neurological deficits (MCAO, 2.08±0.23; MCAO + EA, 1.42±0.19; P<0.05) (Table II), and decreased the cerebral infarct volume (MCAO, 35.39±1.56%; MCAO + EA, 22.39±2.50%; P<0.01) (Fig. 1), demonstrating the therapeutic efficacy of EA against cerebral I/R injury.

Table II

Assessment of neurological deficits.

Table II

Assessment of neurological deficits.

Group2 h after I/R3 days after I/R
Sham00
MCAO2.33±0.192.08±0.23
MCAO + EA2.42±0.191.42±0.19a
Sham + EA00

{ label (or @symbol) needed for fn[@id='tfn2-ijmm-36-05-1215'] } Data shown are the means ± SEM from 18 individual rats in each group.

a P<0.05 vs. the MCAO group. Sham, sham operation, controls; MCAO, middle cerebral artery occlusion; EA, electro-acupuncture; I/R, ischemia/reperfusion; SEM, standard error of the mean.

EA accelerates the proliferation of GFAP-positive reactive astrocytes in the cortical peri-infarct area after stroke

The border of the infarct core was defined by MAP2 staining (Fig. 2), and the subsequent results identified a few GFAP-positive cells in the non-ischemic cortex (Fig. 3E and H); however, the majority of GFAP-positive cells were observed in the post-stroke cortex, specifically in the peri-infarct area (Fig. 3F and G). Western blot analysis also revealed a significant increase in GFAP expression in the MCAO and MCAO + EA groups within the post-stroke cortex, compared with the comparable MCA area in the sham-operated (sham) and sham + EA groups (sham, 1±0; MCAO, 1.95±0.24; MCAO + EA, 3.10±0.44; and sham + EA, 0.97±0.07; P<0.01 vs.sham and sham + EA groups; Fig. 3I and J). In addition, overall GFAP expression was significantly higher in the MCAO + EA group than in the MCAO group (P<0.05), suggesting that treatment with EA promoted the proliferation of GFAP-positive reactive astrocytes in the cortical peri-infarct area after stroke.

EA enhances the proliferation of NPCs in the cortical peri-infarct area after stroke

The presence of nestin/GFAP-positive cells within the post-stroke cortex was investigated in order to assess the generation of injury-induced NPCs. Previous studies have reported that nestin and GFAP-positive cells can acquire stem cell activity in the cortical peri-infarct area after stroke (21,22). In the present study, a nestin-positive subpopulation of NPCs formedon the ipsilateral, but not the contralateral side of the brain after stroke. At 3 days after stroke, the number of nestin-positive cells was significantly higher in the MCAO + EA group than in the MCAO group. Similarly, the number of nestin/GFAP-positive cells was significantly higher in the MCAO + EA group (MCAO, 257.72±49.73; MCAO + EA, 379.56±20.05; P<0.05; Fig. 4), demonstrating that EA potentially increased the proliferation of the NPCs in the cortical peri-infarct area after stroke.

EA regulates the activation of the Wnt pathway in the cortical peri-infarct area after stroke

To examine the effects of EA on the Wnt signaling pathway, we measured the protein and mRNA levels of Wnt1, GSK3 and β-catenin in the ischemic cortex by western blot analysis and RT-PCR. As shown in Fig. 5, focal cerebral I/R injury significantly reduced the expression of Wnt1 and β-catenin, while the transcription of GSK3 was significantly increased in the ischemic cortex at 3 days following stroke when compared to the sham and sham + EA groups (P<0.05). Of note, the decrease in the expression of Wnt1 and β-catenin was circumvented by treatment with EA, and the upregulated transcription of GSK3 was significantly reduced following treatment with EA (P<0.05). Taken together, these results suggest that EA applied at the Quchi and Zusanli acupoints significantly promotes the activation of the Wnt signaling pathway in the peri-infarct cortex.

Discussion

In response to stroke, subpopulations of cortical reactive astrocytes proliferate and express several proteins commonly associated with neural stem/progenitor cells, such as GFAP, nestin and RC2 (2123). Shimada et al (21) demonstrated that GFAP-expressing reactive astrocytes can be isolated from the cortical peri-infarct area 3 days after stroke, and de-differentiated into reactive astrocyte-derived neural stem/progenitor cells with self-renewal and multipotent properties when grown under neurosphere conditions. Lineage tracing identified reactive astrocytes as a cell of origin for neural stem cells (NSCs) derived from cortical peri-infarct tissues after stroke (21).

In this study, in order to investigate the effects of EA on NPC proliferation via Wnt signaling, we treated sham-operated and rats subjected to MCAO with electric stimulation at the Quchi (LI11) and Zusanli (ST36) acupoints on the contralateral paralyzed limb. Our results revealed that EA applied at these acupoints 1 day following cerebral I/R injury and once daily for 3 consecutive days, significantly improved neurological function (MCAO, 2.08±0.23; MCAO + EA, 1.42±0.19; P<0.05) and attenuated the increase in the cerebral infarct volume (MCAO, 35.39±1.56%; MCAO + EA, 22.39±2.50%; P<0.01) induced by MCAO. Our findings corroborate those of previous studies that used a model of transient focal cerebral ischemia to demonstrate the therapeutic efficacy of EA (1417). Furthermore, immunofluorescence staining was performed to observe several markers associated with the activation of NPCs. Our results revealed that the number of proliferating nestin/GFAP-positive NPCs was significantly increased within the post-stroke cortex in the MCAO group (257.72±49.73) (Fig. 4) but even more so in the MCAO + EA group (379.56±20.05, P<0.05 vs. MCAO group), suggesting that EA promoted the proliferation of neural stem/progenitor cells in rats subjected to MCAO. The emergence of NPCs in the peri-infarct area is a documented event in the brain after stroke (2123). However, the signaling pathway controlling the proliferation of these NPCs is poorly defined.

Of note, previous studies have indicated that Wnt/β-catenin signaling is critically involved in the regulation of the proliferation and differentiation of NPCs (3,8,9). The Wnt/β-catenin pathway is activated when a Wnt ligand binds to its seven-trans-membrane receptors, the Frizzled proteins. The activation of the Wnt pathway inhibits GSK-3β, which results in the cytoplasmic accumulation of β-catenin. Stabilized β-catenin then translocates into the nucleus and interacts with the transcription factors TCF/Lef to activate downstream genes such as cyclin D1 and c-myc (26,27).

Cerebral ischemia profoundly reduced the transcription of Wnt1 and β-catenin and increased the expression of GSK3. Treatment with EA reversed these effects (Fig. 5). Moreover, this is consistent with the results of previous reports that the Wnt pathway is markedly degraded after stroke (35). In a previous study, when assessed at 3 days following an endothelin-1 (Et-1) injection, treatment with lithium ions prevented the decrease in the expression of β-catenin in the ischemic cortex (5). In this study, the treatment of NPCs with EA significantly increased the expression of Wnt1 and β-catenin, while inhibiting the transcription of GSK3. These data indicate that EA applied at the Quchi (LI11) and Zusanli (ST36) acupoints promoted the proliferation of NPCs in the cortical peri-infarct area via the Wnt/β-catenin pathway.

In conclusion, the results of the present study strongly suggest that treatment with EA provides robust protection against transient cerebral ischemic injury and promotes the proliferation of neural stem/progenitor cells in response to isch-emia via the Wnt/β-catenin pathway. Our data are supported by evidence in the current literature. These results may provide a theoretical and experimental basis for the future clinical application of EA and its potential use in the treatment of cerebral ischemia.

However, even with optimal stimulation parameters, treatment with EA targets multiple mechanisms in order to achieve its protective effects against ischemic insults. Therefore, the precise mechanisms of action associated with this treatment the reparative process in the post-ischemic brain requires further investigation.

Acknowledgments

This study was sponsored by the National Natural Science Foundation of China (grant nos. 81273835 and 81373778). We would like to thank Clarity Manuscript Consultants, LLC, for their assistance in the editing of this manuscript.

Abbreviations:

EA

electro-acupuncture

NPCs

neural progenitor cells

MCAO

middle cerebral artery occlusion

SVZ

subventricular zone

SGZ

subgranular zone

TTC

2,3,5-triphenyltetrazolium chloride

GFAP

glial fibrillary acidic protein

MAP2

microtubule-associated protein 2

CCA

common carotid artery

ECA

external carotid artery

ICA

internal carotid artery

References

1 

Donnan GA, Fisher M, Macleod M and Davis SM: Stroke. Lancet. 371:1612–1623. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Roger VL, Go AS, Lloyd-Jones DM, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS, et al American Heart Association: Statistics Committee and Stroke Statistics Subcommittee: Heart disease and stroke statistics - 2012 update: a report from the American Heart Association. Circulation. 125. pp. e2–e220. 2012, View Article : Google Scholar

3 

Hirabayashi Y, Itoh Y, Tabata H, Nakajima K, Akiyama T, Masuyama N and Gotoh Y: The Wnt/beta-catenin pathway directs neuronal differentiation of cortical neural precursor cells. Development. 131:2791–2801. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Zhang H, Ren C, Gao X, Takahashi T, Sapolsky RM, Steinberg GK and Zhao H: Hypothermia blocks beta-catenin degradation after focal ischemia in rats. Brain Res. 1198:182–187. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Mastroiacovo F, Busceti CL, Biagioni F, Moyanova SG, Meisler MH, Battaglia G, Caricasole A, Bruno V and Nicoletti F: Induction of the Wnt antagonist, Dickkopf-1, contributes to the development of neuronal death in models of brain focal ischemia. J Cereb Blood Flow Metab. 29:264–276. 2009. View Article : Google Scholar

6 

Scott EL and Brann DW: Estrogen regulation of Dkk1 and Wnt/β-Catenin signaling in neurodegenerative disease. Brain Res. 1514:63–74. 2013. View Article : Google Scholar :

7 

Sun FL, Wang W, Zuo W, Xue JL, Xu JD, Ai HX, Zhang L, Wang XM and Ji XM: Promoting neurogenesis via Wnt/β-catenin signaling pathway accounts for the neurorestorative effects of morroniside against cerebral ischemia injury. Eur J Pharmacol. 738:214–221. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Pöschl J, Grammel D, Dorostkar MM, Kretzschmar HA and Schüller U: Constitutive activation of β-catenin in neural progenitors results in disrupted proliferation and migration of neurons within the central nervous system. Dev Biol. 374:319–332. 2013. View Article : Google Scholar

9 

Lie DC, Colamarino SA, Song HJ, Désiré L, Mira H, Consiglio A, Lein ES, Jessberger S, Lansford H, Dearie AR and Gage FH: Wnt signalling regulates adult hippocampal neurogenesis. Nature. 437:1370–1375. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Zhai XF, Chen Z, Li B, Shen F, Fan J, Zhou WP, Yang YK, Xu J, Qin X, Li LQ and Ling CQ: Traditional herbal medicine in preventing recurrence after resection of small hepatocellular carcinoma: a multicenter randomized controlled trial. J Integr Med. 11:90–100. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Liang J, Li F, Wei C, Song H, Wu L, Tang Y and Jia J: Rationale and design of a multicenter, phase 2 clinical trial to investigate the efficacy of traditional Chinese medicine SaiLuoTong in vascular dementia. J Stroke Cerebrovasc Dis. 23:2626–2634. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Wang LN, Wang Y, Lu Y, Yin ZF, Zhang YH, Aslanidi GV, Srivastava A, Ling CQ and Ling C: Pristimerin enhances recombinant adeno-associated virus vector-mediated transgene expression in human cell lines in vitro and murine hepatocytes in vivo. J Integr Med. 12:20–34. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Ling CQ, Wang LN, Wang Y, Zhang YH, Yin ZF, Wang M and Ling C: The roles of traditional Chinese medicine in gene therapy. J Integr Med. 12:67–75. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Liu H, Shen X, Tang H, Li J, Xiang T and Yu W: Using microPET imaging in quantitative verification of the acupuncture effect in ischemia stroke treatment. Sci Rep. 3:10702013. View Article : Google Scholar : PubMed/NCBI

15 

Kim JH, Choi KH, Jang YJ, Bae SS, Shin BC, Choi BT and Shin HK: Electroacupuncture acutely improves cerebral blood flow and attenuates moderate ischemic injury via an endothelial mechanism in mice. PLoS One. 8:e567362013. View Article : Google Scholar : PubMed/NCBI

16 

Jin Z, Liang J, Wang J and Kolattukudy PE: Delayed brain ischemia tolerance induced by electroacupuncture pretreatment is mediated via MCP-induced protein 1. J Neuroinflammation. 10:632013. View Article : Google Scholar : PubMed/NCBI

17 

Tao J, Xue XH, Chen LD, Yang SL, Jiang M, Gao YL and Wang XB: Electroacupuncture improves neurological deficits and enhances proliferation and differentiation of endogenous nerve stem cells in rats with focal cerebral ischemia. Neurol Res. 32:198–204. 2010. View Article : Google Scholar

18 

Mazighi M, Meseguer E, Labreuche J and Amarenco P: Bridging therapy in acute ischemic stroke: a systematic review and meta-analysis. Stroke. 43:1302–1308. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Wu P, Mills E, Moher D and Seely D: Acupuncture in post-stroke rehabilitation: a systematic review and meta-analysis of randomized trials. Stroke. 41:e171–e179. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Tao J, Chen B, Gao Y, Yang S, Huang J, Jiang X, Wu Y, Peng J, Hong Z and Chen L: Electroacupuncture enhances hippocampal NSCs proliferation in cerebral ischemia-reperfusion injured rats via activation of notch signaling pathway. Int J Neurosci. 124:204–212. 2014. View Article : Google Scholar

21 

Shimada IS, LeComte MD, Granger JC, Quinlan NJ and Spees JL: Self-renewal and differentiation of reactive astrocyte-derived neural stem/progenitor cells isolated from the cortical peri-infarct area after stroke. J Neurosci. 32:7926–7940. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Nakagomi T, Taguchi A, Fujimori Y, Saino O, Nakano-Doi A, Kubo S, Gotoh A, Soma T, Yoshikawa H, Nishizaki T, et al: Isolation and characterization of neural stem/progenitor cells from post-stroke cerebral cortex in mice. Eur J Neurosci. 29:1842–1852. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Nakagomi T, Molnár Z, Nakano-Doi A, Taguchi A, Saino O, Kubo S, Clausen M, Yoshikawa H, Nakagomi N and Matsuyama T: Ischemia-induced neural stem/progenitor cells in the pia mater following cortical infarction. Stem Cells Dev. 20:2037–2051. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Longa EZ, Weinstein PR, Carlson S and Cummins R: Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke. 20:84–91. 1989. View Article : Google Scholar : PubMed/NCBI

25 

Swanson RA, Morton MT, Tsao-Wu G, Savalos RA, Davidson C and Sharp FR: A semiautomated method for measuring brain infarct volume. J Cereb Blood Flow Metab. 10:290–293. 1990. View Article : Google Scholar : PubMed/NCBI

26 

He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B and Kinzler KW: Identification of c-MYC as a target of the APC pathway. Science. 281:1509–1512. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Shtutman M, Zhurinsky J, Simcha I, Albanese C, D'Amico M, Pestell R and Ben-Ze'ev A: The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci USA. 96:5522–5527. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2015
Volume 36 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen B, Tao J, Lin Y, Lin R, Liu W and Chen L: Electro-acupuncture exerts beneficial effects against cerebral ischemia and promotes the proliferation of neural progenitor cells in the cortical peri-infarct area through the Wnt/β-catenin signaling pathway. Int J Mol Med 36: 1215-1222, 2015
APA
Chen, B., Tao, J., Lin, Y., Lin, R., Liu, W., & Chen, L. (2015). Electro-acupuncture exerts beneficial effects against cerebral ischemia and promotes the proliferation of neural progenitor cells in the cortical peri-infarct area through the Wnt/β-catenin signaling pathway. International Journal of Molecular Medicine, 36, 1215-1222. https://doi.org/10.3892/ijmm.2015.2334
MLA
Chen, B., Tao, J., Lin, Y., Lin, R., Liu, W., Chen, L."Electro-acupuncture exerts beneficial effects against cerebral ischemia and promotes the proliferation of neural progenitor cells in the cortical peri-infarct area through the Wnt/β-catenin signaling pathway". International Journal of Molecular Medicine 36.5 (2015): 1215-1222.
Chicago
Chen, B., Tao, J., Lin, Y., Lin, R., Liu, W., Chen, L."Electro-acupuncture exerts beneficial effects against cerebral ischemia and promotes the proliferation of neural progenitor cells in the cortical peri-infarct area through the Wnt/β-catenin signaling pathway". International Journal of Molecular Medicine 36, no. 5 (2015): 1215-1222. https://doi.org/10.3892/ijmm.2015.2334