Electroacupuncture promotes neurological functional recovery via the retinoic acid signaling pathway in rats following cerebral ischemia-reperfusion injury

  • Authors:
    • Jiangcong Hong
    • Guangwen Wu
    • Yulong Zou
    • Jing Tao
    • Lidian Chen
  • View Affiliations

  • Published online on: October 26, 2012     https://doi.org/10.3892/ijmm.2012.1166
  • Pages: 225-231
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Neurogenesis is regulated by a number of signaling pathways, including the retinoic acid (RA) pathway, a key regulator of neurogenesis in the subventricular zone (SVZ) and hippocampus. Acupuncture has been used to treat neurological conditions and is known to potentially enhance cell proliferation in the neurogenic area (hippocampal dentate gyrus and the SVZ of the lateral ventricle walls) in pathological conditions, which is associated with improved brain function. However, whether or not the neuroprotective effects of electroacupuncture (EA) are mediated by the regulation of the RA signaling pathway remains to be determined. Using a transient middle cerebral artery occlusion model, in the present study we evaluated the effect of EA on the neurological functional recovery, infarction volume and investigated the underlying molecular mechanisms. Two hundred and sixteen SD rats were randomly divided into 3 groups: sham, model group (ischemic rats without EA stimulation) and EA group (ischemic rats with EA stimulation on ST36 and LI11). Behavioral deficits were detected with high-resolution digital analysis of 24-h home-cage video recordings. Infarct volume was determined by triphenyltetrazolium hydrochloride staining and the expression of RA mRNA and protein was measured using RT-PCR and western blotting, respectively. We found that EA decreased the infarct volume, promoted neurological functional recovery and increased the RA mRNA and protein expression, compared with the model group. Findings of this study suggest that promoting neurological functional recovery by modulating RA expression in the post-ischemic brain is one of the mechanisms by which EA can be effective in the treatment of ischemic stroke.

Introduction

Cerebral ischemia is considered a major cause of morbidity and mortality worldwide. Excitotoxicity is a major pathophysiological mechanism associated with stroke-induced inflammation and oxidative stress-associated disruption of the blood-brain barrier, resulting in enhancing neuronal cell death during ischemic stroke (1). Recombinant tissue plasminogen activator (t-PA) is the preferred treatment for acute ischemic stroke, however, due to a limited time window few patients are able to receive this therapy. Furthermore, t-PA treatment is likely to increase the risk of hemorrhage (2). Therefore, development of alternative therapies for ischemic stroke is important.

Mounting evidence indicates that potential neural stem/progenitor cells are present in various brain regions outside of the subventricular zone (SVZ) and subgranular zone of the hippocampal dentate gyrus (3,4). Stroke-induced neurogenesis has also been demonstrated in the adult human brain, even in patients of advanced age (57), indicating potential involvement of neural stem cells (8) and neurogenesis-regulating factors (9). Various factors regulate neurogenesis during development. One developmental molecule of particular interest in this regard is retinoic acid (RA), which is involved in the normal development of the central nervous system (10) and is also critical in the adult brain (11,12). RA expression and signaling continues in the postnatal and adult brain, stimulating neonatal SVZ and adult hippocampal neurogenesis (1316). The hypothesis that RA is crucial in the transcriptional model after nerve injury is supported by indirect evidence regarding the regulation of trauma-related genes, by observations of retinoid receptors and binding proteins after nerve injury and by dorsal root ganglia, retinal and spinal cord explant cultures (17). Retinoid-binding proteins are expressed in the SVZ-olfactory bulb pathway and RA receptors persist in the olfactory bulb (18). Transplantation of neural cells obtained from RA-treated cynomolgus monkey embryonic stem cells successfully enhanced the motor function of hemiplegic mice with experimental brain injury (19).

Acupuncture has been used to treat neurologic conditions, and acupuncture reportedly enhances cell proliferation in the neurogenic area (hippocampal dentate gyrus and the SVZ of the lateral ventricle walls) in pathological conditions (2022), which is associated with enhanced brain function (23,24). The LI11 (Quchi) and ST36 (Zusanli) acupoints have been selected for brain injury rehabilitation. Electroacupuncture (EA) at these acupoints has the potential to stimulate cell proliferation and reduce brain injury (20,21,2527). However, the mechanisms underlying the effects of acupuncture are poorly understood. Thus, whether or not the neuroprotective effects of EA are mediated by regulation of the RA signaling pathway should be investigated.

To extend the clinical observations of the potential neuroprotective effect of EA and help to establish a scientific foundation for further research, the effect of EA on the infarct volume, neurological functional recovery, and the expression of RA mRNA and protein were examined in this study. EA was found to decrease the infarct volume, promote neurological functional recovery and regulate the expression of RA mRNA and protein. This finding suggests that promoting neurological functional recovery through modulating RA expression in the post-ischemic brain is one of the mechanisms by which EA can be effective in the treatment of ischemic stroke.

Materials and methods

Reagents

The Raldh1, Raldh2 and β-actin primers were purchased from Sangon Biotech (Shanghai) Co., Ltd. (Shanghai, China). Raldh1 and Raldh2 antibodies, horseradish peroxidase (HRP)-conjugated secondary antibodies and the antibody against β-actin were obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA). Any other chemicals used, unless otherwise stated, were obtained from Sigma Chemical Company (St. Louis, MO, USA).

Animals

Healthy and pathogen-free male Sprague-Dawley rats (n=216), weighing 220–280 g, were purchased from SLAC Laboratory Animal Co., Ltd., Shanghai, China (Laboratory Animal Use Certificate no. SCXK(SH)2007000509570) and raised in a sterile environment. The care and use of the laboratory animals complied with the Guidance Suggestions for the Care and Use of Laboratory Animals, enforced by the Ministry of Science and Technology, China (2006) (28).

Animal model of transient middle cerebral artery occlusion

Animals were housed in a 12-h reverse light-dark cycle and were provided with food and water ad libitum. Rats were anesthetized with 10% chloral hydrate (3.5 ml/kg body weight). Body temperature was maintained using a 37°C water-recirculating pad. Transient middle cerebral artery occlusion (tMCAO) was produced for 120 min using the external carotid artery insertion method as described previously (29). Briefly, the left common carotid artery was exposed through a midline incision, and the internal and external carotid arteries were separated. A 3.0-mm nylon monofilament with a tip rounded by heat was placed in the external carotid artery and advanced through the internal carotid artery until resistance occurred. The monofilament was left in place for 120 min and then removed under anesthesia. Animals were observed until recovery from anesthesia. The sham group was treated in the same way, although tMCAO was not induced. This protocol produced infarcts involving the striatum and cortex, with a mortality rate of ~30%. The rats were subsequently randomized into 3 experimental groups: EA group (n=72), model group (n=72) and sham group (n=72). The rats were tested at 1, 7, 14 and 28 days subsequent to tMCAO. Eighteen rats were decapitated at each time point in each experimental group, after the neurological function was tested. Six rats were used for triphenyltetrazolium hydrochloride (TTC) staining, 6 rats were used for western blotting (Raldh1 and Raldh2 protein measurement), and the remaining 6 were used for Raldh1 and Raldh2 mRNA measurement.

Treatment

In the EA group, EA was applied to acupoints for hemiplegia. The acupoint prescription included ST36 (Zusanli, 5 mm below the head of fibula under the knee joint and 2 mm lateral to the anterior tibial tubercle) and LI11 (Quchi, in the depression lateral to the anterior aspect of the radius joint of the forelimb) at a depth of 5 mm into the skin with a stainless needle measuring 0.25×20 mm in length with a guide-tube for 20 min (Wujiang Shenli Medical & Health Material Co., Ltd., Wujiang, China). Electric stimulation was generated using an electrical stimulator (Huatuo SDZ-II; Suzhou Medical Appliance Factory Co., Ltd., China) for 20 min and the stimulation parameter exhibited disperse-dense waves of a frequency of 5/20 Hz (28.5 msec/15 msec pulse duration) and a current density of 2–4 mA. Model and sham groups did not undergo any EA, which was administered once a day for a period of 4 weeks.

Behavioral testing

Neurological function deficits were evaluated using the HomeCageScan system at 1, 7, 14 and 28 days after tMCAO. Each rat was placed in a separate cage identical to its home-cage, with fresh bedding, food and water. A video camera (Sony DCR-HC62) was positioned perpendicular to the long axis of the cage so that the field of view included the entire length of the cage. The rats were acclimatized to their new environment for 1 h and activity was recorded for 24 h (12-hour light/dark cycle) under standard fluorescent lighting. The rats were then returned to their home-cages. The full length of each video was analyzed using HomeCageScan (version 3.0; Clever Systems, Inc.) for the quantification of rat motor deficits, specifically quantifying behaviors including grooming, feeding, walking and rearing. The sum of the time spent in each behavior was calculated, yielding an index of total activity.

Measurement of infarct volume

Infarct volume was assessed at 1, 7, 14 and 28 days after tMCAO using TTC staining. Animals were sacrificed by 10% chloral hydrate overdose, brains were rapidly removed, sectioned coronally at 2-mm intervals from the frontal pole and immersed in TTC (2%) at 37°C for 20 min, followed by formaldehyde (4%) for 15 min. The hemispheric infarcted area in each section was calculated by subtracting the area of normal TTC-stained brain in the ipsilateral cortex from the contralateral area. The data were analyzed with a computer-based image analysis system (Adobe Photoshop 8.0). The sum of the areas of all the sections was calculated and then multiplied by 2 mm, yielding the volume.

RNA extraction and RT-PCR analysis

A total of 72 rats (n=6/group) were sacrificed by decapitation at 1, 7, 14 and 28 days after tMCAO. The ischemic zone of rat brain was rapidly removed and stored at −80°C for subsequent analysis. Total RNA from brain tissue was isolated with TRIzol reagent (Invitrogen). Oligo (dT)-primed RNA (1 μg) was reverse-transcribed with SuperScript II reverse transcriptase (Promega) according to the manufacturer’s instructions. The obtained cDNA was used to determine the mRNA amount of Raldh1 or Raldh2 by PCR with Taq DNA polymerase (Fermentas). β-actin was used as an internal model. The primers used in these reactions are listed in Table I. The amplified products were analyzed by 1.5% agarose gel electrophoresis. Optical density ratios for Raldh1, Raldh2 and β-actin were used for the semi-quantitative analyses.

Table I

PT-PCR primers.

Table I

PT-PCR primers.

GenePrimer sequenceAnnealing temperature (°C)
Raldh1Forward 5′-CTTCTTTGTCCAGCCCACAGTCT-3′
Reverse 5′-GTTCACCCAGTTCTCGTCCATTT-3′55
Raldh2Forward 5′-GTCGTCAACATTCTGCCAGGGTAT-3′
Reverse 5′-CTGCTCCACAGCGTAGTCCAAGTC-3′56
β-actinForward 5′-ACTGGCATTGTGATGGACTC-3′
Reverse 5′-CAGCACTGTGTTGGCATAGA-3′55
Western blot analysis

The ischemic zone of rat brain was sectioned, and the corresponding samples were homogenized with a homogenizer in lysis buffer (NP-40 lysis buffer and 100 mM PMSF). The homogenates were centrifuged at 14,000 rpm for 30 min at 4°C. The supernatant was collected and the total protein content was determined using a Micro BCA protein assay kit with bovine serum albumin as the standard (Pierce Chemical, Rockford, IL, USA). Equal amounts of protein (20 μg) were boiled in loading buffer and separated by 12% SDS-PAGE gel under reducing condition using 80 V for 1 h. The proteins were then electrophoretically transferred onto nitrocellulose membranes using the iBlot Western Detection Stack/iBlot Dry Blotting system (Invitrogen). Membranes were blocked for 30 min with agitation at RT in SuperBlock T20 (TBS) blocking buffer (Thermo Scientific, Rockford, IL, USA). Membranes were washed in TBS with 0.25% Tween-20 (TBST) and exposed to primary antibodies against Raldh1 or Raldh2 (1:1,000) overnight at 4°C on a rocking platform or rotator. β-actin (1:1,000) was also measured as an internal control for protein loading. After membranes were washed in TBST, secondary horseradish peroxidase (HRP)-conjugated antibodies (anti-goat) were added at 1:5,000 dilution for 1 h at room temperature and the membranes were washed again in TBST. The antibody-bound protein bands were then detected with ECL, and images were captured using a Bio-Rad ChemiDoc XRS+ (Bio-Rad, Hercules, CA, USA). The ratio of gray scale values of the target protein to the internal control was used to measure the relative amount of Raldh1 and Raldh2.

Statistical analysis

Data are presented as the means ± SD for the indicated number of independently performed experiments. Comparisons between multiple groups were conducted with one-way ANOVA, whereas within each group data were analyzed with analysis of intraclass variance analysis. Differences between the 2 groups were assessed using the Student’s t-test. P-values of <0.05 and <0.01 were considered as significant and highly significant differences, respectively.

Results

RA reduces infarct volume in rats following tMCAO

TTC staining was performed to determine the infarct volumes in brain sections at 1, 7, 14 and 28 days after tMCAO. The infarct volume was found to be decreased in a time-dependent manner in both the EA and model groups (P<0.05) (Fig. 1). At 7 and 14 days, the infarct volume was significantly decreased in the EA group (P<0.05) vs. the model group. No significant differences were detected in the infarct volume between the EA and model groups at 1 and 28 days after tMCAO (P>0.05), indicating that rats were able to recover naturally after tMCAO.

RA improves neurological function in rats following tMCAO

Cerebral cortex infarction caused neurological function deficits in rats, mainly manifested as right forelimb paralysis. To quantify this deficit, 24-h video recordings were analyzed using a software program designed for rat home-cage behavioral assessment. Neurological function was found to have improved in a time-dependent manner in the EA and model groups (P<0.05) (Fig. 2). Time spent walking, rearing and grooming was increased, while time spent feeding was reduced in the EA group 7 and 14 days after tMCAO (P<0.05) compared with the model group. No significant differences in neurological function were observed between the EA and model groups at 1 and 28 days after tMCAO (P>0.05). No neurological function deficits were evident in the sham group.

EA treatment regulates the expression of Raldh1 and Raldh2 after tMCAO

To explore the mechanism of the potential neuroprotective effect of EA, RT-PCR and western blot analysis were carried out to examine the mRNA and protein expression in the ischemic brain after tMCAO. In the model group, Raldh1 and Raldh2 mRNA was greatly increased at 7 days, peaked at 14 days and then gradually decreased (Fig. 3). Additionally, in the model group, higher Raldh1 and Raldh2 mRNA levels were found at 7 and 14 days (P<0.05) compared with the sham group. In the EA group, the Raldh1 and Raldh2 mRNA levels were higher than those in the model group at 7 and 14 days (P<0.05). No significant differences were detected in the Raldh1 and Raldh2 mRNA expression levels between the EA and model groups at 1 and 28 days after tMCAO (P>0.05), and the pattern of protein expression of Raldh1 and Raldh2 was similar to their respective mRNA levels (Fig. 4). These findings suggest that EA promotes neurological functional recovery in rats following tMCAO through the regulation of expression of the RA family.

Discussion

Neural stem cells from the adult forebrain SVZ give rise to olfactory bulb interneurons, while those in the hippocampal dentate gyrus generate new neurons in the granule cell layer (18). Brain insults such as seizure, stroke and trauma activate neural stem cells to proliferate more rapidly, migrate into injured regions and form new neurons and glia (3036).

Retinoids, in particularly the active metabolite of vitamin A, RA, is known to stimulate neonatal SVZ and adult hippocampal neurogenesis (1416). Retinol and its derivatives exert their biological actions via specific nuclear receptors (RARs and RXRs) that regulate gene transcription (37). RA receptors also interact with other nuclear receptors that possess neuroprotective properties, such as 1,25-dihydroxyvitamin D (9) that is neuroprotective against stroke (38) and RXR receptors that form heterodimers with the vitamin D3 receptor (RXR-VDR). RXR is also able to form a dimer with thyroid hormone receptors, and thyroid hormone derivatives are protective against infarction (39). Thus, the neuroprotective effects of retinol and RA may be mediated, at least in part, by their formation of heterodimers with other nuclear receptors. In addition to its effect on nuclear receptors, retinol exerts acute effects on other potentially protective biological sites including calcium channels and gap junction channels, as well as having antioxidant properties. Calcium channel antagonists and antioxidants protect the brain from ischemia in animal models of stroke (40). RA also induced the expression of midkine, which is protective against brain ischemia (41).

RA is synthesized from retinol in a two-step pathway involving oxidation first to retinaldehyde, then to RA. The former step is predominantly catalyzed by retinol dehydrogenases, the latter step by retinaldehyde dehydrogenases (Raldhs), which are encoded by Raldh1, Raldh2 and Raldh3 expressed in non-overlapping patterns. Raldh3 is expressed only in the eye and facial region/ventral retina and the ganglionic eminence of the telencephalon. Later in development, this enzyme can be detected by zymography in a variety of sites, including the liver and adult skin. Raldh1 is present in only the dorsal retina, ventral mesencephalon and projections from here to the corpus striatum and the otic vesicle regions (data not shown). Although Raldh2 is absent from most of the brain proper, it is strongly expressed in the leptomeninges that surround it (42). In the present study, we found that the Raldh1 and Raldh2 mRNA levels of the EA and model groups were greatly increased at 7 days, peaked at 14 days and gradually decreased at 28 days after tMCAO. A marked increase was observed in the Raldh1 and Raldh2 mRNA levels at 7 and 14 days in the EA group (P<0.05) compared with the model group. Expression of the Raldh1 and Raldh2 protein was similar to that of Raldh1 and Raldh2 mRNA.

Behavior is known to be the ultimate and most complex output of the brain. To quantify neurological function deficit, 24-h video recordings were conducted and digitally analyzed. The assessment of locomotor deficits using this method has been established in models of Huntington’s and prion disease (43), with a similar approach having been described recently in a rat ICH model (44). Compared with traditional methods of behavioral assessment in stroke models, this digital approach has a number of benefits (4547). First, it requires minimal investigator participation, reducing labor costs and the potential for bias. Second, it facilitates the analysis of night-time behavior. The validity of daytime testing of nocturnal animals in stroke models has never been established and is questionable given the behavioral inhibition and cognitive disturbances observed in rats during light phase testing. Third, this digital approach measures behaviors that are directly relevant to the animal’s functional status. Most importantly, however, it eliminates the variability inherent in measuring brief episodes of behavior in animals stressed by environmental variation and human interference.

In this study, we found that time spent walking, rearing and grooming was markedly increased, whereas time spent feeding was reduced with locomotor activity improvement in the EA group at 7 and 14 days after tMCAO, compared with that in the model group (P<0.05). The changes we found in the neurological function were concomitant with improvements in RA mRNA and protein expression. These results therefore suggest that the administration of EA is likely to promote neurological functional recovery and regulate the expression of RA mRNA and protein. To the best of our knowledge, this is the first study to report that EA regulates RA expression.

It was also demonstrated that there was no significant difference in the infarct volume, neurological function, expression of RA mRNA and protein between the EA and model groups at 28 days after tMCAO (P>0.05), indicating that rats were able to recover naturally after tMCAO. Spontaneous recovery may be due to neurite regeneration and synapse remodeling, as well as a reduction in brain edema. A series of plasticity responses after tMCAO, including the release of neurotrophic factors, synthesis of neuron-specific proteins and rise of synaptic excitation, is involved in the process of functional recovery (48). Additionally, our study examined the correlation between neurogenesis and the use of young rats in behavioral recovery. The findings of this study are in contrast with the reality that the elderly population is most often affected by strokes and these people, unlike young healthy rodents, also have a multiplicity of other disease co-morbidities, such as hypertension and diabetes that along with increased age, are likely to limit or alter the neurogenic response post-stroke. Indeed, aged animals exhibit stroke-induced neurogenesis, however, their response is ‘muted’ compared with young animals (49,50). Although the spontaneous recovery of neurological function deficit may occur after tMCAO, the administration of EA significantly promoted neural function recovery and decreased infarct volume. Raldh1 and Raldh2 mRNA and protein expression was increased by EA treatment following tMCAO compared to the model group.

In conclusion, administration of EA decreased the infarct volume, promoted neurological functional recovery and demonstrated that this neuroprotective effect is attributed, at least in part, to the expression of RA mRNA and protein. These findings likely provide an experimental basis for the treatment of ischemic stroke by electroacupuncture.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (no. 30901935) and the Key Laboratory for Orthopedics and Sports Rehabilitation of the Ministry of Education, China.

References

1 

Lo EH, Dalkara T and Moskowitz MA: Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci. 4:399–415. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Madden K: Optimal timing of thrombolytic therapy in acute ischaemic stroke. CNS Drugs. 16:213–218. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Willaime-Morawek S and van der Kooy D: Cortex- and striatum-derived neural stem cells produce distinct progeny in the olfactory bulb and striatum. Eur J Neurosci. 27:2354–2362. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Nakagomi T, Taguchi A, Fujimori Y, Saino O, Nakano-Doi A, Kubo S, Gotoh A, Soma T, Yoshikawa H, Nishizaki T, Nakagomi N, Stern DM and Matsuyama T: 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

5 

Jin K, Wang X, Xie L, Mao XO, Zhu W, Wang Y, Shen J, Mao Y, Banwait S and Greenberg DA: Evidence for stroke-induced neurogenesis in the human brain. Proc Natl Acad Sci USA. 103:13198–13202. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Macas J, Nern C, Plate KH and Momma S: Increased generation of neuronal progenitors after ischemic injury in the aged adult human forebrain. J Neurosci. 26:13114–13119. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Minger SL, Ekonomou A, Carta EM, Chinoy A, Perry RH and Ballard CG: Endogenous neurogenesis in the human brain following cerebral infarction. Regen Med. 2:69–74. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Landgren H and Curtis MA: Locating and labeling neural stem cells in the brain. J Cell Physiol. 226:1–7. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Hodge RD and Hevner RF: Expression and actions of transcription factors in adult hippocampal neurogenesis. Dev Neurobiol. 71:680–689. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Ross SA, McCaffery PJ, Drager UC and De Luca LM: Retinoids in embryonal development. Physiol Rev. 80:1021–1054. 2000.PubMed/NCBI

11 

McCaffery P, Zhang J and Crandall JE: Retinoic acid signaling and function in the adult hippocampus. J Neurobiol. 66:780–791. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Werner EA and Deluca HF: Retinoic acid is detected at relatively high levels in the CNS of adult rats. Am J Physiol Endocrinol Metab. 282:E672–E678. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Calza L, Giuliani A, Fernandez M, Pirondi S, D’Intino G, Aloe L and Giardino L: Neural stem cells and cholinergic neurons: regulation by immunolesion and treatment with mitogens, retinoic acid, and nerve growth factor. Proc Natl Acad Sci USA. 100:7325–7330. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Haskell GT and LaMantia AS: Retinoic acid signaling identifies a distinct precursor population in the developing and adult forebrain. J Neurosci. 25:7636–7647. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Wang TW, Zhang H and Parent JM: Retinoic acid regulates postnatal neurogenesis in the murine subventricular zone-olfactory bulb pathway. Development. 132:2721–2732. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Jacobs S, Lie DC, DeCicco KL, Shi Y, DeLuca LM, Gage FH and Evans RM: Retinoic acid is required early during adult neurogenesis in the dentate gyrus. Proc Natl Acad Sci USA. 103:3902–3907. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Mey J and McCaffery P: Retinoic acid signaling in the nervous system of adult vertebrates. Neuroscientist. 10:409–421. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Plane JM, Whitney JT, Schallert T and Parent JM: Retinoic acid and environmental enrichment alter subventricular zone and striatal neurogenesis after stroke. Exp Neurol. 214:125–134. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Ikeda R, Kurokawa MS, Chiba S, Yoshikawa H, Ide M, Tadokoro M, Nito S, Nakatsuji N, Kondoh Y, Nagata K, Hashimoto T and Suzuki N: Transplantation of neural cells derived from retinoic acid-treated cynomolgus monkey embryonic stem cells successfully improved motor function of hemiplegic mice with experimental brain injury. Neurobiol Dis. 20:38–48. 2005. View Article : Google Scholar

20 

Kim EH, Jang MH, Shin MC, Lim BV, Kim HB, Kim YJ, Chung JH and Kim CJ: Acupuncture increases cell proliferation and neuropeptide Y expression in dentate gyrus of streptozotocin-induced diabetic rats. Neurosci Lett. 327:33–36. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Kim EH, Kim YJ, Lee HJ, Huh Y, Chung JH, Seo JC, Kang JE, Lee HJ, Yim SV and Kim CJ: Acupuncture increases cell proliferation in dentate gyrus after transient global ischemia in gerbils. Neurosci Lett. 297:21–24. 2001. View Article : Google Scholar

22 

Liu Q, Yu J, Mi WL, Mao-Ying QL, Yang R, Wang YQ and Wu GC: Electroacupuncture attenuates the decrease of hippocampal progenitor cell proliferation in the adult rats exposed to chronic unpredictable stress. Life Sci. 81:1489–1495. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Cheng H, Yu J, Jiang Z, Zhang X, Liu C, Peng Y, Chen F, Qu Y, Jia Y, Tian Q, Xiao C, Chu Q, Nie K, Kan B, Hu X and Han J: Acupuncture improves cognitive deficits and regulates the brain cell proliferation of SAMP8 mice. Neurosci Lett. 432:111–116. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Naylor AS, Bull C, Nilsson MK, Zhu C, Bjork-Eriksson T, Eriksson PS, Blomgren K and Kuhn HG: Voluntary running rescues adult hippocampal neurogenesis after irradiation of the young mouse brain. Proc Natl Acad Sci USA. 105:14632–14637. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Yun SJ, Park HJ, Yeom MJ, Hahm DH, Lee HJ and Lee EH: Effect of electroacupuncture on the stress-induced changes in brain-derived neurotrophic factor expression in rat hippocampus. Neurosci Lett. 318:85–88. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Gao J, Wang S, Wang X and Zhu C: Electroacupuncture enhances cell proliferation and neuronal differentiation in young rat brains. Neurol Sci. 32:369–374. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Jang MH, Shin MC, Lee TH, Lim BV, Shin MS, Min BI, Kim H, Cho S, Kim EH and Kim CJ: Acupuncture suppresses ischemia-induced increase in c-Fos expression and apoptosis in the hippocampal CA1 region in gerbils. Neurosci Lett. 347:5–8. 2003. View Article : Google Scholar : PubMed/NCBI

28 

The Ministry of Science and Technology of the People’s Republic of China. Guidance Suggestions for the Care and Use of Laboratory Animals. 2006 09 30

29 

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

30 

Arvidsson A, Collin T, Kirik D, Kokaia Z and Lindvall O: Neuronal replacement from endogenous precursors in the adult brain after stroke. Nat Med. 8:963–970. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Goings GE, Sahni V and Szele FG: Migration patterns of subventricular zone cells in adult mice change after cerebral cortex injury. Brain Res. 996:213–226. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Lichtenwalner RJ and Parent JM: Adult neurogenesis and the ischemic forebrain. J Cereb Blood Flow Metab. 26:1–20. 2006. View Article : Google Scholar

33 

Parent JM, Valentin VV and Lowenstein DH: Prolonged seizures increase proliferating neuroblasts in the adult rat subventricular zone-olfactory bulb pathway. J Neurosci. 22:3174–3188. 2002.PubMed/NCBI

34 

Parent JM, Vexler ZS, Gong C, Derugin N and Ferriero DM: Rat forebrain neurogenesis and striatal neuron replacement after focal stroke. Ann Neurol. 52:802–813. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Parent JM, Yu TW, Leibowitz RT, Geschwind DH, Sloviter RS and Lowenstein DH: Dentate granule cell neurogenesis is increased by seizures and contributes to aberrant network reorganization in the adult rat hippocampus. J Neurosci. 17:3727–3738. 1997.PubMed/NCBI

36 

Zhang RL, Zhang ZG, Zhang L and Chopp M: Proliferation and differentiation of progenitor cells in the cortex and the subventricular zone in the adult rat after focal cerebral ischemia. Neuroscience. 105:33–41. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Liden M and Eriksson U: Understanding retinol metabolism: structure and function of retinol dehydrogenases. J Biol Chem. 281:13001–13004. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Wang Y, Chiang YH, Su TP, Hayashi T, Morales M, Hoffer BJ and Lin SZ: Vitamin D(3) attenuates cortical infarction induced by middle cerebral arterial ligation in rats. Neuropharmacology. 39:873–880. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Doyle KP, Suchland KL, Ciesielski TM, Lessov NS, Grandy DK, Scanlan TS and Stenzel-Poore MP: Novel thyroxine derivatives, thyronamine and 3-iodothyronamine, induce transient hypothermia and marked neuroprotection against stroke injury. Stroke. 38:2569–2576. 2007. View Article : Google Scholar

40 

Sato Y, Meller R, Yang T, Taki W and Simon RP: Stereo-selective neuroprotection against stroke with vitamin A derivatives. Brain Res. 1241:188–192. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Harvey BK, Shen H, Chen GJ, Yoshida Y and Wang Y: Midkine and retinoic acid reduce cerebral infarction induced by middle cerebral artery ligation in rats. Neurosci Lett. 369:138–141. 2004. View Article : Google Scholar : PubMed/NCBI

42 

McCaffery P and Drager UC: Regulation of retinoic acid signaling in the embryonic nervous system: a master differentiation factor. Cytokine Growth Factor Rev. 11:233–249. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Steele AD, Jackson WS, King OD and Lindquist S: The power of automated high-resolution behavior analysis revealed by its application to mouse models of Huntington’s and prion diseases. Proc Natl Acad Sci USA. 104:1983–1988. 2007.PubMed/NCBI

44 

Otero L, Zurita M, Aguayo C, Bonilla C, Rodríguez A and Vaquero J: Video-Tracking-Box linked to Smart software as a tool for evaluation of locomotor activity and orientation in brain injured rats. J Neurosci Methods. 188:53–57. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Roedel A, Storch C, Holsboer F and Ohl F: Effects of light or dark phase testing on behavioural and cognitive performance in DBA mice. Lab Anim. 40:371–381. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Spruijt BM and DeVisser L: Advanced behavioural screening: automated home cage ethology. Drug Discov Today Technol. 3:231–237. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Chen L, Zhang X, Chen-Roetling J and Regan RF: Increased striatal injury and behavioral deficits after intracerebral hemorrhage in hemopexin knockout mice. J Neurosurg. 114:1159–1167. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Stichel CC and Muller HW: Experimental strategies to promote axonal regeneration after traumatic central nervous system injury. Prog Neurobiol. 56:119–148. 1998. View Article : Google Scholar : PubMed/NCBI

49 

Gao P, Shen F, Gabriel RA, Law D, Yang E, Yang GY, Young WL and SU H: Attenuation of brain response to vascular endothelial growth factor-mediated angiogenesis and neurogenesis in aged mice. Stroke. 40:3596–3600. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Leasure JL and Grider M: The effect of mild post-stroke exercise on reactive neurogenesis and recovery of somatosensation in aged rats. Exp Neurol. 226:58–67. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January 2013
Volume 31 Issue 1

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
Hong J, Wu G, Zou Y, Tao J and Chen L: Electroacupuncture promotes neurological functional recovery via the retinoic acid signaling pathway in rats following cerebral ischemia-reperfusion injury. Int J Mol Med 31: 225-231, 2013
APA
Hong, J., Wu, G., Zou, Y., Tao, J., & Chen, L. (2013). Electroacupuncture promotes neurological functional recovery via the retinoic acid signaling pathway in rats following cerebral ischemia-reperfusion injury. International Journal of Molecular Medicine, 31, 225-231. https://doi.org/10.3892/ijmm.2012.1166
MLA
Hong, J., Wu, G., Zou, Y., Tao, J., Chen, L."Electroacupuncture promotes neurological functional recovery via the retinoic acid signaling pathway in rats following cerebral ischemia-reperfusion injury". International Journal of Molecular Medicine 31.1 (2013): 225-231.
Chicago
Hong, J., Wu, G., Zou, Y., Tao, J., Chen, L."Electroacupuncture promotes neurological functional recovery via the retinoic acid signaling pathway in rats following cerebral ischemia-reperfusion injury". International Journal of Molecular Medicine 31, no. 1 (2013): 225-231. https://doi.org/10.3892/ijmm.2012.1166