Open Access

Transplantation of mesenchymal stem cells promotes the functional recovery of the central nervous system following cerebral ischemia by inhibiting myelin‑associated inhibitor expression and neural apoptosis

  • Authors:
    • Nianping Feng
    • Guang Hao
    • Fenggang Yang
    • Fujun Qu
    • Haihong Zheng
    • Songlan Liang
    • Yonghua Jin
  • View Affiliations

  • Published online on: February 19, 2016     https://doi.org/10.3892/etm.2016.3089
  • Pages: 1595-1600
  • Copyright: © Feng 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

Cerebral ischemia, which may lead to cerebral hypoxia and damage of the brain tissue, is a leading cause of human mortality and adult disability. Mesenchymal stem cells (MSCs) are a class of adult progenitor cells with the ability to differentiate into multiple cell types. The transplantation of bone marrow‑derived MSCs is a potential therapeutic strategy for cerebral ischemia. However, the underlying mechanism has yet to be elucidated. In the present study, primary MSCs were isolated from healthy rats, labeled and transplanted into the brains of middle cerebral artery occlusion rat models. The location of the labeled MSCs in the rat brains were determined by fluorescent microscopy, and the neurological functions of the rats were scored. Immunohistochemical analyses demonstrated that the protein expression levels of myelin‑associated inhibitors of regeneration, including Nogo‑A, oligodendrocyte myelin glycoprotein and myelin‑associated glycoprotein, were decreased following transplantation of the bone marrow‑derived MSCs. Furthermore, the mRNA expression levels of Capase‑3 and B‑cell lymphoma 2, as determined by reverse transcription‑quantitative polymerase chain reactions, were downregulated and upregulated, respectively, in the MSC‑transplanted rats; thus suggesting that neural apoptosis was inhibited. The results of the present study suggested that the transplantation of bone marrow‑derived MSCs was able to promote the functional recovery of the central nervous system following cerebral ischemia. Accordingly, inhibitors targeting myelin‑associated inhibitors and apoptosis may be of clinical significance for cerebral ischemia in the future.

Introduction

Cerebral ischemia occurs as a result of insufficient blood flow to the brain (1,2), which leads to limited oxygen supply, or cerebral hypoxia, and may eventually result in the death of brain tissue, cerebral infarction or ischemic stroke (3). It is a leading cause of death and the predominant cause of adult disability (4). At present, there is no specific therapy for improving functional recovery.

Mesenchymal stem cells (MSCs) are a class of adult progenitor cells capable of differentiating into several cell types (58). Several previous studies have indicated that bone marrow derived MSCs may serve as a source of cells for cell transplantation therapy following cerebral ischemia and have been successfully used for the treatment of experimental stroke (911). In the aforementioned studies, bone marrow derived MSCs selectively targeted injured brain tissue and promoted functional recovery via various cell delivery routes. Although the transplantation of bone marrow derived MSCs has been revealed to provide therapeutic benefit to cerebral ischemia, the underlying mechanisms have yet to be elucidated.

The adult mammalian central nervous system (CNS) possesses a poor capability to regenerate axons following injury. The failure of CNS axons to regenerate after damage has significant consequences for brain ischemia (12). Several factors contributing to this regenerative failure include the intrinsic state of the injured neuron, the formation of the inhibitory glial scar and the presence of inhibitory myelin debris. A previous study demonstrated that myelin-associated neurite growth inhibitors had a crucial role in preventing CNS regeneration after brain injury (13). Three inhibitors of regeneration have been identified in myelin thus far, including: Nogo-A, oligodendrocyte myelin glycoprotein (OMgp) and myelin-associated glycoprotein (MAG). These molecules are membrane proteins of myelin and expressed predominately by oligodendrocytes in the adult mammalian CNS (12,1417).

It has been previously reported that numerous brain neurons in ischemia penumbia undergo apoptosis following either global or focal ischemic insults (18). Apoptosis is a unique, gene-directed form of cell death, and is characterized by nuclear condensation and cytoplasmic fragmentation that contributes to physiological and pathological processes (19). The Caspases, in particular Caspase-3, perform a key role in the execution of apoptosis (20). B-cell lymphoma 2 (Bcl-2) is the crucial member of the Bcl-2 family of regulator proteins that regulate apoptosis, and is able to inhibit apoptosis without affecting cellular proliferation (21,22).

In the present study, the intraluminal model of transient middle cerebral artery occlusion (MCAO) was established in rats to investigate the effects of the transplantation of bone marrow derived MSCs on CNS functional recovery. It was determined in the present study that the transplantation of bone marrow derived MSCs promoted the functional recovery of the CNS following brain ischemia via the inhibition of the expression of Nogo-A, OMgp and MAG, as well as neuronal apoptosis.

Materials and methods

Rats

A total of 90 healthy adult male Sprague-Dawley (SD) rats weighing 220–280 g, and 15 healthy adult male SD rats weighing 80–120 g, were obtained from the Experimental Animal Center of the Second Affiliated Hospital of Harbin Medical University (Harbin, China). The rats were maintained in a room at 23±1°C under a 12-h light/dark cycle with ad libitum access to food and water. All experiments were performed with approval from the Ethics Committee of Harbin Medical University.

Rat MCAO model

Adult male SD rats (n=90; weight, 220–280 g) were used to establish the intraluminal model of transient MCAO. A transient (2-h) MCAO was induced using a modified version of the method described by Longa et al (23). Briefly, rats were anesthetized by intraperitoneal injection with 10% chloral hydrate (0.35 ml/100 g; Sigma-Aldrich, St. Louis, MO, USA) and placed in the supine position. The left common carotid artery (CCA), external carotid artery and internal carotid artery (ICA) were exposed and isolated through a carotid midline incision (2.0–2.5 cm). A surgical nylon suture 0.234 mm in diameter and wax-coated tip was advanced from the CCA into the lumen of the ICA until it blocked the origin of the middle cerebral artery. From the bifurcation of the CCA, 18–20 mm of the nylon suture was in the lumen and 1 cm was left outside. After 2 h occlusion, the suture was withdrawn to permit reperfusion until resistance was felt. Neurological examinations, as described by Longa et al (23), were performed subsequent to the onset of occlusion. The neurological results were scored using a 5-point scale as follows: A score of 0 indicated no neurological deficit; 1, (failure to extend left forepaw fully) indicated a mild focal neurological deficit; 2, (circling to the left) indicated a moderate focal neurological deficit and 3, (falling to the left) indicated a severe focal deficit. Rats with a score of 4 did not walk unaided and had a decreased level of consciousness. A total of 90 rats with a score of 1–3 were considered as suitable models of MCAO and were used in further experiments.

Bone marrow-derived MSCs culture

MSCs were harvested from 15 SD rats (weight, 80–120 g). Primary MSCs were isolated from the bone marrow of the tibias and femurs of the aforementioned rats by their adherence to plastic in vitro culture. At 3 days following MCAO, the rats were sacrificed by cervical dislocation following anesthetization by intraperitoneal injection with 10% chloral hydrate (0.35 ml/100 g). Under sterile conditions, tibias and femurs were harvested, the adherent soft tissue was removed and the ends of the bones were excised toward the start of the marrow cavity. Fresh bone marrow was harvested aseptically by flushing the cavity of the bone with needles filled with Dulbecco's modified Eagle's medium-low glucose (DMEM-LG; HyClone; GE Healthcare Life Sciences, Logan, UT, USA) supplemented with 10% fetal bovine serum (FBS; Hyclone). A single cell suspension was prepared by gentle pipetting several times and passing the cell suspension through a 200 mesh metal strainer (Thermo Fisher Scientific, Inc., Waltham, MA, USA). The cells were seeded into each tissue culture flask at a density of 106 cells/ml and cultured in an incubator (Forma Scientific; Thermo Fisher Scientific, Inc.) containing 5% CO2 at 37°C. After 48 h, non-adherent cells were removed and the medium renewed and changed every 2 days thereafter. Once cells reached ~90% confluency, as determined by phase contrast microscopy (Eclipse 80i; Nikon Corporation, Tokyo, Japan), they were passaged into culture flasks at a 1:2 ratio.

Cell labeling

In order to monitor the distribution of bone marrow-derived MSCs in rat brains, 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester (CFSE; Fluka, Buchs, Switzerland) was used to label the MSCs. Briefly, after trypsinization (Invitrogen; Thermo Fisher Scientific, Inc.), cells at passage 3 were washed twice and resuspended in 200 µl DMEM. The cell suspension was then mixed with 200 µl labeling solution in a tube for 10 min at 37°C, with periodical tapping of the tube. The reaction was terminated by the addition of FBS, and the CFSE labeled cells were washed twice and resuspended in 0.01 ml PBS. Cell viability was evaluated by Trypan blue staining (Sigma-Aldrich). Only single cell suspensions with 95% viability were used for implantation. Approximately 3×106 cells/ml of MSCs in 1 ml PBS were transplanted into each rat.

Experimental groups and transplantation

All rats (n=90) were randomly divided into 2 groups at 24 h following the induction of MCAO or sham operation: MCAO group (n=40), and MSCs group (n=50). Rats in the MCAO group received 1 ml 0.01 M PBS via an intravenous injection. Rats in the MSCs group received CFSE-labeled 3×106 MSCs in 1 ml 0.01 M PBS via an intravenous injection. All transplantation procedures were performed under aseptic conditions and rats that received transplantation were selected at random.

Distribution of CFSE-labeled bone marrow derived MSCs

Three days subsequent to the establishment of MCAO, 10 rats in the MSCs group were sacrificed, and their brains were removed and stored in liquid nitrogen. Coronal blocks (2 mm) containing brain tissues were cut anterior and posterior to the optic chiasm, and cryostat sections were prepared. Subsequently, the survival and distribution of CFSE-labelled MSCs were observed using an inverted fluorescence microscope (Eclipse 80i).

Behavioral testing

Modified neurological severity scores (mNSS) described by Chen et al (9) were employed to evaluate neurological deficits in rats. In all rats, a series of NSS tests was performed on days 3, 7 and 14 following the establishment of MCAO by 2 investigators who were blind to the experimental groups. Neurological function was scored on a scale of 0 to 11 (normal score, 0; maximal deficit score, 11). The severity of injury was scored by awarding 1 point each time the rat was unable to perform the assessment or for the lack of a tested reflex; thus, the higher the score, the more severe the injury. This modified version of mNSS is composite of the motor, sensory and reflex assessments.

Immunohistochemical assessment

Rats were anesthetized with 10% chloral hydrate (0.35 ml/100 g, intraperitoneal injection). The rat brains were removed and were postfixed in 4% paraformaldehyde (Sigma-Aldrich) for 24 h, and embedded in paraffin. Sections were deparaffinized and incubated with 3% H2O2 for 10 min to block endogenous peroxidase activity. Brain sections were placed in citrate buffer (pH 6) at 80 kPa for 2 min. Tissues were treated with primary antibodies and incubated at 4°C for 12 h. Tissues were incubated at room temperature for 20 min with horseradish peroxidase-labeled goat anti-rat IgG antibody (1:200; cat. no. ZB2307; ZSGB-BIO, Beijing, China). Stable 3,3′-diaminobenzidine (ZSGB-BIO) was then used as a chromogen for light microscopy (Eclipse 80i). Counterstaining of sections was performed with hematoxylin and eosin (Shanghai BetterBioChem Co., Ltd., Shanghai, China). The primary antibodies used in the study were as follows: Rabbit anti-Caspase-3 polyclonal antibody (1:100; PA5-16335; LabVision/Neomarkers; Thermo Fisher Scientific, Inc.), rabbit anti-Bcl-2 antibody (1:80; sc-492; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), rabbit anti-Nogo-A antibody (1:30; PA1060; Wuhan Boster Biological Technology, Ltd., Wuhan, China), rabbit anti-OMgp antibody (1:80; BS-1200R; BIOSS, Beijing, China) and rabbit anti-MAG antibody (1:100; BS-0257R; BIOSS). Photomicrographs were acquired at ×200 magnification under an inverted microscope (Eclipse 80i). The distribution of Nogo-A, OMgp, MAG, Caspase-3, in addition to Bcl-2 expression levels, were assessed by counting the number of positively stained cells in 10 distinct regions of ischemic penumbra in each section.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA from the rat brain tissue was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Brain tissue was homogenized in ~1 ml TRIzol reagent. After homogenization, insoluble material from the homogenate was removed by centrifuging at 10,387 × g for 10 min at 4°C. Following isopropyl alcohol precipitation, total RNA was treated with DNase (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol, in order to remove contaminating DNA. cDNA was synthesized from 1 µg total RNA using the SuperScript III Reverse-Transcriptase kit (Invitrogen; Thermo Fisher Scientific, Inc.) in a final volume of 20 µl. The reactions were incubated in 96-well optical plates at 95°C for 10 min, followed by 40 cycles of 2 min at 95°C, 45 sec at 54°C, 45 sec at 68°C, and extension at 68°C for 8 min. qPCR was performed using the SYBR Prime Script kit (Invitrogen; Thermo Fisher Scientific, Inc.) and the Applied Biosystems 7500 Fluorescent Quantitative PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The primer sequences used were as follows: Caspase-3 forward, 5′-CATGGCCCTGAAATACGAAGTC-3′ and reverse, 5′-CCGTCCCTTGAATTTCTCCAG-3′; Bcl-2 forward, 5′-GGGAGATCGTGATGAAGTACATCC-3′ and reverse, 5′-AGTTCCACAAAGGCATCCCAG-3′; β-actin forward, 5′-AAGAGAGGCATCCTGACCCTG-3′ and reverse, 5′-TCCAGACGCAGGATGGCATG-3′ (GenePharma Co., Ltd., Shanghai, China). Relative quantification was performed using the 2−ΔΔCq method (24), with normalization to the β-actin gene. As a negative control, RT-qPCR was performed using all reagents except for the template, and for an RT-minus control, RT-qPCR was conducted in the absence of reverse transcriptase.

Statistical analysis

Statistical analyses were performed using the SPSS 13.0 software (SPSS, Inc., Chicago, IL, USA). Statistical significance of mNSS was analyzed using one-way analysis of variance. All data are presented as the mean ± standard deviation and analyzed for statistical significance by Pearson's χ2 test. P<0.05 was considered to indicate a statistically significant difference.

Results

Morphology and MSC migration

MSCs were collected from the bone marrow of femurs and tibias of SD rats and were evaluated morphologically using a phase contrast microscope. Subsequent to 14 days of culture, primary bone marrow derived MSCs reached 70% confluence and exhibited a fibroblast-like morphology (Fig. 1A). The third passage of MSCs also exhibited a fibroblastic morphology and reached 80–90% confluency after 7 days of culture (Fig. 1B). A model of MCAO was then established in rats to detect the effect of transplantation of MSCs by intravenous injection to cerebral ischemia. Green fluorescent CFSE-labeled MSCs were observed 3 days following the establishment of MCAO using a fluorescence microscope. CFSE-labeled MSCs migrated to the brains of rats and survived in the ischemic brain (Fig. 1C).

Evaluation of neurological deficit

The cerebral ischemia-induced neurological deficits were determined by mNSS assessments at 3, 7 and 14 days. As displayed in Fig. 2, no significant differences in mNSS scores were observed between the MSCs group and the PBS group on day 3. However, on days 7 and 14, the mNSS scores of the MSCs group were significantly lower compared with those of the PBS group (P<0.05), indicating that the transplantation of MSCs promoted the functional recovery of the CNS following cerebral ischemia.

Expression of Nogo-A, MAG and OMgp

As Nogo-A, MAG and OMgp are involved in the prevention of CNS regeneration following brain injury, we subsequently investigated whether transplantation of MSCs was able to affect their cellular expression. The MSCs group displayed a significantly reduced number of Nogo-A positive cells compared with the PBS group on days 3 and 14 (P<0.05); however, the difference was not significant on day 7 (Fig. 3 and Table I). With regard to OMgp, the PBS and the MCAO group displayed a similar number of OMgp-positive cells on day 3. However, the MSCs group displayed a significantly lower number of OMgp-positive cells compared with the MCAO group on days 7 and 14 (P<0.05; Table II). Furthermore, the MSCs group displayed a significantly lower number of MAG-positive cells compared with the MCAO group between days 3 and 14 (P<0.05; Table III). The aforementioned results suggest that the transplantation of MSCs reduced the cellular expression levels of Nogo-A, OMgp and MAG and thus contributed to the functional recovery of the CNS following cerebral ischemia in rats.

Table I.

Nogo-A-positively stained cells at different time points (n=10).

Table I.

Nogo-A-positively stained cells at different time points (n=10).

GroupDay 3Day 7Day 14
PBS31.02±3.8525.07±3.0052.28±4.55
MSCs   24.97±4.20a21.93±2.68 40.77±6.68a

a P<0.05 vs. PBS group. PBS, phosphate-buffered saline; MSCs, mesenchymal stem cells.

Table II.

Oligodendrocyte myelin glycoprotein-positively stained cells at different time points (n=10).

Table II.

Oligodendrocyte myelin glycoprotein-positively stained cells at different time points (n=10).

GroupDay 3Day 7Day 14
PBS32.60±5.1440.87±3.1246.02±3.35
MSCs30.42±3.58   31.22±3.22a   32.55±5.94a

a P<0.05 vs. PBS group. PBS, phosphate-buffered saline; MSCs, mesenchymal stem cells.

Table III.

Myelin-associated glycoprotein-positively stained cells at different time points (n=10).

Table III.

Myelin-associated glycoprotein-positively stained cells at different time points (n=10).

GroupDay 3Day 7Day 14
MCAO68.48±6.3566.70±5.0167.32±6.69
MSCs 51.68±7.21a 55.28±9.91a 53.52±7.70a

a P<0.05 vs. the MCAO group. MCAO, middle cerebral artery occlusion; MSCs, mesenchymal stem cells.

Expression of Caspase-3 and Bcl-2

Finally, it was determined whether the transplantation of MSCs was able to affect cell apoptosis, which is crucial factor in the functional recovery of CNS after cerebral ischemia. MSCs group had significantly lower Caspase-3 mRNA expression levels compared with the MCAO group on days 2 and 3 (P<0.01; Fig. 4A and B). Conversely, the MSCs group displayed significantly higher Bcl-2 expression levels compared with the MCAO group on days 2 and 3 (P<0.01; Fig. 4C and D). Thus, the present results indicate that the transplantation of MSCs is able to reduce apoptosis, which was likely to have accounted for the functional recovery of the CNS following cerebral ischemia.

Discussion

In the present study, the effect of transplantation of bone marrow derived MSCs on functional recovery of CNS after brain ischemia in a rat MCAO model was assessed, and the possible underlying mechanisms investigated.

Initially, it was confirmed that transplantation of MSCs provided therapeutic benefit to cerebral ischemia through the establishment of a rat model of MCAO. MSCs were isolated from rats and injected intravenously into rats with MCAO. The MSCs were able to survive and migrate to the damaged brain, and the transplantation of MSCs significantly improved the functional recovery of the CNS.

The possible mechanisms by which transplantation of MSCs promoted the functional recovery of the CNS following cerebral ischemia were then examined. The present study determined that transplantation of MSCs reduced the expression of axon regeneration inhibitors, including Nogo-A, MAG and OMgp. Furthermore, transplantation of MSCs inhibited neuronal apoptosis, decreasing Caspase-3 and increasing Bcl-2 protein expression levels. The two mechanisms function synergistically to promote the functional recovery of the CNS following cerebral ischemia.

The underlying mechanisms by which transplantation of MSCs reduces the expression of axon regeneration inhibitors and inhibits neuronal apoptosis have yet to be elucidated. It has previously been confirmed that MSCs constitutively secrete several growth factors, including nerve growth factors (NGF), brain derived neurotrophic factors (BDNF) and glioma derived neurotrophic factors (GDNF) (25). Exogenous NGF has an important role in neuronal plasticity and regenerative potential, in addition to the inhibition of neural apoptosis (26). BDNF is able to support the survival of injured CNS neurons in vitro and in vivo and inducing neurite outgrowth (27). GDNF is able to protect against neuronal death after brain injury (28,29). Therefore, it is plausible that MSCs secrete these growth factors, which in turn reduce the expression of axon regeneration inhibitors and inhibit neuronal apoptosis.

In conclusion, the present study revealed that transplantation of bone marrow derived MSCs promotes the functional recovery of the CNS subsequent to cerebral ischemia through decreasing the expression of myelin-associated inhibitors (MAIs) and inhibiting neuronal apoptosis. We propose that inhibitors targeting the aforementioned MAIs and the apoptotic pathway may have potential for the treatment of cerebral ischemia.

Acknowledgements

This work was supported by grants from the Science and Technology Project of the Education Department of Heilongjiang Province (grant nos. 12531311 and 12521336) and the Science Project of the Health Department of Heilongjiang Province (grant no. 2010-095).

References

1 

DeGracia DJ, Sullivan JM, Neumar RW, Alousi SS, Hikade KR, Pittman JE, White BC, Rafols JA and Krause GS: Effect of brain ischemia and reperfusion on the localization of phosphorylated eukaryotic initiation factor 2 alpha. J Cereb Blood Flow Metab. 17:1291–1302. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Kumar R, Azam S, Sullivan JM, Owen C, Cavener DR, Zhang P, Ron D, Harding HP, Chen JJ, Han A, et al: Brain ischemia and reperfusion activates the eukaryotic initiation factor 2alpha kinase, PERK. J Neurochem. 77:1418–1421. 2001. View Article : Google Scholar : PubMed/NCBI

3 

White BC, Sullivan JM, DeGracia DJ, O'Neil BJ, Neumar RW, Grossman LI, Rafols JA and Krause GS: Brain ischemia and reperfusion: Molecular mechanisms of neuronal injury. J Neurol Sci. 179:1–33. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Krumholz HM, Brindis RG, Brush JE, Cohen DJ, Epstein AJ, Furie K, Howard G, Peterson ED, Rathore SS, Smith SC Jr, et al: Standards for statistical models used for public reporting of health outcomes: An American Heart Association scientific statement from the Quality of Care and Outcomes Research Interdisciplinary Writing Group: Cosponsored by the Council on Epidemiology and Prevention and The Stroke Council. Endorsed by the American College of Cardiology Foundation. Circulation. 113:456–462. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Eglitis MA, Dawson D, Park KW and Mouradian MM: Targeting of marrow-derived astrocytes to the ischemic brain. Neuroreport. 10:1289–1292. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Prockop DJ: Marrow stromal cells as stem cells for continual renewal of nonhematopoietic tissues and as potential vectors for gene therapy. J Cell Biochem. (Suppl 30–31): 284–285. 1998. View Article : Google Scholar

7 

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S and Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 284:143–147. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Eglitis MA and Mezey E: Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice. Proc Natl Acad Sci USA. 94:4080–4085. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Chen J, Li Y, Wang L, Lu M, Zhang X and Chopp M: Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats. J Neurol Sci. 189:49–57. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Zhao LR, Duan WM, Reyes M, Keene CD, Verfaillie CM and Low WC: Human bone marrow stem cells exhibit neural phenotypes and ameliorate neurological deficits after grafting into the ischemic brain of rats. Exp Neurol. 174:11–20. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Honmou O, Onodera R, Sasaki M, Waxman SG and Kocsis JD: Mesenchymal stem cells: Therapeutic outlook for stroke. Trends Mol Med. 18:292–297. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Xie F and Zheng B: White matter inhibitors in CNS axon regeneration failure. Exp Neurol. 209:302–312. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Grados-Munro EM and Fournier AE: Myelin-associated inhibitors of axon regeneration. J Neurosci Res. 74:479–485. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Schwab ME: Nogo and axon regeneration. Curr Opin Neurobiol. 14:118–124. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Prinjha R, Moore SE, Vinson M, Blake S, Morrow R, Christie G, Michalovich D, Simmons DL and Walsh FS: Inhibitor of neurite outgrowth in humans. Nature. 403:383–384. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Domeniconi M, Cao Z, Spencer T, Sivasankaran R, Wang K, Nikulina E, Kimura N, Cai H, Deng K, Gao Y, et al: Myelin-associated glycoprotein interacts with the Nogo66 receptor to inhibit neurite outgrowth. Neuron. 35:283–290. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Vourc'h P and Andres C: Oligodendrocyte myelin glycoprotein (OMgp): Evolution, structure and function. Brain Res Brain Res Rev. 45:115–124. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Lipton P: Ischemic cell death in brain neurons. Physiol Rev. 79:1431–1568. 1999.PubMed/NCBI

19 

Mattson MP and Chan SL: Calcium orchestrates apoptosis. Nat Cell Biol. 5:1041–1043. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Mohan S, Abdul AB, Abdelwahab SI, Al-Zubairi AS, Sukari MA, Abdullah R, Taha Elhassan MM, Ibrahim MY and Syam S: Typhonium flagelliforme induces apoptosis in CEMss cells via activation of caspase-9, PARP cleavage and cytochrome c release: Its activation coupled with G0/G1 phase cell cycle arrest. J Ethnopharmacol. 131:592–600. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Cleary ML, Smith SD and Sklar J: Cloning and structural analysis of cDNAs for bcl-2 and a hybrid bcl-2/immunoglobulin transcript resulting from the t(14;18) translocation. Cell. 47:19–28. 1986. View Article : Google Scholar : PubMed/NCBI

22 

Otake Y, Soundararajan S, Sengupta TK, Kio EA, Smith JC, Pineda-Roman M, Stuart RK, Spicer EK and Fernandes DJ: Overexpression of nucleolin in chronic lymphocytic leukemia cells induces stabilization of bcl2 mRNA. Blood. 109:3069–3075. 2007.PubMed/NCBI

23 

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

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Salgado AJ, Sousa JC, Costa BM, Pires AO, Mateus-Pinheiro A, Teixeira FG, Pinto L and Sousa N: Mesenchymal stem cells secretome as a modulator of the neurogenic niche: Basic insights and therapeutic opportunities. Front Cell Neurosci. 9:2492015. View Article : Google Scholar : PubMed/NCBI

26 

Zhang H, Wu F, Kong X, Yang J, Chen H, Deng L, Cheng Y, Ye L, Zhu S, Zhang X, et al: Nerve growth factor improves functional recovery by inhibiting endoplasmic reticulum stress-induced neuronal apoptosis in rats with spinal cord injury. J Transl Med. 12:1302014. View Article : Google Scholar : PubMed/NCBI

27 

Johnson JE, Barde YA, Schwab M and Thoenen H: Brain-derived neurotrophic factor supports the survival of cultured rat retinal ganglion cells. J Neurosci. 6:3031–3038. 1998.

28 

Chen J, Li Y, Wang L, Zhang Z, Lu D, Lu M and Chopp M: Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats. Stroke. 32:1005–1011. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Chen Q, Long Y, Yuan X, Zou L, Sun J, Chen S, Perez-Polo JR and Yang K: Protective effects of bone marrow stromal cell transplantation in injured rodent brain: Synthesis of neurotrophic factors. J Neurosci Res. 80:611–619. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 11 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Feng N, Hao G, Yang F, Qu F, Zheng H, Liang S and Jin Y: Transplantation of mesenchymal stem cells promotes the functional recovery of the central nervous system following cerebral ischemia by inhibiting myelin‑associated inhibitor expression and neural apoptosis. Exp Ther Med 11: 1595-1600, 2016
APA
Feng, N., Hao, G., Yang, F., Qu, F., Zheng, H., Liang, S., & Jin, Y. (2016). Transplantation of mesenchymal stem cells promotes the functional recovery of the central nervous system following cerebral ischemia by inhibiting myelin‑associated inhibitor expression and neural apoptosis. Experimental and Therapeutic Medicine, 11, 1595-1600. https://doi.org/10.3892/etm.2016.3089
MLA
Feng, N., Hao, G., Yang, F., Qu, F., Zheng, H., Liang, S., Jin, Y."Transplantation of mesenchymal stem cells promotes the functional recovery of the central nervous system following cerebral ischemia by inhibiting myelin‑associated inhibitor expression and neural apoptosis". Experimental and Therapeutic Medicine 11.5 (2016): 1595-1600.
Chicago
Feng, N., Hao, G., Yang, F., Qu, F., Zheng, H., Liang, S., Jin, Y."Transplantation of mesenchymal stem cells promotes the functional recovery of the central nervous system following cerebral ischemia by inhibiting myelin‑associated inhibitor expression and neural apoptosis". Experimental and Therapeutic Medicine 11, no. 5 (2016): 1595-1600. https://doi.org/10.3892/etm.2016.3089