Abstract
We investigated the probability of newly generated neurons that could survive and mature in the ischemic hippocampal CA1 region (CA1) of a gerbil model of transient cerebral ischemia. Neuronal death was shown in the stratum pyramidale (SP) from 4 days post‐ischemia, and a significant increase in NeuN‐positive (+) neurons was found in the SP at 180 days post‐ischemia. 5‐Bromo‐2‐deoxyuridine (BrdU)+ cells were co‐stained with NeuN and glutamic decarboxylase 67 (GAD67). Brain‐derived neurotrophic factor (BDNF) immunoreactivity and protein level was shown in nonpyramidal cells from 4 days post‐ischemia, and the immunoreactivity was strong at 30 days post‐ischemia and not significantly changed until 180 days post‐ischemia. Furthermore, TrkB immunoreactivity was co‐stained with GAD67 when we examined at 180 days post‐ischemia. Myelin basic protein (MBP)+ nerve fibers were reduced at 4 days post‐ischemia and maintained until 60 days post‐ischemia, and MBP immunoreactivity and levels were significantly increased at 180 days post‐ischemia. In the passive avoidance test, cognitive dysfunction was improved at 180 days post‐ischemia. These results suggest that the differentiation of neural progenitor cells into new GABAergic neurons may be promoted via BDNF in the ischemic CA1 and that the neurogenesis may partially mediate the recovery of cognitive impairments via increasing myelinated nerve fibers.
Keywords: GABAergic neuron, hippocampal CA1, myelinated nerve fibers, transient cerebral ischemia, neurogenesis
Introduction
Transient cerebral ischemia causes fatal delayed degeneration of specific vulnerable neurons in some regions of the brain including the hippocampal CA1 region 19. Neuronal death in the hippocampal CA1 region occurs some days after transient ischemic damage and the neuronal death is referred to as “delayed neuronal death” 19. The pronounced death of the CA1 pyramidal neurons is associated with severe impairments of hippocampal‐dependent brain functions, such as spatial learning and memory 13.
Neurogenesis is important in neurological repair and in the fundamental function of the central nervous system (CNS), which contains progenitor cells that are capable of generating new neurons, astrocytes and oligodendrocytes 9, 48. In adult mammals, persistent neurogenesis has been demonstrated in two specific brain areas; the subventricular zone (SVZ), which borders the lateral ventricles, and the subgranular zone (SGZ), which is in the hippocampal dentate gyrus (DG) 10, 30, 39. Previous studies have shown that cerebral ischemia stimulates neurogenesis and the neuronal regeneration takes place from endogenous neural progenitor cells in animal models of global ischemia 18, 29, 32, 52. Although the adult CA1 region is extremely vulnerable to ischemic insults, the production of new neurons appears to be very limited in the adult brain including the CA1 region after global cerebral ischemia 42. However, the origin and differentiation of neural progenitor cells, which give rise to new neurons in the adult CNS, has been a subject of debate.
The present study aims to address whether neurogenesis following cerebral ischemia would be able to appear in the adult gerbil CA1 region. We examined long‐term changes in neuronal distribution in the CA1 region of the gerbil induced by 5 minutes of transient global cerebral ischemia and determined whether newly born cells were differentiated into specific neuronal phenotypes. Moreover, we examined whether this neurogenesis in the CA1 region was related with the re‐myelination and the recovery of cognitive impairment.
Materials and Methods
Experimental animals
We used male Mongolian gerbils (Meriones unguiculatus) obtained from the Experimental Animal Center, Kangwon National University, Chunchon, South Korea. The gerbils were used at 24 weeks (body weighting 65–75 g) of age and maintained in pathogen‐free conditions under adequate temperature (23°C) and humidity (60%) control with a 12‐h light/12‐h dark cycle. The procedures for animal handling and care adhered to guidelines that are in compliance with the current international laws and policies (Guide for the Care and Use of Laboratory Animals, The National Academies Press, 8th Ed., 2011) and they were approved by the Institutional Animal Care and Use Committee (IACUC) at Kangwon University.
Induction of transient cerebral ischemia
Transient cerebral ischemia was developed according to our previous method 25. In brief, the experimental animals were anesthetized with a mixture of 2.5% isoflurane in 33% oxygen and 67% nitrous oxide. Ischemia was induced by occluding the arteries with nontraumatic aneurysm clips (Yasargil FE 723K, Aesculap, Tuttlingen, Germany). After 5 minutes of occlusion, the aneurysm clips were removed from the common carotid arteries. The body (rectal) temperature under free‐regulating or normothermic (37 ± 0.5°C) conditions was monitored with a rectal temperature probe (TR‐100; Fine Science Tools, Foster City, CA) and maintained using a thermometric blanket before, during and after the surgery until the animals completely were recovered from anesthesia. Thereafter, animals were kept on the thermal incubator (temperature, 23°C; humidity, 60%, Mirae Medical Industry, Seoul, South Korea) to maintain the body temperature until the animals were sacrificed. Sham‐operated animals were exposed to similar surgery without carotid artery occlusion.
5‐bromo‐2′‐deoxyuridine (BrdU) administration and tissue processing
To examine the cumulative labeling of proliferative cells in the hippocampal CA1 region after transient cerebral ischemia, gerbils were received intraperitoneal BrdU injection (50 mg/kg in saline, Sigma‐Aldrich, St. Louis, MO) once a week for 2 months prior to 180 days after post‐ischemia. Sham‐ and ischemia‐operated animals (n = 7 at 4, 10, 30, 60 and 180 days after I‐R were anesthetized with pentobarbital sodium, and perfused transcardially with 0.1 M phosphate‐buffered saline (PBS, pH 7.4) followed by 4% paraformaldehyde in 0.1 phosphate‐buffer (PB, pH 7.4). The brains were removed and postfixed in the same fixative for 6 h. The brain tissues were embedded in tissue‐freezing medium and serially sectioned into 30 µm coronal sections on a cryostat (Leica, Wetzlar, Germany).
Cresyl violet staining
To investigate the morphological and neuronal changes in the hippocampus after transient cerebral ischemia, the hippocampal sections in each group were stained with cresyl violet (CV) as we descried previously 35. In brief, the sections were mounted on gelatin‐coated microscopy slides. CV acetate (Sigma, St. Louis, MO) was dissolved at 1.0% (w/v) in distilled water, and glacial acetic acid (Sigma) was added to this solution. Before and after staining for 2 minutes at room temperature, the sections were washed twice in distilled water. The fixed brain tissues were dehydrated by immersing for 2 h in 50%, 70%, 80%, 90%, 95% and 100% ethanol baths in succession at room temperature. After dehydration, the sections were mounted with Canada Balsam (Kanto, Tokyo, Japan).
Immunohistochemistry and immunofluorescence for NeuN, GAD67 BDNF and MBP
To investigate the neuronal changes in the CA1 region after transient cerebral ischemia, according to previous study 25, the NeuN+, GAD67+, BDNF+ and MBP+ staining was performed in the present study. The sections were sequentially treated with 0.3% hydrogen peroxide (H2O2) in PBS for 30 minutes and 10% normal horse serum in 0.05 M PBS for 30 minutes. The sections were then incubated with diluted mouse anti‐NeuN (1:1000, Chemicon International, Temecula, CA), mouse anti‐GAD67 (1:50, Chemicon International) rabbit anti‐BDNF (1:1000; Abcam Incorporated) and rabbit anti‐MBP (1:1000; Abcam Incorporated) overnight at room temperature. Thereafter the tissues were exposed to biotinylated horse anti‐mouse (1:200, Vector, Burlingame, CA), anti‐goat (1:200, Vector) and anti‐rabbit IgG (1:200, Vector) and streptavidin peroxidase complex (1:200, Vector). Then, the sections were visualized with 3, 3′‐diaminobenzidine tetrachloride (DAB) in 0.1 M Tris‐HCl buffer and mounted on the gelatin‐coated slides. For myelin basic protein (MBP) detection, the sections were incubated in FITC‐conjugated goat anti‐rabbit IgG (1:600; Jackson ImmunoResearch, West Grove, PA). After dehydration the sections were mounted with Canada balsam (Kanto chemical, Tokyo, Japan).
Cell counts
As previously described 24, all the measurements were performed to ensure objectivity in blind conditions (three observers for each experiment) and control and experimental samples were assayed under the same conditions. According to anatomical landmarks corresponding to AP −1.4 to −2.2 mm of the gerbil brain atlas, the studied tissue sections were selected with 120‐μm interval, and cell counts were obtained by averaging the total cell numbers 15 sections taken from each animal per group. The number of NeuN‐positive (+) and GAD67+ neurons was counted in a 250 × 250 µm square, applied approximately at the center of the CA1 region in the stratum pyramidale using an image analyzing system (software: Optimas 6.5, CyberMetrics, Scottsdale, AZ). Cell counts were obtained by averaging the total number of NeuN‐immunoreactive neurons from each animal per group.
To quantitatively analyze BDNF and MBP immunoreactivities, the corresponding areas of the CA1 region were measured from 15 sections per animal. Images of all BDNF‐immunoreactive structures were taken from three layers (strata oriens, pyramidale and radiatum in the hippocampus proper) through an AxioM1 light microscope (Carl Zeiss, Göttingen, Germany) equipped with a digital camera (Axiocam, Carl Zeiss) connected to a PC monitor. Images were calibrated into an array of 512 × 512 pixels corresponding to a tissue area of 250 × 250 µm (40 × primary magnifications). The densities of all BDNF+ and MBP+ structures were evaluated by optical density (OD), which was obtained after the transformation of the mean gray level using the formula: OD = log (256/mean gray level). The background OD was taken from areas adjacent to the measured area. After the background density was subtracted, a ratio of the OD of the image file was calibrated as % [relative optical density, (ROD)] using Adobe Photoshop version 8.0 and then analyzed using NIH Image 1.59 software.
Double immunofluorescence for BrdU/NeuN or GAD67, BDNF/GFAP or Iba‐1 and TrkB/GAD67
To confirm the differentiation from newly generated cells to mature GABAergic neurons and the cell type containing TrkB immunoreactivity, the sections at 180 days after ischemia were used for double immunofluorescence staining with BrdU/NeuN, BrdU/GAD67 and TrkB/GAD67. In addition, we performed double immunofluorescence staining for BDNF/GFAP and BDNF/Iba‐1 to identify the cell type of nonpyramidal cells expressing BDNF at 30 days after ischemia. In brief, DNA denaturation was conducted, and then sections were incubated in 2N HCl and in boric acid (only for BrdU staining). The sections were incubated with the mixture of diluted mouse anti‐NeuN (1:400, Chemicon International), rat anti‐BrdU (1:250, Serotec, Bicester, UK), rabbit anti‐TrkB (1:50, Abcam Incorporated), mouse anti‐GAD67 (1:50, Chemicon International), rabbit anti‐BDNF (1:200, Santa Cruz Biotechnology), mouse anti‐glial fibrillary acidic protein (GFAP) (1:400, Chemicon International) and goat anti‐ionized calcium‐binding adapter molecule 1 (Iba‐1) (1:500, Santa Cruz Biotechnology) overnight at 4°C. After washing three times for 10 minutes with PBS, the sections were incubated in a mixture of both FITC‐conjugated goat anti‐rabbit IgG (1:600; Jackson ImmunoResearch), FITC‐conjugated donkey anti‐rat IgG (1:600; Jackson ImmunoResearch), Cy3‐conjugated donkey anti‐mouse IgG (1:200; Jackson ImmunoResearch) and Cy3‐ conjugated donkey anti‐goat IgG (1:200; Jackson ImmunoResearch) for 2 h at room temperature. The immunoreactions were observed under the confocal microscope (LSM510 META NLO, Carl Zeiss). Negative control test was also performed using preimmune serum instead of primary antibody to establish the specificity of the immunostaining. The negative control resulted in the absence of immunoreactivity in all structures. For calculating the number of BrdU+/NeuN+ cells and BrdU+/GAD67+ cells in the CA1 region, z stack images were taken from the tissue area of 200 × 200 µm with 30 µm thick 10 sections per animal. Cell counts were performed following the above‐mentioned method.
Western blot analysis
To confirm changes in level of BDNF and MBP proteins in the hippocampus after transient cerebral ischemia, at designated times (4, 30 and 180 days after the surgery), the animals (n = 7 at each point in time) were sacrificed and used for the western blot analysis. After sacrificing them and removing the hippocampus, the tissues were homogenized in 50 mM PBS (pH 7.4) containing 0.1 mM ethylene glycol bis (2‐aminoethyl ether)‐N,N,N′,N′ tetraacetic acid (EGTA) (pH 8.0), 0.2% Nonidet P‐40, 10 mM ethylendiamine tetraacetic acid (EDTA) (pH 8.0), 15 mM sodium pyrophosphate, 100 mM β‐glycerophosphate, 50 mM NaF, 150 mM NaCl, 2 mM sodium orthovanadate, 1 mM phenylmethylsulfonyl fluoride (PMSF) and 1 mM dithiothreitol (DTT). After centrifugation, the protein level was determined in the supernatants using a Micro BCA protein assay kit with bovine serum albumin as the standard (Pierce Chemical, Rockford, IL). Aliquots containing 20 µg of total protein were boiled in loading buffer containing 150 mM Tris (pH 6.8), 3 mM DTT, 6% SDS, 0.3% bromophenol blue and 30% glycerol. The aliquots were then loaded onto a 10% polyacrylamide gel. After electrophoresis, the gels were transferred to nitrocellulose transfer membranes (Pall Crop, East Hills, NY). To reduce background staining, the membranes were incubated with 5% nonfat dry milk in PBS containing 0.1% Tween 20 for 45 minutes, followed by incubation with rabbit anti‐BDNF antibody (1:2000; Abcam Incorporated) or rabbit anti‐MBP (1:2000; Abcam Incorporated), peroxidase‐conjugated goat anti‐rabbit IgG (Vector, Cat# PI‐1000) and ECL kit (Pierce Chemical). Loading controls were performed using antibodies against beta actin (Abcam Incorporated). The results of the western blot analysis were scanned, and densitometric analysis for the quantification of the bands was done using Scion Image software (Scion Corp., Frederick, MD), which was used to count ROD: A ratio of the ROD was calibrated as %, with sham‐group designated as 100%.
Passive avoidance test
Short‐term memory ability was evaluated by assessing the latency of the passive avoidance test. This test was performed using passive avoidance apparatus (GEM 392, San Diego instruments) according to the method of Sharma et al 44 with slight modification. The apparatus is divided into two sections. One compartment is large, white and illuminated, the other is small, dark and without illumination. The two compartments communicate through a sliding door system and the floor is a steel grid in both compartments. The bars of the dark compartment floor are wired to a constant current scrambler circuit. The test was divided into a training and test session. The experimental session was divided into three phases: habituation trial, acquisition trial and retention trial. During the habituation trial, which was executed on the third day after ischemic insults, the gerbil was placed in the white and illuminated compartment. In this phase, the sliding door was initially closed and it opened after 5 s. The gerbil can then explore both compartments for 60 s and after this period it was taken off the apparatus. After 10 minutes, it was placed again in the white compartment. The sliding door opened after 5 s and successively closed when the gerbil crossed the cage, entering the dark room, where it remained for 10 s, then was removed from the cage. The acquisition trial was performed 60 minutes after the habituation trial. In this phase, the gerbil was replaced in the white room and when it crossed the sliding door entering the dark room, it received an electric shock (2 mA for 5 s), released from the grid. Twenty‐four hours later, the retention trial was performed. The gerbils were replaced in the white room and the sliding doors opened as the acquisition trial. The latency times for entering the dark compartment and the number of error times were recorded. When an animal did not enter the dark room within 180 s, the latency was recorded as 180 s. Response latency was measured as an index of learning ability. Passive avoidance test were performed at 1 day before ischemia‐reperfusion, 4, 10, 30, 60 and 180 days after I‐R. On the 3rd, 9th, 29th, 59th and 179th days, the retention trial was performed again.
Statistical analysis
Data are expressed as the mean ± standard error of the mean. The data of cell counts were evaluated by a one‐way ANOVA SPSS program and comparisons among groups were made using parametric two‐way ANOVA. Further comparisons were assessed using Duncan's multiple‐range test. The latency of entrance to the dark compartment of the passive avoidance test was analyzed using a Kruskal–Wallis nonparametric one‐way analysis of variance, followed by a two‐tailed Mann–Whitney U test. Statistical significance was set at P < 0.05.
Results
Changes of cresyl violet‐positive cells
Neuronal death in the hippocampus after transient cerebral ischemia was examined using CV staining. In the sham‐operated group, neurons stained with CV were easily observed in all of the subregions of the hippocampus. In the ischemia‐operated groups, a loss of cresyl violet‐positive (CV+) pyramidal neurons was observed in the stratum pyramidale of the entire CA1 region; however, neurons in the other subregions were well stained with CV (Figure 1A).
Figure 1.

Changes of CV‐positive (CV+) cells and NeuN‐immunoreactive (NeuN+) neurons in the hippocampal CA1 region after I‐R. A. CV staning in the hippocampus of the sham‐ and ischemia‐operated groups. CV+ cells are dramatically decreased only in the CA1 region after ischemia. CA, cornus ammonis; DG, dentate gyrus. Scale bar = 400 µm. B. Immunohistochemistry for NeuN in the CA1 region of the sham‐ and ischemia‐operated groups. SP, stratum pyramidale; SO, stratum oriens and SR, stratum radiatum. Scale bar = 50 µm. C. The mean number of NeuN+ neurons/250 × 250 µm of the CA1 region at sham, 4, 10, 30, 60 and 180 days after I‐R (n = 7 per group; *P < 0.05, significantly different from the sham‐group; #P < 0.05, significantly different from the former group). The bars indicate the means ± SEM. D. Double immunofluorescence staining for BrdU (green), NeuN (red) and merged images (yellow) in the CA1 region 180 days after I‐R. Most of the BrdU+ cells are colocalized with NeuN+ neurons (arrows). Scale bar = 50 μm. E. The mean number of NeuN+, BrdU+ and BrdU+/NeuN+ cells/200 × 200 µm of the CA1 region 180 days after I‐R. (n = 7 per group). The bars indicate the means ± SEM.
Changes of NeuN‐immunoreactive neurons
Delayed neuronal death in the CA1 region following ischemia‐reperfusion (I‐R) was examined using NeuN immunohistochemistry. In the sham‐operated group, neurons in the CA1 region were well stained showing the presence of NeuN (92 ± 4.52 cells/250 × 250 µm, Figure 1B,C). Four days after I‐R, NeuN‐immunoreactive (NeuN+) neurons were hardly detected in the CA1 region due to the delayed cell death of pyramidal neurons in the stratum pyramidale (7 ± 1.05 cells/250 × 250 µm, Figure 1B,C). Thereafter, there were no significant changes in NeuN immunoreactivity and in the number of NeuN+ neurons in the CA1 region for 60 days after I‐R (Figure 1B,C). However, there was a significant increase in the number of NeuN+ neurons in the CA1 region 180 days after I‐R (24 ± 3.32 cells/250 × 250 µm, Figure 1B,C); the NeuN+ neurons were generally distributed near or within the stratum pyramidale. To elucidate that BrdU+ cells were newly generated and transformed into mature neurons, double immunofluorescence staining was performed in the CA1 region 180 days after I‐R. All of the BrdU+ cells revealed co‐expression with NeuN+ cells in the CA1 region 180 days after I‐R (Figure 1D,E).
Changes of glutamic acid decarboxylase 67 cells
In the sham‐operated group, glutamic acid decarboxylase 67 (GAD67+) neurons were detected near the stratum pyramidale of the CA1 region (4.3 ± 0.51 cells/250 × 250 µm, Figure 2A,B). In the ischemia‐operated groups, four days after I‐R, GAD67 immunoreactivity and GAD67+ cells were similar to that the sham‐operated group (3.9 ± 0.42 cells/250 × 250 µm, Figure 2A,B). Until 60 days after I‐R, GAD67 immunoreactivity and its pattern were not changed to those 4 days after I‐R (Figure 2A,B). At 180 days after I‐R, the GAD67 immunoreactivity and GAD67+ cells were significantly increased near the stratum pyramidale compare with those 4 days after I‐R (11.3 ± 0.53 cells/250 × 250 µm, Figure 2A,B). In addition, when the double labeling of BrdU and GAD67 at180 days after I‐R was performed, all of the BrdU+ cells in the ischemic CA1 region were co‐labeled with GAD67 (Figure 2C,D).
Figure 2.

Changes of GAD67‐immunoreactive (GAD67+) neurons in the hippocampal CA1 region after I‐R. A. Immunohistochemistry for GAD67 in the CA1 region of the sham‐ and ischemia‐operated groups. SP, stratum pyramidale; SO, stratum oriens and SR, stratum radiatum. Scale bar = 50 µm. B. The mean number of GAD67+ neurons/250 × 250 µm of the CA1 region at sham, 4, 10, 30, 60 and 180 days after I‐R (n = 7 per group; *P < 0.05, significantly different from the sham‐group; #P < 0.05, significantly different from the former group). The bars indicate the means ± SEM. C. Double immunofluorescence staining for BrdU (green), GAD67 (red) and merged images (yellow) in the CA1 region 180 days after I‐R. All of the BrdU+ cells are co‐labeled with GAD67+ neurons (arrows). Scale bar = 50 μm. D. The mean number of BrdU+, GAD67+ and BrdU+/GAD67+ cells/200 × 200 µm of the CA1 region 180 days after I‐R. (n = 7 per group). The bars indicate the means ± SEM.
Changes of brain‐derived neurotrophic factor immunoreactivity and protein level
In the sham‐operated group, BDNF immunoreaction was presented in the neurons of stratum pyramidale in the CA1 region (Figure 3A,B). Four days after I‐R, a conspicuous reduction of BDNF immunoreactivity was noted in the stratum pyramidale, and strong BDNF immunoreaction was expressed in nonpyramidal cells in the strata oriens and radiatum 30 days after I‐R (Figure 3A,C). To identify the cell type of nonpyramidal cells expressing BDNF 30 days after I‐R, double immunofluorescence staining for BDNF/GFAP or BDNF/Iba‐1 was performed and we found that BDNF immunoreactive cells were colocalized with GFAP+ astrocytes (Figure 3B); however, BDNF immunoreactivity was not found in the Iba‐1+ microglia (data not shown). Thereafter, the distribution pattern of the distribution of BDNF+ cells until 180 days after I‐R was similar those 4 days after I‐R, although BDNF immunoreactivity was a little changed with time (Figure 3A,C).
Figure 3.

Changes of BDNF‐immunoreactive (BDNF+) neurons in the hippocampal CA1 region after I‐R. A. Immunohistochemistry for BDNF in the CA1 region of the sham‐ and ischemia‐operated groups. SP, stratum pyramidale; SO, stratum oriens and SR, stratum radiatum. Scale bar = 50 µm. B. Double immunofluorescence staining for BDNF (green), GFAP (red) and merged images (yellow) in the CA1 region 30 days after I‐R. BDNF immunoreactivity is shown in GFAP+ positive astrocytes (arrows). Scale bar = 50 μm. The bars indicate the means ± SEM. C. Relative optical density (ROD) of BDNF immunoreactivity of the CA1 region at sham, 4, 10, 30, 60 and 180 days after I‐R. The ROD as % values of sham is represented (n = 7 per group; *P < 0.05, significantly different from the sham‐group). The bars indicate the means ± SEM. D. Western blot analysis of BDNF in the CA1 region of the sham‐ and ischemia‐operated groups. ROD as % values of immunoblot band is also represented (n = 7 per group; *P < 0.05, significantly different from the sham‐group; #P < 0.05, significantly different from the former group). E. Double immunofluorescence staining for TrkB (green), GAD67 (red) and merged images (yellow) in the CA1 region 180 days after I‐R. Most of the TrkB+ cells are colocalized with GAD67+ cells (arrows). Scale bar = 50 μm. The bars indicate the means ± SEM.
Western blot analysis showed that the pattern of change in BDNF protein level in the CA1 region after I‐R was similar to that observed in the immunohistochemical data. In the ischemia‐operated groups, BDNF protein level was markedly increased 30 days after I‐R compared with that in the sham‐operated group. Thereafter, BDNF protein level was not significantly changed until 180 days post‐ischemia (Figure 3D). In addition, to elucidate that newly generated cells express BDNF receptor, TrkB, double immunofluorescence staining was performed in the CA1 region 180 days after I‐R. Double labeled TrkB+/GAD67+ cells were examined near the stratum pyramidale of the CA1 region 180 days after I‐R (Figure 3E).
Changes of myelin basic protein immunoreactivity and protein level
The assessment of myelin was evaluated by immunohistochemistry and western blot for MBP in the CA1 region after I‐R. In the sham‐operated group, MBP+ myelinated nerve fibers were easily observed in all of the layers of the CA1 region, especially, in the strata oriens and radiatum (Figure 4A). In the ischemia‐operated groups, MBP+ myelinated nerve fibers were decreased in the CA1 region from 4 days after I‐R (Figure 4A,B). At 10 days post‐ischemia, MBP+ myelinated nerve fibers were significantly lower (28% vs. sham) than those 4 days after I‐R, and MBP+ myelinated nerve fibers were not significantly changed until 60 days after I‐R (Figure 4A,B). However, 180 days after I‐R, MBP+ myelinated nerve fibers were significantly increased (44% vs. sham) in the CA1 region compared with those at 60 days post‐ischemia (Figure 4A,B).
Figure 4.

Changes of MBP‐immunoreactive (MBP+) neurons in the hippocampal CA1 region after I‐R. A. Immunohistochemistry for MBP in the CA1 region of the sham‐ and ischemia‐operated groups. SP, stratum pyramidale; SO, stratum oriens and SR, stratum radiatum. Scale bar = 50 µm. B. Relative optical density (ROD) of MBP+ myelinated fibers of the CA1 region at sham, 4, 10, 30, 60 and 180 days after I‐R. ROD as % values of Sham is represented (n = 7 per group; *P < 0.05, significantly different from the sham‐group; #P < 0.05, significantly different from the former group). The bars indicate the means ± SEM. C. Western blot analysis of MBP in the CA1 region of the sham‐ and ischemia‐operated groups. The ROD as % values of immunoblot band is also represented (n = 7 per group; *P < 0.05, significantly different from the sham‐group; #P < 0.05, significantly different from the former group). The bars indicate the means ± SEM.
Conversely, western blot analysis showed that the pattern of change in MBP protein level in the CA1 region after I‐R was similar to that observed in the immunohistochemical data (Figures 4C).
Passive avoidance response after global ischemia
To examine whether the formation of new CA1 neurons was related with any functional recovery, we observed hippocampal‐dependent learning and memory performance 1 day before I‐R, and 4, 10, 30, 60 and 180 days after I‐R. The mean latency time of the passive avoidance response in the retention trial was 52.2 ± 12.4 s 1 day before I‐R (Figure 5). Transient global cerebral ischemia significantly induces shorter escape latencies. At 4, 10, 30 and 60 days after I‐R, the latency time is decreased by 14.1 ± 9.5 s, 12.8 ± 6.2 s, 11.5 ± 5.4 s, 11.7 ± 4.1 s (P < 0.05 vs. 1 day before I‐R); however, 180 days after I‐R, the latency time was significantly increased by 24.5 ± 9.2 s (Figure 5).
Figure 5.

Escape latency of entrance to the dark compartment of passive avoidance apparatus during memory retention test 1 day before I‐R, 4, 10, 30, 60 and 180 days after I‐R. Although a significant reduction in response latency after I‐R is observed, there is a significant improvement in response latency 180 days after I‐R. (n = 7 per group; *P < 0.05, significantly different from the sham‐group; #P < 0.05, significantly different from the former group). The bars indicate the means ± SEM.
Discussion
Over the past several years, stroke‐stimulated endogenous neurogenesis has been applied as one of effective strategies for the treatment of stroke associated with aging 2, 36. Cerebral ischemia is known to stimulate the proliferation and differentiation of neural precursor cells, which have a potential to enable regeneration and restoration; however, the functional relation of this phenomenon is not clear because of the poor survival and low neuronal differentiation rate of newly born cells in the brain, especially, in the aged brain 8, 15. Furthermore, it has been reported that hypoxia could induce neurogenesis in the hippocampus of the rodent 5, 43. Conversely, it has been demonstrated that transplanted neural precursor cells could increase endogenous neurogenesis after cerebral ischemia in the aged rat and suggested that cell based therapy for stroke might be able to increase endogenous adaptive processes including neurogenesis 16. On the basis of the above reports, in this study, we examined neurogenesis in the gerbil hippocampal CA1 region for a long time after transient cerebral ischemia.
In the present study, a massive regeneration of GAD+ neurons was found in the CA1 region very long time later following transient cerebral ischemic insult, which might be due to the lack of the process of spontaneously endogenous neurogenesis. Interestingly, we found that newly born NeuN+, BrdU+ and GAD67+ cells, which were co‐localized with TrkB, were distributed near the stratum pyramidale 180 days after I‐R and that the neurogenesis might be stimulated by increasing the production of endogenous BDNF. Moreover, this neurogenesis might mediate new myelination and the recovery of cognitive impairments.
Several studies have documented that experimental ischemia in the adult brain can trigger neurogenesis as a compensatory mechanism for neural cell death 27. Liu et al 29 reported an increase in cell proliferation in the hippocampal DG after transient global ischemia in the adult gerbil. A 12‐fold increase in cell birth in the DG was observed 1–2 weeks after 10 minutes of global ischemia; in this case, the neurogenesis represented an amplification of normal neurogenesis rather than the induction of neurogenesis in a normally non‐neurogenic area. However, such neurogenesis does not serve to replace the loss of the CA1 pyramidal neurons induced by transient ischemia. Nevertheless, it is of interest to note that the spontaneous regeneration of CA1 pyramidal neurons occurs after ischemic injury. Nakatomi et al 32 demonstrated the spontaneous regeneration of CA1 neurons after global cerebral ischemia using both BrdU administration and retroviral injection into the periventricular region. In addition, many studies have shown that an increased proliferation of neural progenitor cells starts at 3–4 days post‐ischemia, peaks at 7–10 days post‐ischemia and returns to the control level by 3–5 weeks after global cerebral ischemia 29, 47, 50, 52. However, Bendel and coworkers 37 reported that about 40% of CA1 neurons had reappeared 90 days after transient ischemic insult, whereas some researchers reported that the tertiary cell death had appeared between 90 and 125 days after ischemic damage 3, 51. Furthermore, Bueters et al 4 reporetd that newly formed CA1 cells had partly disappeared 250 days after ischemia. Considering their researches, we could not tell how new neurogenesis occurs in ischemic region with time after I‐R. In this study, we observed that many NeuN+ neurons were found near the stratum pyramidale of the CA1 region 180 days post‐ischemia and the NeuN+ neurons were colocalized with BrdU+ cells. Although BrdU immunocytochemistry is a powerful method in studies regarding adult neurogenesis, it has some shortcomings that can complicate studies on adult neurogenesis following brain injuries 33, 38. It is of more concern that damaged neurons in ischemic regions may re‐enter the cell cycle in a process of abortive DNA synthesis prior to dying via apoptosis 21. Thus, BrdU+ neurons in ischemic region cannot be assumed to be newly born. However, it has been thought that such damaged cells always die within 28 days 21, so analyses at longer points in time should eliminate such concerns. In addition, recent data suggest that some postmitotic neurons may survive long after taking up BrdU by DNA synthesis 6. We, in this study, found the significant increase of BrdU+/NeuN+ neurons near the stratum pyramidale of the CA1 region 180 days after I‐R. In addition, we recently reported that F‐J B+ cells in the stratum pyramidale were hardly observed from 60 days after I‐R 24. These findings are in agreement with the report by Teramoto et al 49, which showed a gradual increase of BrdU/NeuN double‐positive neurons up to 13 weeks after focal ischemia. Taken together, these findings suggest that NeuN+ cells are produced during the division of neural progenitor cells rather than through the repair process of preexisting neurons. Therefore, our present study indicates that neurogenesis in the ischemic CA1 region occurs very late after transient cerebral ischemia.
Another fundamental feature is that adult neural progenitor cells can regenerate into specific neuronal subtypes, which are appropriate for damaged sites. In the hippocampal DG, most of newly formed dentate granule cells differentiate into NeuN+ neurons by 3–4 weeks after ischemia 20. In the striatum, partial progenitor cells migrate into the striatum and develop into GAD67+ GABAergic and acetyltransferase+ cholinergic neurons 26. Furthermore, in the hippocampus proper, neurogenesis in adults may include some hippocampal cell types, which contain GABAergic neurons 41. On the basis of these findings, in this study, we found observed a significant increase of GABAergic neurons in the CA1 region 180 days after I‐R using GAD67 that is well known as one of excellent immunohistochemical markers for identifying GABAergic neurons 45, 46. In addition, we found that all of the BrdU+ cells were colocalized with GAD67 in the ischemic CA1 region 180 days after I‐R via double immunofluorescence staining for BrdU/GAD67. This finding indicates that a renewal of GABAergic neurons would contribute to plasticity and help to maintain the integrity of neural networks in the adult brain 11, because it was reported that newly generated neurons could develop into mature neurons displaying functional neurotransmission after cerebral ischemia in the adult brain 14.
Interestingly, BDNF acts as a powerful trophic agent for GABAergic population. Actually, BDNF increases the differentiation of hippocampal and neocortical inhibitory interneurons, promotes a GABAergic phenotype in hippocampal granule cells 12, 28, and enhances the expression of GAD67 in primary hippocampal cultures obtained from adult mice 1. In addition, BDNF stimulates neurogenesis in the SGZ or SVZ regions following cerebral ischemia 7, 40. In the present study, we investigated the change of BDNF immunoreactivity and its level in the ischemic CA1 region to explain its relationship with the increase of neurogenesis after I‐R. We found a significant increase of BDNF expression in the nonpyramidal cells, which were colocalized with GFAP+ astrocytes, at 30 days post‐ischemia, and the increased BDNF expression was maintained until 180 days post‐ischemia. It was reported that BDNF was expressed in astrocytes in the mouse hippocampus following transient cerebral ischemia and suggested that the increased BDNF production in astrocytes might be related with the enhancement of hippocampal neurogenesis after brain ischemia 34. Therefore, we state that BDNF mediates, at least in part, the enhancement of hippocampal neurogenesis induced by an ischemic insult, although neural progenitor cells may not produce BDNF; newly generated cells express BDNF receptor (TrkB) and can respond to BDNF 22. Conversely, to better understand the mechanism controlling cell differentiation in the ischemic CA1 region, we found that GAD67+ cells were co‐localized with TrkB when we examined 180 days post‐ischemia. Therefore, our observation suggests that BDNF may act on neural progenitor cells to promote their differentiation into GABAergic neurons via TrkB.
The pronounced deletion of CA1 pyramidal neurons is associated with severe impairments of hippocampal‐dependent brain functions 23. Cognitive function in ischemic animals is impaired at 14 days post‐ischemia, and the cognitive dysfunction subsides at 90 or 125 days post‐ischemia 3, 51. In particular, ischemic hippocampal damage leads to cognitive dysfunction as an evident from reduction in response latency in passive avoidance test 17. As expected, we found that cognitive dysfunction, as evaluated by the passive avoidance test, appeared from 4 days to 60 days after ischemia and improved significantly at 180 days post‐ischemia. This possibility is supported by a significant increase in NeuN+ neurons that were found at 180 days after post‐ischemia as compared with those at 60 days post‐ischemia. In addition, recent studies have demonstrated that enhanced learning and memory functions as well as behavioral effects are associated with neurogenesis in the hippocampus following cerebral ischemia 3.
Conversely, myelin is responsible not only for the protection and insulation of axons but also for the production of supportive neurotropic factors. Thus, a loss of myelin displays an adverse effect on axonal function and neuronal survival, and myelination may play an important role in cognitive function 31. In the present study, MBP staining showed that MBP immunoreactivity in the ischemic CA1 region was decreased from 4 days after I‐R, persisted until at least 60 days post‐ischemia and the immunoreactivity was restored to about 40% of the sham‐group 180 days after I‐R. Therefore, some renewal of myelinated nerve fibers in the ischemic CA1 region could contribute to the recovery of cognitive impairments, which suggests that the new CA1 neurons can restore disrupted neuronal circuits following cerebral ischemia.
In conclusion, our study provides the first evidence in the development of newly‐generated GABAergic neurons in the ischemic CA1 region and may enhance our understanding of neurogenesis in ischemic adult brain. Taken together, our findings suggest that increasing BDNF can enhance the induction of GABAergic neurons in the ischemic CA1 region very long time later after I‐R and that the renewal of GABAergic neurons in the ischemic CA1 region can partially restore disrupted neuronal circuit following cerebral ischemia.
Acknowledgments
The authors would like to thank Mr. Seung Uk Lee for their technical help. This research was supported by the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (2010‐0010580), by the Bio‐Synergy Research Project (NRF‐2014M3A9C4066454) of the Ministry of Science, ICT and Future Planning through the National Research Foundation, and by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT and future Planning (NRF‐2013R1A2A2A01068190).
Conflict of interests The authors declare that there are no conflicts of interest.
References
- 1. Babu H, Cheung G, Kettenmann H, Palmer TD, Kempermann G (2007) Enriched monolayer precursor cell cultures from micro‐dissected adult mouse dentate gyrus yield functional granule cell‐like neurons. PloS one 2:e388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Balseanu AT, Buga AM, Catalin B, Wagner DC, Boltze J, Zagrean AM et al (2014) Multimodal approaches for regenerative stroke therapies: combination of granulocyte colony‐stimulating factor with bone marrow mesenchymal stem cells is not superior to G‐CSF alone. Front Aging Neurosci 6:130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Bendel O, Alkass K, Bueters T, von Euler M, von Euler G (2005) Reproducible loss of CA1 neurons following carotid artery occlusion combined with halothane‐induced hypotension. Brain Res 1033:135–142. [DOI] [PubMed] [Google Scholar]
- 4. Bueters T, von Euler M, Bendel O, von Euler G (2008) Degeneration of newly formed CA1 neurons following global ischemia in the rat. Exp Neurol 209:114–124. [DOI] [PubMed] [Google Scholar]
- 5. Buga AM, Vintilescu R, Balseanu AT, Pop OT, Streba C, Toescu E, Popa‐Wagner A (2012) Repeated PTZ treatment at 25‐day intervals leads to a highly efficient accumulation of doublecortin in the dorsal hippocampus of rats. PloS One 7:e39302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Burns TC, Ortiz‐Gonzalez XR, Gutierrez‐Perez M, Keene CD, Sharda R, Demorest ZL et al (2006) Thymidine analogs are transferred from prelabeled donor to host cells in the central nervous system after transplantation: a word of caution. Stem Cells 24:1121–1127. [DOI] [PubMed] [Google Scholar]
- 7. Chen J, Zacharek A, Zhang C, Jiang H, Li Y, Roberts C et al (2005) Endothelial nitric oxide synthase regulates brain‐derived neurotrophic factor expression and neurogenesis after stroke in mice. J Neurosci 25:2366–2375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Choi JH, Yoo KY, Lee CH, Park JH, Yan BC, Kwon SH et al (2012) Comparison of neurogenesis in the dentate gyrus between the adult and aged gerbil following transient global cerebral ischemia. Neurochem Res 37:802–810. [DOI] [PubMed] [Google Scholar]
- 9. Emsley JG, Mitchell BD, Kempermann G, Macklis JD (2005) Adult neurogenesis and repair of the adult CNS with neural progenitors, precursors, and stem cells. Prog Neurobiol 75:321–341. [DOI] [PubMed] [Google Scholar]
- 10. Eriksson PS, Perfilieva E, Bjork‐Eriksson T, Alborn AM, Nordborg C, Peterson DA, Gage FH (1998) Neurogenesis in the adult human hippocampus. Nat Med 4:1313–1317. [DOI] [PubMed] [Google Scholar]
- 11. Ge S, Goh EL, Sailor KA, Kitabatake Y, Ming GL, Song H (2006) GABA regulates synaptic integration of newly generated neurons in the adult brain. Nature 439:589–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Gutierrez R (2005) The dual glutamatergic‐GABAergic phenotype of hippocampal granule cells. Trends Neurosci 28:297–303. [DOI] [PubMed] [Google Scholar]
- 13. Hartman RE, Lee JM, Zipfel GJ, Wozniak DF (2005) Characterizing learning deficits and hippocampal neuron loss following transient global cerebral ischemia in rats. Brain Res 1043:48–56. [DOI] [PubMed] [Google Scholar]
- 14. Hou SW, Wang YQ, Xu M, Shen DH, Wang JJ, Huang F et al (2008) Functional integration of newly generated neurons into striatum after cerebral ischemia in the adult rat brain. Stroke 39:2837–2844. [DOI] [PubMed] [Google Scholar]
- 15. Jin K, Minami M, Xie L, Sun Y, Mao XO, Wang Y et al (2004) Ischemia‐induced neurogenesis is preserved but reduced in the aged rodent brain. Aging cell 3:373–377. [DOI] [PubMed] [Google Scholar]
- 16. Jin K, Xie L, Mao X, Greenberg MB, Moore A, Peng B et al (2011) Effect of human neural precursor cell transplantation on endogenous neurogenesis after focal cerebral ischemia in the rat. Brain Res 1374:56–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Karasawa Y, Araki H, Otomo S (1994) Changes in locomotor activity and passive avoidance task performance induced by cerebral ischemia in Mongolian gerbils. Stroke 25:645–650. [DOI] [PubMed] [Google Scholar]
- 18. Kee NJ, Preston E, Wojtowicz JM (2001) Enhanced neurogenesis after transient global ischemia in the dentate gyrus of the rat. Exp Brain Res 136:313–320. [DOI] [PubMed] [Google Scholar]
- 19. Kirino T (1982) Delayed neuronal death in the gerbil hippocampus following ischemia. Brain Res 239:57–69. [DOI] [PubMed] [Google Scholar]
- 20. Komitova M, Perfilieva E, Mattsson B, Eriksson PS, Johansson BB (2002) Effects of cortical ischemia and postischemic environmental enrichment on hippocampal cell genesis and differentiation in the adult rat. J Cereb Blood Flow Metab 22:852–860. [DOI] [PubMed] [Google Scholar]
- 21. Kuan CY, Schloemer AJ, Lu A, Burns KA, Weng WL, Williams MT et al (2004) Hypoxia‐ischemia induces DNA synthesis without cell proliferation in dying neurons in adult rodent brain. J Neurosci 24:10763–10772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Lachyankar MB, Condon PJ, Quesenberry PJ, Litofsky NS, Recht LD, Ross AH (1997) Embryonic precursor cells that express Trk receptors: induction of different cell fates by NGF, BDNF, NT‐3, and CNTF. Exp Neurol 144:350–360. [DOI] [PubMed] [Google Scholar]
- 23. Langdon KD, Granter‐Button S, Corbett D (2008) Persistent behavioral impairments and neuroinflammation following global ischemia in the rat. Eur J Neurosci 28:2310–2318. [DOI] [PubMed] [Google Scholar]
- 24. Lee CH, Moon SM, Yoo KY, Choi JH, Park OK, Hwang IK, Sohn Y, Moon JB, Cho JH, Won MH (2010) Long‐term changes in neuronal degeneration and microglial activation in the hippocampal CA1 region after experimental transient cerebral ischemic damage. Brain Res 1342:138–149. [DOI] [PubMed] [Google Scholar]
- 25. Lee CH, Park JH, Yoo KY, Choi JH, Hwang IK, Ryu PD, Kim DH et al (2011) Pre‐ and post‐treatments with escitalopram protect against experimental ischemic neuronal damage via regulation of BDNF expression and oxidative stress. Exp Neurol 229:450–459. [DOI] [PubMed] [Google Scholar]
- 26. Li Y, Blanco GD, Lei Z, Xu ZC (2010) Increased GAD expression in the striatum after transient cerebral ischemia. Mol Cell Neurosci 45:370–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Lichtenwalner RJ, Parent JM (2006) Adult neurogenesis and the ischemic forebrain. J Cereb Blood Flow Metab 26:1–20. [DOI] [PubMed] [Google Scholar]
- 28. Lin PY, Hinterneder JM, Rollor SR, Birren SJ (2007) Non‐cell‐autonomous regulation of GABAergic neuron development by neurotrophins and the p75 receptor. J Neurosci 27:12787–12796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Liu J, Solway K, Messing RO, Sharp FR (1998) Increased neurogenesis in the dentate gyrus after transient global ischemia in gerbils. J Neurosci 18:7768–7778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Lois C, Alvarez‐Buylla A (1994) Long‐distance neuronal migration in the adult mammalian brain. Science 264:1145–1148. [DOI] [PubMed] [Google Scholar]
- 31. Miki K, Ishibashi S, Sun L, Xu H, Ohashi W, Kuroiwa T, Mizusawa H (2009) Intensity of chronic cerebral hypoperfusion determines white/gray matter injury and cognitive/motor dysfunction in mice. J Neurosci Res 87:1270–1281. [DOI] [PubMed] [Google Scholar]
- 32. Nakatomi H, Kuriu T, Okabe S, Yamamoto S, Hatano O, Kawahara N et al (2002) Regeneration of hippocampal pyramidal neurons after ischemic brain injury by recruitment of endogenous neural progenitors. Cell 110:429–441. [DOI] [PubMed] [Google Scholar]
- 33. Nowakowski RS, Hayes NL (2000) New neurons: extraordinary evidence or extraordinary conclusion? Science 288:771. [DOI] [PubMed] [Google Scholar]
- 34. Okuyama S, Shimada N, Kaji M, Morita M, Miyoshi K, Minami S et al (2012) Heptamethoxyflavone, a citrus flavonoid, enhances brain‐derived neurotrophic factor production and neurogenesis in the hippocampus following cerebral global ischemia in mice. Neurosci Lett 528:190–195. [DOI] [PubMed] [Google Scholar]
- 35. Park JH, Joo HS, Yoo KY, Shin BN, Kim IH, Lee CH et al (2011) Extract from Terminalia chebula seeds protect against experimental ischemic neuronal damage via maintaining SODs and BDNF levels. Neurochem Res 36:2043–2050. [DOI] [PubMed] [Google Scholar]
- 36. Popa‐Wagner A, Buga AM, Doeppner TR, Hermann DM (2014) Stem cell therapies in preclinical models of stroke associated with aging. Front cell Neurosci 8:347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Rahimi A, Keilig L, Bendels G, Klein R, Buzug TM, Abdelgader I, Abboud M, Bourauel C (2005) 3D reconstruction of dental specimens from 2D histological images and microCT‐scans. Comput Methods Biomech Biomed Eng 8:167–176. [DOI] [PubMed] [Google Scholar]
- 38. Rakic P (2002) Adult neurogenesis in mammals: an identity crisis. J Neurosci 22:614–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Rakic P (2002) Neurogenesis in adult primate neocortex: an evaluation of the evidence. Nat Rev Neurosci 3:65–71. [DOI] [PubMed] [Google Scholar]
- 40. Scharfman H, Goodman J, Macleod A, Phani S, Antonelli C, Croll S (2005) Increased neurogenesis and the ectopic granule cells after intrahippocampal BDNF infusion in adult rats. Exp Neurol 192:348–356. [DOI] [PubMed] [Google Scholar]
- 41. Scharfman HE (2002) Does the development of a GABAergic phenotype by hippocampal dentate gyrus granule cells contribute to epileptogenesis. Epilepsy Curr 2:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Schmidt W, Reymann KG (2002) Proliferating cells differentiate into neurons in the hippocampal CA1 region of gerbils after global cerebral ischemia. Neurosci Lett 334:153–156. [DOI] [PubMed] [Google Scholar]
- 43. Schmoll H, Badan I, Grecksch G, Walker L, Kessler C, Popa‐Wagner A (2003) Kindling status in sprague‐dawley rats induced by pentylenetetrazole: involvement of a critical development period. Am J Pathol 162:1027–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Sharma SS, Dhar A, Kaundal RK (2007) FeTPPS protects against global cerebral ischemic‐reperfusion injury in gerbils. Pharmacol Res 55:335–342. [DOI] [PubMed] [Google Scholar]
- 45. Shetty AK, Turner DA (2001) Glutamic acid decarboxylase‐67‐positive hippocampal interneurons undergo a permanent reduction in number following kainic acid‐induced degeneration of ca3 pyramidal neurons. Exp Neurol 169:276–297. [DOI] [PubMed] [Google Scholar]
- 46. Stanley DP, Shetty AK (2004) Aging in the rat hippocampus is associated with widespread reductions in the number of glutamate decarboxylase‐67 positive interneurons but not interneuron degeneration. J Neurochem 89:204–216. [DOI] [PubMed] [Google Scholar]
- 47. Takagi Y, Nozaki K, Takahashi J, Yodoi J, Ishikawa M, Hashimoto N (1999) Proliferation of neuronal precursor cells in the dentate gyrus is accelerated after transient forebrain ischemia in mice. Brain Res 831:283–287. [DOI] [PubMed] [Google Scholar]
- 48. Taupin P (2006) Neurogenesis in the adult central nervous system. C R Biol 329:465–475. [DOI] [PubMed] [Google Scholar]
- 49. Teramoto T, Qiu J, Plumier JC, Moskowitz MA (2003) EGF amplifies the replacement of parvalbumin‐expressing striatal interneurons after ischemia. J Clin Investig 111:1125–1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Tonchev AB, Yamashima T, Zhao L, Okano HJ, Okano H (2003) Proliferation of neural and neuronal progenitors after global brain ischemia in young adult macaque monkeys. Mol Cell Neurosci 23:292–301. [DOI] [PubMed] [Google Scholar]
- 51. von Euler M, Bendel O, Bueters T, Sandin J, von Euler G (2006) Profound but transient deficits in learning and memory after global ischemia using a novel water maze test. Behav Brain Res 166:204–210. [DOI] [PubMed] [Google Scholar]
- 52. Yagita Y, Kitagawa K, Ohtsuki T, Takasawa K, Miyata T, Okano H et al (2001) Neurogenesis by progenitor cells in the ischemic adult rat hippocampus. Stroke 32:1890–1896. [DOI] [PubMed] [Google Scholar]
