Icariin promotes cell proliferation and regulates gene expression in human neural stem cells in vitro

  • Authors:
    • Pan Yang
    • Yun‑Qian Guan
    • Ya‑Li Li
    • Li Zhang
    • Lan Zhang
    • Lin Li
  • View Affiliations

  • Published online on: June 8, 2016     https://doi.org/10.3892/mmr.2016.5377
  • Pages: 1316-1322
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Icariin (ICA), which is an essential bioactive component extracted from the herb Epimedium, possesses neuroprotective properties. The aim of the present study was to investigate the regulatory roles of ICA in cell proliferation and gene expression in human neural stem cells (NSCs) in vitro. Single cells were isolated from the corpus striatum of 16‑20‑week human fetuses obtained following spontaneous abortion. The cells were cultured in Dulbecco's modified Eagle's medium/F12 complete medium and were characterized by immunostaining and cell differentiation assay. NSCs were treated with ICA, and cell proliferation was assessed using the Cell Counting kit‑8 cell proliferation assay kit. In addition, neurosphere formation was comparatively studied between the ICA‑treated and control cells. cDNA microarray analysis was performed to examine the effects of ICA on gene expression. Altered expression of genes important for regulating NSC proliferation was further analyzed by quantitative polymerase chain reaction (qPCR). The results demonstrated that typical neurospheres appeared after 7‑10 days of culturing of individual cells isolated from the corpus striatum. These cells expressed nestin, an important NSC marker, and in the presence of differentiation medium they expressed β‑III‑tubulin, a specific neuronal marker, and glial fibrillary acidic protein, an astrocyte marker. Treatment with ICA enhanced NSC proliferation and the formation of neurospheres. Microarray data and pathway analysis revealed that the genes regulated by ICA were involved in several signaling pathways, including the Wnt and basic fibroblast growth factor (bFGF) pathways, which are important for the regulation of NSC function. Upregulation of frizzled class receptor 7 and dishevelled segment polarity protein 3, which are key players in the Wnt pathway, and fibroblast growth factor receptor 1, which is the receptor for bFGF, and downregulation of glycogen synthase kinase‑3β, which is a Wnt pathway inhibitor, was further validated by qPCR. In conclusion, ICA promoted proliferation and regulated gene expression in human NSCs, thus suggesting that ICA may exert its neuroprotective effects by regulating NSC activity.

Introduction

Neural stem cells (NSCs) are multipotent stem cells, which are derived from neural tissues, either from the central or peripheral nervous systems. NSCs are self-renewing and can give rise to various cell types of the nervous system, including neurons, astrocytes and oligodendrocytes, through asymmetric cell division (1). NSC-derived neurogenesis is critical for various aspects of central nervous function, including spatial learning and memory, mood regulation and motor control (24). Previous studies have suggested that NSC transplantation may have beneficial effects in the treatment of numerous pathological conditions, such as neurodegenerative diseases, seizures and brain tumors (57). Therefore, NSCs may be considered a promising therapeutic option for the treatment of ischemic stroke and neurodegenerative diseases (8). Nevertheless, how to promote cell survival and proliferation following cell transplantation remains a significant challenge for stem cell therapy using NSCs to treat various neurodegenerative disorders and strokes.

Epimedium is a plant of the Epimedium brevicornum Maxim species of the Berberidaceae family, which contains various bioactive compounds, including flavonoids and alkaloids (9). Icariin (ICA) is a type of flavonoid, and is the most metabolically active component extracted from Epimedium. ICA possesses various bioactivities, including antidepressant-like effects and anti-inflammatory activity (10,11). In addition, it has previously been reported that ICA improves memory impairment in a murine model of Alzheimer's disease, and ameliorates motor dysfunction in spinal cord injury in mice (12,13). Due to the known actions of ICA, and the important role NSCs have in neuroregeneration, the present study hypothesized that ICA may stimulate cell proliferation and regulate gene expression in NSCs.

Materials and methods

Materials

ICA (purity >98%, high-performance liquid chromatography grade) with the molecular formula C33H42O15 and a molecular weight of 676.65 was purchased from Shaanxi Scidoor Hi-tech Biology Co., Ltd. (Xi'an, China). Hank's Balanced Salt Solution (HBSS), Dulbecco's modified Eagle's medium (DMEM)/F12 medium, fetal bovine serum (FBS), B27, epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), Glutmax and poly-L-lysine-coated plates were purchased from Thermo Fisher Scientific, Inc. (Beijing, China). Mouse anti-nestin was obtained from BD Pharmingen (cat. no. 560341; San Jose, CA, USA), mouse anti-β-III-tubulin (cat. no. T8578), mouse anti-glial fibrillary acidic protein (GFAP; cat. no. G3893) and rabbit anti-galactocerebroside (GalC; cat. no. G9152) were obtained from Sigma Aldrich (Shanghai) Trading. Co., Ltd.(Shanghai, China). Cy2- or Cy3-conjugated secondary antibodies were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). RNeasy kit and AMV reverse transcriptase were obtained from Qiagen, Inc. (Valencia, CA, USA). Cy3-dCTP and Cy5-dCTP were purchased from CapitalBio, Inc. (Beijing, China). SYBR Premix Ex Taq for quantitative polymerase chain reaction (qPCR) was obtained from Sangon Biotech Co., Ltd. (Shanghai, China).

NSC isolation and characterization

The use of human fetuses obtained following spontaneous abortion was approved by the Ethic Committee of Xuanwu Hospital of Capital Medical University (Beijing, China), and written informed consent was obtained from the study participants. The corpus striatum of 16–20-week fetuses were obtained from 5 fetuses following spontaneous abortion, and were dissected and chopped in cooled HBSS using a tissue chopper. The tissue was further triturated by pipetting up and down 10 times to dissociate cells. After filtering through a 300 micron stainless steel sieve, in order to remove the large pieces of tissue, the dissociated cells were pelleted by centrifugation at 200 × g for 5 min at room temperature, washed once with HBSS, and were re-suspended at a density of 2×106 cells/ml in DMEM/F12 medium containing 2% B27, 20 ng/ml EGF, 20 ng/ml bFGF and 1% Glutmax (DMEM/F12 complete medium). Cells were seeded in 100 mm dishes and cultured at 37°C in a humidified atmosphere containing 5% CO2. Cell medium was changed every 3 days. After an initial culture for 7–10 days typical NSC clusters appeared, which were pelleted by centrifugation at 200 × g for 5 min at room temperature and were treated with accutase. Accutase-digested cells were washed once with HBSS, re-suspended in DMEM/F12 complete medium and used for all subsequent assays. For NSC characterization, the cells were seeded onto poly-L-lysine-coated chamber slides (1×104/cm2). Following a 12 h culture, the cells were subjected to immunostaining for the analysis of nestin expression.

NSC differentiation

Growth factor-free DMEM/F12 medium supplemented with 1% FBS was used to induce cell differentiation. Cell clusters were digested with accutase as aforementioned, were washed once with HBSS, re-suspended in differentiation medium, and were seeded onto poly-L-lysine-coated chamber slides (1×104/cm2). Cells were cultured for 7 days, and were subsequently subjected to fixation for immunostaining, in order to determine the expression of β-III-tubulin, GFAP and GalC.

Immunostaining

Cells were fixed in ice-cold 4% paraformaldehyde for 15 min, followed by three washes with phosphate-buffered saline (PBS; pH 7.4). After blocking with 10% goat serum (Jackson ImmunoResearch, Inc., West Grove, PA, USA) in PBS containing 0.3% Triton X-100 at 37°C for 1 h, the cells were incubated with various primary antibodies at 4°C overnight. The antibodies used were as follows: Mouse anti-nestin (1:1,000), mouse anti-β-III-tubulin (1:1,000), mouse anti-GFAP (1:500) and rabbit anti-GalC (1:100). After three washes with PBS, Cy2- or Cy3-conjugated secondary antibodies (1:500) were added to the cells and were incubated at 37°C for 30 min, followed by three washes with PBS. Cells were then mounted with VectaShield mounting medium (Vector Laboratories, Inc., Burlingame, CA, USA) and 4′,6-diamidino-2-phenylindole was used for nuclear counter-staining. Fluorescent signals were visualized using a confocal laser microscope system (MRC1024; Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Cell treatment and proliferation assay

To study the effects of ICA on NSC proliferation, accutase-dissociated single cells were suspended in DMEM/F12 complete medium at a density of 2×105 cells/ml. A 100 µl cell suspension was added into each well of a 96-well plate, which had previously been coated with poly-L-lysine. Following a 24 h culture, the medium was replaced with serum- and growth factor-free medium containing various concentrations of ICA (final concentrations, 0.1, 1 and 10 µM). After a further 24 h of culture, the medium was removed and serum- and growth factor-free medium (without phenol red) containing 10% Cell Counting kit (CCK)-8 reagent was added (Dojundo Molecular Technologies, Inc., Kumamoto, Japan). The cells were cultured for 4 h at 37°C, and the optical density (OD) of each well was measured at 450 nm (OD450) using a microplate reader. Cells cultured in serum- and growth factor-free medium served as a control. In a separate experiment, individual NSCs in DMEM/F12 complete medium were seeded into each well of a 24-well plate (104 cells/cm2) coated with poly-L-lysine. Following a 24 h culture, the medium was replaced with serum- and growth factor-free medium containing 10 µM ICA (final concentration). Cells were maintained in culture for 7 days, and the formation of neurospheres was observed by microscopy. The diameters of 15 randomly chosen neurospheres from each well were measured using a micrometer installed in the microscope (IX70; Olympus Corporation, Tokyo, Japan). The results from three wells of each treatment group were averaged and compared.

cDNA microarray analysis

cDNA microarray analysis was performed for gene expression profiling, and the results were compared between the cells treated with ICA and the untreated control cells. Cell treatment with 10 µM ICA was conducted, as aforementioned. NSCs cultured in serum- and growth factor-free medium without ICA served as a control. RNA was extracted from the cells using the RNeasy kit, according to manufacturer's protocol, and was quantified using the spectrophotometric method. Total RNA (2 µg) was used for first-strand cDNA synthesis using AMV reverse transcriptase, according to the manufacturer's protocol. Cy3-dCTP was added to the cDNA of the control cells, whereas Cy5-dCTP was added to the cDNA of ICA-treated cells. Microarray analysis was performed using the 22 K Human Genome Array (CapitalBio, Inc.). The slide contained gene-specific 70-mer oligonucleotides representing 21,329 human genes, including four human housekeeping genes as positive controls, and 12 negative controls, which were designed to have no significant homology with known human gene sequences. The labeled samples were quantitatively adjusted based on the efficiency of Cy-dye incorporation and mixed into 80 µl hybridization solution [3X saline-sodium citrate (SSC), 0.2% sodium dodecyl sulfate (SDS), 25% formamide and 5X Denhardt's). cDNA in hybridization solution was denatured at 95°C for 3 min prior to loading onto the microarray. The array was hybridized at 42°C overnight, and washed once with washing solution (0.2% SDS and 2X SSC) at 42°C for 5 min, followed by one wash with 0.2% SSC at room temperature for 5 min. Finally, the array was scanned using a confocal LuxScan 10 KA scanner (CapitalBio, Inc.). The data of the obtained images were extracted using LuxScan 3.0 software (CapitalBio, Inc.). Genes with a signal intensity >800 (Cy3 or Cy5) were considered to be expressed. In every two channel slides, the intensity ratio of Cy5 to Cy3 of each spot was calculated following normalization with LOWESS regression. Statistical data and differential analysis files were generated using SAM software 3.0 (Stanford University, Stanford, CA, USA). Significantly altered genes were selected based on a P<0.05 and >2 fold change. All differentially expressed genes were analyzed using the free web-based Molecular Annotation System 2.0 (MAS 2.0; http://bioinfo.capitalbio.com/mas3/).

qPCR

qPCR was performed to verify the array results. Primers (Table I) were designed based on published gene sequences using the Oligo 5.0 program (Molecular Biology Insights, Inc., Colorado Springs, CO, USA), and were synthesized by Sangon Biotech Co., Ltd. The PCR mix (20 µl) contained 10 µl 2X SYBR Taq Premix buffer, 1 µl cDNA, 200 nM of each primer (final concentration) and 9.0 µl nuclease-free water. The reaction mixture was incubated at 95°C for 10 sec, followed by 40 amplification cycles at 95°C for 5 sec, 60°C for 10 sec and 72°C for 10 sec. A melting curve analysis was run to determine the specificity of amplification following each qPCR analysis. Relative mRNA expression levels were calculated using the 2−∆∆Cq method (14), following normalization against glyceraldehyde 3-phosphate dehyrdrogenase levels.

Table I

Quantitative polymerase chain reaction primer sequences.

Table I

Quantitative polymerase chain reaction primer sequences.

GenesReference numberSequenceProduct size
FZD7NM_003507F: 5′-GGCGCCTCTGTTCGTCTAC-3′106 bp
R: 5′-GGTCTTGGTGCCGTCGTGT-3′
CTNNB1NM_001904F: 5′-CACGTGCAATCCCTGAACTGA-3′121 bp
R: 5′-CGCATGATAGCGTGTCTGGAA-3′
DVL3NM_004423F: 5′-GGTACTGCGGGAGATTGTG-3′168 bp
R: 5′-GGAAGGTGCCGGTCAT-3′
GSK-3βNM_002093F: 5′-GTCCGATTGCGTTATT-3′176 bp
R: 5′-TTCGGAACAGCTGATACAT-3′
FGFR1NM_023105F: 5′-AAGGCATCATTTGGTGAACAGAAC-3′133 bp
R: 5′-TCACACAACATTGCTTCAAGGTAGG-3′
GAPDHNM_002046F: 5′-ATGACATCAAGAAGGTGGTG-3′177 bp
R: 5′-CATACCAGGAAATGAGCTTG-3′

[i] F, forward primer; R, reverse primer; bp, base pair; FZD7, frizzled class receptor 7; CTNNB1, catenin beta-1; DVL3, dishevelled segment polarity protein 3; GSK-3β, glycogen synthase kinase-3β; FGRF1, fibroblast growth factor receptor 1; GAPDH, glyceraldehyde 3-phosphate dehyrdrogenase.

Statistical analysis

All results are expressed as the mean ± standard error of the mean, and were analyzed using one-way analysis of variance followed by Tukey's post-hoc test using SPSS 16.0 software (SPSS Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

NSC isolation and characterization

NSCs were isolated from the corpus striatum of 16–20-week human fetuses obtained following spontaneous abortion. After culturing in DMEM/F12 complete medium for 7–10 days, typical NSC clusters appeared in the culture. Immunostaining analysis revealed that neurosphere-forming cells expressed nestin, a marker of NSCs (Fig. 1A). Cell differentiation assay demonstrated that NSCs were able to differentiate into different cell types of the nervous system, as indicated by the expression of β-III-tubulin, a specific neuronal marker (Fig. 1B); and GFAP, an astrocyte marker (Fig. 1C). Few cells expressed GalC, an oligodendrocyte marker (data not shown).

NSC proliferation is stimulated by icariin in vitro

Using a CCK-8 cell proliferation assay kit, the effects of ICA on NSC proliferation were determined. As shown in Fig. 2, NSCs were incubated with various concentrations of ICA. In the presence of 0.1 or 1 µM ICA, cells did not exhibit markedly increased proliferation compared with the control cells (Fig. 2A). Conversely, cells incubated with 10 µM ICA exhibited a significantly increased proliferation rate compared with the untreated cells (n=4, P<0.05; Fig. 2A). When ICA concentration increased to 50 µM, cell toxicity was observed (data not shown). In a separate assay, the formation of neurospheres under the influence of ICA was examined. Neurospheres formed in ICA-treated cells appeared larger compared with those in the control group (Fig. 2B and C). By measuring neurosphere diameter, it was demonstrated that the diameter of neurospheres formed in the ICA-treated group was significantly greater than that of the neurospheres in the control group (n=5, P<0.05; Fig. 2D).

Gene expression profiling by cDNA microarray

Gene expression in 10 µM ICA-treated cells and control cells was profiled by cDNA microarray, and the results were compared. A total of 779 genes were differentially expressed between the ICA-treated group and the control group, based on P<0.05 and >2 fold change criteria (data not shown). Of the altered genes, those involved in the top 10 cell signaling pathways analyzed using MAS software were listed in Table II. Since the Wnt signaling pathway and the bFGF pathway have important roles in NSC proliferation and neurogenesis, several differentially expressed genes important in the Wnt signaling pathway were selected (Table III). Fibroblast growth factor receptor 1 was also further analyzed by qPCR to validate the array results.

Table II

Differences in gene expression in the top 10 pathways analyzed using Molecular Annotation System software.

Table II

Differences in gene expression in the top 10 pathways analyzed using Molecular Annotation System software.

PathwayTotal genesP-valueGenes
Regulation of actin cytoskeleton192.0E-8ITGA7, PDGFRA, ACTN1, PFN1, GSN, PDGFRB↓, GNG12, FGFR2, FGD3↓, PAK3↓, APC2↓, FGFR1, PFN2↓, ARHGEF7↓, ITGA2, ITGB5, TIAM1↓, PIP5K1B↓, FGFR3↓
Focal adhesion222.3E-7CAST, ITGA7, PDGFRA, JUN, ACTN1, LAMC1, PDGFRB↓, COL1A2, TNC, CCND1, SHC3, THBS4, THBS2↓, MAPK10↓, PAK3↓, SPP1, ITGA2, COL4A1, ITGB5, PDGFD↓, LAMA4, SRC
MAPK pathway205.2E-7PRKX↓, MEF2C↓, PDGFRA, JUN, CACNB3↓, DUSP4, NTRK2↓, DUSP6, PDGFRB↓, MYC, GNG12, FGFR2, MAPK10↓, CACNG4↓, MKNK1↓, GADD45A, FGFR1, TNFRSF1A, HSPA5, FGFR3↓
ECM-receptor interaction128.5E-7ITGA7, SDC3, LAMC1, COL1A2, TNC, THBS4, THBS 2↓, SPP1, ITGA2, COL4A1, ITGB5, LAMA4
Axon guidance122.0E-6EPHA3↓, NCK2↓, EPHA2, PAK3↓, SEMA6D↓, ROBO2↓, RGS3, DPYSL2↓, SEMA5B↓, PLXNA2↓, EPHB3↓, EPHB4↓
Cell communication117.0E-6INA↓, LAMC1, GJA1↓, COL1A2, TNC, THBS4, THBS2↓, VIM, SPP1, COL4A1, LAMA4
TGF-beta pathway91.2E-5ACVR1↓, RPS6KB2↓, MYC, THBS4, THBS2↓, AMH↓, INHBB↓, LTBP1, BMP7
Wnt pathway103.8E-5WNT7B, FZD7, DVL3, GSK- 3β↓, MYC, CCND1, CTNNB1, TCF7, LEF1, JUN
Gap junction95.2E-5TUBB3↓, PRKX↓, PDGFRA, PDGFRB↓, EDG2, GJA1↓, SRC↓, PDGFD↓, CSNK1E↓
Glutathione metabolism65.9E-5GSS, GSTA4↓, GSTO1, MGST2, GSTZ1, GPX7

[i] ↑Indicates gene upregulation and ↓indicates gene downregulation by icariin. MAPK, mitogen-activated protein kinase; ECM, extracellular matrix; TGF, transforming growth factor.

Table III

Differentially expressed genes in the Wnt signaling pathway.

Table III

Differentially expressed genes in the Wnt signaling pathway.

GeneFold change (ICA/control)Molecular functions
WNT7B   2.482Wnt ligand
FZD7   2.850Non-G-protein coupled receptor
DVL3   2.191Catalytic activity
GSK-3β   0.468Kinase and inactivating agent
MYC   4.390Transcription regulator
CCND111.677Mitotic and cell cycle regulation
CTNNB1   2.500Coactivator of transcription
TCF7   2.804RNA polymerase II transcription
LEF110.663Transcription regulator
JUN   2.086Transcription cofactor

[i] ICA, icariin.

Gene expression validation by qPCR

The mRNA expression levels of fibroblast growth factor receptor 1 (FGFR1), and four genes of the Wnt signaling pathway: Frizzled class receptor 7 (FZD7), dishevelled segment polarity protein 3 (DVL3), catenin beta-1 (CTNNB1) and glycogen synthase kinase-3β (GSK-3β), were further determined by qPCR. The results indicated that the mRNA expression levels of FZD7 (Fig. 3A) and DVL3 (Fig. 3B) were significantly elevated in the ICA-treated group compared with the control group (P<0.01 and P<0.05, respectively); CTNNB1 expression was increased in ICA-treated cells; however, the increase was not significantly different compared with the control cells, as determined by statistical analysis (Fig. 3C). The mRNA expression levels of GSK-3β were markedly reduced in the ICA-treated cells (n=6, P<0.01; Fig. 3D). In addition, FGFR1 expression was significantly upregulated by ICA (P<0.01; Fig. 3E).

Discussion

NSCs possess the characteristics of self-renewal and multipotency (15). The present study isolated and cultured NSCs from the corpus striatum of 16–20-week fetuses obtained following spontaneous abortion, and demonstrated that these cells expressed nestin, which is an intermediate filament protein selectively expressed in NSCs and commonly used as a marker for NSCs (15). Cultured in differentiation medium, the NSCs expressed β-III-tubulin, a specific neuronal marker, and GFAP, a marker for astrocytes (16,17), thus indicating that the NSCs were multipotent.

In our previous study, Epimedium flavonoids were revealed to promote the proliferation and differentiation of NSCs derived from neonatal rats in vitro (9). ICA, which is an important flavonoid extracted from Epimedium, has not yet been investigated regarding its role in regulating cell proliferation and gene expression in NSCs. The present study examined the effects of ICA on NSC proliferation. The results demonstrated that NSCs treated with 10 µM ICA exhibited a significantly higher proliferation rate compared with untreated cells. When NSCs were incubated with 10 µM ICA for 7 days, larger neurospheres were formed compared with those in the control group. Measurement of neurosphere diameter revealed the diameter of neurospheres formed in the ICA-treated group was significantly greater compared with in the control group. These data suggested that ICA promoted NSC proliferation and growth.

In order to provide information regarding the mechanisms underlying ICA actions, the present study analyzed ICA-regulated gene expression in NSCs. A cDNA microarray analysis, which is an important tool used to uncover the molecular basis of several biological processes (18), was used to profile gene expression in NSCs. The results indicated that treatment with ICA regulated gene expression in NSCs, affecting numerous signaling pathways (Tables II and III). Of these pathways, the Wnt signaling pathway has been recognized as a key regulator for self-renewal and maintenance of neural progenitors during neurogenesis (1921), and bFGF signaling has been reported to potently induce NSC proliferation (2225). The canonical Wnt pathway cascade is initiated when Wnt signaling proteins bind to Frizzled protein receptors, such as FZD7, which is a family of G protein-coupled receptors (2628). Subsequently, the receptor interacts with a downstream mediator, the Dishevelled protein, which is an essential effector for the accumulation of β-catenin encoded by CTNNB130 (27,29,30). The accumulated β-catenin then translocates to the nucleus where it regulates target gene expression to control cell function, including cell proliferation. Conversely, GSK-3β negatively regulates Wnt signaling (27,31). The microarray analysis conducted in the present study demonstrated that the expression levels of several genes in the Wnt and bFGF pathways were altered following ICA treatment. The expression levels of FGFR1, and four important Wnt family members: FZD7, DVL3, CTNNB1 and GSK-3β were further determined by qPCR. The results demonstrated that ICA significantly enhanced expression of FGFR1, FZD7 and DVL3, and significantly reduced the expression of GSK-3β. It was hypothesized that ICA-regulated expression of these genes presumably favored the proliferation of human NSCs.

In conclusion, NSCs derived from the corpus striatum of 16–20-week human fetuses obtained following spontaneous abortion were successfully isolated. The NSCs expressed nestin, an NSC marker, and could differentiate into various types of cells of the nervous system. Treatment with ICA promoted the proliferation and modulated gene expression in NSCs, thus suggesting that ICA may exert its neuroprotective effects by regulating NSC activity. Taken together, the findings suggest that ICA may be potentially applied for the treatment of neurodegenerative disorders.

Acknowledgments

The present study was supported by the National Natural Science Foundation of China (grant nos. 81273498 and 81274120), the Beijing Natural Science Foundation (grant nos. 7112061 and 7132110), the Beijing Science and Technology Program (grant no. Z131102002813066), the Capital Health Development Scientific Grant (grant no. 2011-1001-05), and the Beijing Health and Technical High-level Personnel Plan (grant nos. 2011-1-7 and 2009-3-66).

References

1 

Gage FH: Mammalian neural stem cells. Science. 287:1433–1438. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Zhang CL, Zou Y, He W, Gage FH and Evans RM: A role for adult TLX-positive neural stem cells in learning and behaviour. Nature. 451:1004–1007. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Thomas RM and Peterson DA: Even neural stem cells get the blues: Evidence for a molecular link between modulation of adult neurogenesis and depression. Gene Expr. 14:183–193. 2008.PubMed/NCBI

4 

McBride JL, Behrstock SP, Chen EY, Jakel RJ, Siegel I, Svendsen CN and Kordower JH: Human neural stem cell transplants improve motor function in a rat model of Huntington's disease. J Comp Neurol. 475:211–219. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Carletti B, Piemonte F and Rossi F: Neuroprotection: The emerging concept of restorative neural stem cell biology for the treatment of neurodegenerative diseases. Curr Neuropharmacol. 9:313–317. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Waldau B, Hattiangady B, Kuruba R and Shetty AK: Medial ganglionic eminence-derived neural stem cell grafts ease spontaneous seizures and restore GDNF expression in a rat model of chronic temporal lobe epilepsy. Stem Cells. 28:1153–1164. 2010.PubMed/NCBI

7 

Achanta P, Sedora Roman NI and Quiñones-Hinojosa A: Gliomagenesis and the use of neural stem cells in brain tumor treatment. Anticancer Agents Med Chem. 10:121–130. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Hung CW, Liou YJ, Lu SW, Tseng LM, Kao CL, Chen SJ, Chiou SH and Chang CJ: Stem cell-based neuroprotective and neurorestorative strategies. Int J Mol Sci. 11:2039–2055. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Yao R, Zhang L, Li X and Li L: Effects of Epimedium flavonoids on proliferation and differentiation of neural stem cells in vitro. Neurol Res. 32:736–742. 2010. View Article : Google Scholar

10 

Pan Y, Kong L, Xia X, Zhang W, Xia Z and Jiang F: Antidepressant-like effect of icariin and its possible mechanism in mice. Pharmacol Biochem Behav. 82:686–694. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Xu CQ, Liu BJ, Wu JF, Xu YC, Duan XH, Cao YX and Dong JC: Icariin attenuates LPS-induced acute inflammatory responses: Involvement of PI3K/Akt and NF-kappaB signaling pathway. Eur J Pharmacol. 642:146–153. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Urano T and Tohda C: Icariin improves memory impairment in Alzheimer's disease model mice (5xFAD) and attenuates amyloid β-induced neurite atrophy. Phytother Res. 24:1658–1663. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Tohda C and Nagata A: Epimedium koreanum extract and its constituent icariin improve motor dysfunction in spinal cord injury. Evid Based Complement Alternat Med. 2012:7312082012. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

Messam CA, Hou J and Major EO: Coexpression of nestin in neural and glial cells in the developing human CNS defined by a human-specific anti-nestin antibody. Exp Neurol. 161:585–596. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Katsetos CD, Herman MM and Mörk SJ: Class III beta-tubulin in human development and cancer. Cell Motil Cytoskeleton. 55:77–96. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Menet V, Giménez y Ribotta M, Chauvet N, Drian MJ, Lannoy J, Colucci-Guyon E and Privat A: Inactivation of the glial fibrillary acidic protein gene, but not that of vimentin, improves neuronal survival and neurite growth by modifying adhesion molecule expression. J Neurosci. 21:6147–6158. 2001.PubMed/NCBI

18 

Schena M, Shalon D, Davis RW and Brown PO: Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science. 270:467–470. 1995. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Wexler EM, Paucer A, Kornblum HI, Palmer TD and Geschwind DH: Endogenous Wnt signaling maintains neural progenitor cell potency. Stem Cells. 27:1130–1141. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Machon O, Backman M, Machonova O, Kozmik Z, Vacik T, Andersen L and Krauss S: A dynamic gradient of Wnt signaling controls initiation of neurogenesis in the mammalian cortex and cellular specification in the hippocampus. Dev Biol. 311:223–237. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Reimers D, López-Toledano MA, Mason I, Cuevas P, Redondo C, Herranz AS, Lobo MV and Bazán E: Developmental expression of fibroblast growth factor (FGF) receptors in neural stem cell progeny. Modulation of neuronal and glial lineages by basic FGF treatment. Neurol Res. 23:612–621. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Kelly CM, Tyers P, Borg MT, Svendsen CN, Dunnett SB and Rosser AE: EGF and FGF-2 responsiveness of rat and mouse neural precursors derived from the embryonic CNS. Brain Res Bull. 68:83–94. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Nelson AD and Svendsen CN: Low concentrations of extracellular FGF-2 are sufficient but not essential for neurogenesis from human neural progenitor cells. Mol Cell Neurosci. 33:29–35. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Furusho M, Kaga Y, Ishii A, Hébert JM and Bansal R: Fibroblast growth factor signaling is required for the generation of oligoden-drocyte progenitors from the embryonic forebrain. J Neurosci. 31:5055–5066. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Clevers H and Nusse R: Wnt/β-catenin signaling and disease. Cell. 149:1192–1205. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Saito-Diaz K, Chen TW, Wang X, Thorne CA, Wallace HA, Page-McCaw A and Lee E: The way Wnt works: Components and mechanism. Growth Factors. 31:1–31. 2013. View Article : Google Scholar :

28 

Dann CE, Hsieh JC, Rattner A, Sharma D, Nathans J and Leahy DJ: Insights into Wnt binding and signaling from the structures of two Frizzled cysteine-rich domains. Nature. 412:86–90. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Wallingford JB and Habas R: The developmental biology of dishevelled: An enigmatic protein governing cell fate and cell polarity. Development. 132:4421–4436. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Gan XQ, Wang JY, Xi Y, Wu ZL, Li YP and Li L: Nuclear Dvl, c-Jun, beta-catenin, and TCF form a complex leading to stabilization of beta-catenin-TCF interaction. J Cell Biol. 180:1087–1100. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Hart MJ, de los Santos R, Albert IN, Rubinfeld B and Polakis P: Downregulation of beta-catenin by human Axin and its association with the APC tumor suppressor, beta-catenin and GSK3 beta. Curr Biol. 8:573–581. 1998. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2016
Volume 14 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang P, Guan YQ, Li YL, Zhang L, Zhang L and Li L: Icariin promotes cell proliferation and regulates gene expression in human neural stem cells in vitro. Mol Med Rep 14: 1316-1322, 2016
APA
Yang, P., Guan, Y., Li, Y., Zhang, L., Zhang, L., & Li, L. (2016). Icariin promotes cell proliferation and regulates gene expression in human neural stem cells in vitro. Molecular Medicine Reports, 14, 1316-1322. https://doi.org/10.3892/mmr.2016.5377
MLA
Yang, P., Guan, Y., Li, Y., Zhang, L., Zhang, L., Li, L."Icariin promotes cell proliferation and regulates gene expression in human neural stem cells in vitro". Molecular Medicine Reports 14.2 (2016): 1316-1322.
Chicago
Yang, P., Guan, Y., Li, Y., Zhang, L., Zhang, L., Li, L."Icariin promotes cell proliferation and regulates gene expression in human neural stem cells in vitro". Molecular Medicine Reports 14, no. 2 (2016): 1316-1322. https://doi.org/10.3892/mmr.2016.5377