Open Access

Pro‑neurogenic effects of andrographolide on RSC96 Schwann cells in vitro

  • Authors:
    • Fuben Xu
    • Huayu Wu
    • Kun Zhang
    • Peizhen Lv
    • Li Zheng
    • Jinmin Zhao
  • View Affiliations

  • Published online on: September 6, 2016     https://doi.org/10.3892/mmr.2016.5717
  • Pages: 3573-3580
  • Copyright: © Xu 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

Nerve regeneration remains a challenge to the treatment of peripheral nerve injury. Andrographolide (Andro) is the main active constituent of Andrographis paniculata, which has been applied in the treatment of several diseases, including inflammation, in ancient China. Andro has been reported to facilitate the reduction of edema and to exert analgesic effects in the treatment of various diseases. These findings suggest that Andro may be considered a promising anti‑inflammatory agent that may suppress destruction and accelerate proliferation of Schwann cells following peripheral nerve injury. In the present study, the effects of Andro on RSC96 cells were investigated in vitro. The RSC96 cell line is a spontaneously immortalized rat Schwann cell line, which was originally derived from a long‑term culture of rat primary Schwann cells. RSC96 cells were treated with a range of 0 to 50 µM Andro prior to the MTT assay. Cell proliferation, morphology, synthesis and nerve‑specific gene expression were performed to detect the effect of Andro on RSC96 cells. The results of the present study demonstrated that the recommended doses of Andro ranged between 0.78 and 12.5 µM, among which the most obvious response was observed when used at 3.125 µM (P<0.05). DNA content was improved in Andro groups compared with the control group (P<0.05). In addition, Andro was able to promote the gene expression of glial cell line‑derived neurotrophic factor, brain‑derived neurotrophic factor, ciliary neurotrophic factor, and the specific Schwann cell marker S100β (P<0.05). The results of a viability assay, hematoxylin‑eosin staining, and immunohistochemistry were also improved in Andro groups. These results indicated that Andro may accelerate proliferation of RSC96 cells in vitro, whilst maintaining the Schwann cell phenotype; therefore, the present study may provide valuable evidence for the further exploration of the effects of Andro on peripheral nerves.

Introduction

Peripheral nerve injuries are considered one of the most challenging and difficult problems to treat with reconstructive surgery (1). Fractures, hematomas, contusions and compressions may induce peripheral nerve injury, which is characterized by the disruption of myelin sheaths and axons (2,3). Inflammation has been shown to have an important role in the pathogenesis of several neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease, multiple sclerosis and amyotrophic lateral sclerosis (46). The ability of the mammalian peripheral nervous system (PNS) to regenerate axons following injury is well documented (7). Schwann cells have an important role in axon regeneration post-injury (8,9). Therefore, the identification of an effective anti-neuroinflammatory and neuroprotective agent, which is able to accelerate the proliferation of Schwann cells, thus maintaining the Schwann cell phenotype, is of great importance.

In traditional Chinese medicine, Andrographis paniculata is a traditional herb, which possesses immunological, antibacterial, antiviral, anti-inflammatory, antithrombotic, and lung and hepato-protective properties (1013). Andrographolide (Andro; Fig. 1) is the primary active component of A. paniculata, which is widely used in South Asia and China for the treatment of inflammation-related diseases, due to its potent anti-inflammatory and antiviral properties (1416). Andro and its derivatives, a group of diterpenes, have been reported to exert a protective effect against lipopolysaccharide-induced dopaminergic neurodegeneration in mesencephalic neuron-glia cultures (17). The anti-inflammatory role of Andro has been well-documented in several studies (18,19). Furthermore, Andro exerts proapoptotic effects on tumor cells (20,21). It has also been reported that Andro facilitates cell differentiation (22). These findings suggested that Andro may exert anti-neuroinflammatory and neuroprotective effects during peripheral nerve regeneration, which is a vital long-term strategy in the treatment of peripheral nerve injury.

As myelin-forming cells in the PNS, Schwann cells have a crucial role in peripheral nerve regeneration (23). Schwann cells have been shown to be able to provide bioactive substrates for axonal migration, and release molecules that regulate axonal outgrowth (24). In addition, Schwann cells activate nonresident macrophages to the site of injury, in order to complete myelin phagocytosis, release cytokines, and secrete neurotrophic factors that guide the resultant regeneration (25,26). Schwann cells provide trophic support to axons via the expression of several neurotrophic factors, including brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF) and ciliary neurotrophic factor (CNTF), particularly following nerve injury (27). These findings indicate the specific nature of the relationship between Schwann cells and axons, and thus confirm our hypothesis.

Based on the hypothesis that Andro may be used as a potential anti-inflammatory agent to relieve the destruction and accelerate the proliferation of Schwann cells following peripheral nerve injury, the present study investigated its effects on the growth and phenotypic maintenance of RSC96 cells in vitro. Examination of cell proliferation, morphology, viability, and RSC96-specific gene expression was performed. The results suggested that Andro may exert effects on RSC96 cell attachment, survival and proliferation, and on the release of neurotrophic factors. The present study may provide evidence for the application of Andro in the clinical treatment of peripheral nerve injury.

Materials and methods

Reagents and instruments

Trypsin and antibiotics (100 U/ml penicillin and 100 U/ml streptomycin) were purchased from Beijing Solarbio Science & Technology Co., Ltd. (Beijing, China); 6-well and 96-well cell culture plates were purchased from Costar (Corning Incorporated, Corning, NY, USA). Anti-S100β (S100β; cat. no. BA120; 1:200) antibody and the 3,3-diaminobenzidine tetrahydrochloride (DAB) kit were obtained from Wuhan Boster Biological Technology, Ltd. (Wuhan, China), Dulbecco's modified Eagle's medium/F-12 supplement (DMEM/F-12), fetal bovine serum (FBS) and 3–(4,5)-dimethylthiahiazol(-z-y1)-3,5-di-phenyltetrazo-lium-bromide (MTT) were purchased from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Dimethyl sulfoxide (DMSO), Hoechst 33258 and proteinase K were purchased from Sigma-Aldrich (Merck Millipore, Darmstadt, Germany). Multiskan GO Microplate Spectrophotometer was obtained from Thermo Fisher Scientific, Inc. Other reagents and instruments used in the present study were purchased from the following companies: Hematoxylin-eosin (HE) kit (Nanjing Jiancheng Bioengineering Institute, Nanjing, China); RNeasy RNA extraction kit (Tiangen Biotech Co., Ltd., Beijing, China); reverse transcription (RT) kit (Fermentas; Thermo Fisher Scientific, Inc.); Fast-Start Universal SYBR Green Master Mix (Roche Diagnostics GmbH, Mannheim, Germany); quantitative polymerase chain reaction (qPCR) detection system (RealPlex4; Eppendorf, Hauppauge, NY, USA); LIVE/DEAD viability assay kit (Invitrogen; Thermo Fisher Scientific, Inc.); laser scanning confocal microscope (Nikon Corporation, Tokyo, Japan); and upright microscope (Olympus Corporation, Tokyo, Japan).

Cells culture

The RSC96 cell line consists of spontaneously immortalized rat Schwann cells, which are derived from the long-term culture of rat primary Schwann cells. RSC96 cells were purchased from the China Center for Type Culture Collection (Wuhan, China), and were cultured in DMEM/F-12 supplemented with 10% (v/v) FBS and 1% (v/v) antibiotics in a humidified atmosphere containing 5% CO2 and 95% air at 37°C. The culture medium was replaced every 3 days after plating. RSC96 cells were passaged with 0.25% trypsin when cell confluence reached 80–90%. Confluent RSC96 cells were subsequently treated at the indicated times with the indicated concentrations of Andro.

Chemicals

Andro was purchased from Chengdu Must Bio-technology Co., Ltd. (Chengdu, China). Prior to experimentation, Andro was dissolved in DMSO in order to generate a 100 mM stock solution, and was stored at −4°C. The Andro stock solution was diluted with cultured medium to provided various concentrations and added to the cell culture for subsequent experiments. Prior to use, the culture medium contained 1.5625, 3.125 and 6.25 µM Andro was filtered using 0.22 µm filters for sterilization.

Cell cytotoxicity assay

Cell viability was estimated using a colorimetric assay based on the conversion of MTT into a blue formazan product. The cells were plated at 800 cells/well in 96-well cell culture plates and were pretreated with various concentrations of Andro (0–50 µM) for 3 days in a 5% CO2 humidified incubator at 37°C. MTT (5 mg/ml) was then added to each well and the plates were incubated in the dark at 37°C for 4 h. Subsequently, culture medium was removed and the cells were treated with 150 µl DMSO to dissolve the formazan product. The cells were incubated in DMSO with agitation for 10 min. Optical density of each sample was measured using a Multiskan GO Microplate Spectrophotometer at 570 nm. Five individual cultures were used for each test. The experiments were carried out in quintuplicate.

Cell proliferation analysis

Based on the results of the cytotoxicity assay, three doses of Andro, which exhibited a positive effect, were selected (1.5625, 3.125 and 6.25 µM), alongside a control group (0 µM Andro) for cell proliferation analysis. RSC96 cells in the various groups were cultured for 2, 4 and 6 days in a 5% CO2 humidified incubator at 37°C prior to subsequent experiments. Cells were digested with 0.25% trypsin and were resuspended in phosphate-buffered saline (PBS) containing 60 µg/ml proteinase K for 6 h at 60°C. After dyeing with Hoechst 33258, cell proliferation was determined by detecting DNA production using an ultraviolet spectrofluorometer; calf thymus DNA was used as a standard. The excitation wavelength was 346 nm and the emission wavelength was 460 nm. The experiments were carried out in quintuplicate.

Morphological examination

Cells were cultured for 2, 4 and 6 days, and were fixed in 4% paraformaldehyde for 40 min at room temperature for subsequent HE staining. Cells were incubated with a nuclear dye for 3 min, followed by a 10 sec incubation with HE. Subsequently, the cells were rinsed with PBS, naturally dried and sealed with neutral gum. Cells were then examined, and images were captured under an upright microscope.

Cell viability assay

Cell viability was determined using the LIVE/DEAD viability assay kit. Briefly, cells on coverslips were rinsed quickly with PBS (0.01 mol/l, pH 7.4) to remove the medium. Subsequently, 1 µM calcein-acetoxym-ethyl (calcein-AM) and 1 µM propidium iodide (PI) were added to the cell cultures and were incubated in the dark for 5 min at 37°C. Images were captured using a laser scanning confocal microscope.

Immunohistochemical staining

S100β protein expression was detected by immunohistochemical staining using anti-S100 (S100β), according to the manufacturer's protocol. Briefly, cells on coverslips were rinsed quickly with PBS (0.01 mol/l, pH 7.4) to remove the medium. Subsequently, the cells were fixed in 4% paraformaldehyde at room temperature for 40 min. After washing three times with PBS and permeabilizing with 3% Triton X-100 for 5 min, cells were incubated with 3% H2O2 for 10 min at room temperature, in order to suppress endogenous peroxidase activity. The cells were then treated with goat serum for 10 min at room temperature to block nonspecific staining. Subsequently, the cells were incubated with rat monoclonal anti-S100 antibody (S100β; 1:150 dilution) overnight at 4°C in a humidified chamber. After washing three times with PBS, secondary antibodies (cat. no. SP-9000; 1:50; OriGene Technologies, Inc., Beijing, China) and biotin-labeled horseradish peroxidase (OriGene Technologies, Inc.) were successively added for 15 and 10 min at room temperature. The chromogenic reaction of S100 was visualized using a DAB kit, and the slides were counterstained with hematoxylin. Finally, cells were gradually dehydrated, sealed with neutral gum, observed, and images were captured under an upright microscope.

RT-qPCR analysis

To further explore the effects of Andro on the expression of Schwann cell-specific genes, BDNF, GDNF and CNTF mRNA expression was analyzed by RT-qPCR. Total RNA was extracted from RSC96 cells using an RNeasy RNA extraction kit, according to the manufacturer's protocol. Reverse transcription of RNA was performed at 25°C for 5 min, 42°C 60 min and then 72°C for 5 min using a reverse transcription kit (Fermentas; Thermo Fisher Scientific, Inc.). The RT-qPCR reactions were performed using a qPCR detection system with a FastStart Universal SYBR Green Master Mix under the following conditions: 10 min at 95°C, 15 sec at 95°C and 1 min at 60°C for 35 cycles. The primer sequences (BGI, Shenzhen, China) for BDNF, GDNF, CNTF and glyceraldehyde 3-phosphate dehydrogenase (GAPDH; internal control) are listed in Table I. The melting curve data were collected to verify PCR specificity. Each gene was analyzed in triplicate to diminish operation errors. Relative gene expression levels were calculated using the 2−ΔΔCq method (28), and were normalized to GAPDH gene expression. Each gene was analyzed in quintuplicate to reduce randomization error.

Table I

Primer sequences used in quantitative polymerase chain reaction.

Table I

Primer sequences used in quantitative polymerase chain reaction.

GenePrimer sequence (5′ to 3′)Length (bp)Amplicon size (bp)
GDNFF: AGACCGGATCCGAGGTGC18129
R: TCGAGAAGCCTCTTACCGGC20
BDNFF: TACCTGGATGCCGCAAACAT20182
R: TGGCCTTTTGATACCGGGAC20
CNTFF: ATGGCTTTCGCAGAGCAAAC20191
R: CAACGATCAGTGCTTGCCAC20
GAPDHF: GTCATCATCTCAGCCCCCTC2099
R: GGATGCGTTGCTGACAATCT20

[i] GDNF, glial cell-derived neurotrophic factor; BDNF, brain-derived neurotrophic factor; CNTF, ciliary neurotrophic factor; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; F, forward primer; R, reverse primer; bp, base pairs.

Statistical analysis

Statistical analyses were conducted using SPSS software, version 17.0 (SPSS, Inc., Chicago, IL, USA). Data are presented as the mean ± standard deviation. Statistical significance was determined using one-way analysis of variance followed by Dunnett's post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Cytotoxicity assay

The present study examined the cytotoxicity of various concentrations of Andro on RSC96 cells using the MTT assay. Cells were treated with increasing concentrations of Andro (0–50 µM). As shown in Fig. 2, compared with the control group (0 µM), treatment with Andro between 0.78 and 12.5 µM exhibited low cytotoxicity. In addition, 0.78–12.5 µM Andro significantly accelerated cell growth (P<0.05) with the most obvious effect being observed when used at 3.125 µM (P<0.05). However, Andro exhibited a suppressive effect on RSC96 cells in vitro when used between 12.5 and 50 µM, as compared with the control group.

Cell proliferation

As presented in Fig. 3, RSC96 cells treated with 1.5625, 3.125 and 6.25 µM Andro exhibited increased proliferation compared with the control group (0 µM Andro). Proliferation was determined according to DNA content (P<0.05), which was markedly higher in the Andro groups compared with in the control group after the same culture period. Among the three concentrations, 3.125 µM Andro exhibited the strongest effect on cell growth at all time points.

Cell morphology

HE staining was conducted using an upright microscope to assess the morphology of RSC96 cells. The images indicated that the Andro groups exhibited increased cell growth compared with the control group at the same time point (Fig. 4). There were no marked differences in Schwann cell morphology between the groups after 6 days of culture. Compared with the control group, RSC96 cells in the presence of Andro grew better and had a distinctive proliferative tendency that gradually increased with time. In addition, when used at 3.125 µM, Andro was able to enhance the proliferation of RSC96 cells compared with the other two concentrations in vitro.

Cell viability assay

As presented in Fig. 5 viable cells and dead cells were stained with calcein-AM/PI. The results demonstrated that Andro exerted positive effects on survival. Images of calcein-AM/PI staining demonstrated that the survival of cells in the Andro groups was increased compared with in the control group. Consistent with the results of a cell proliferation assay (Fig. 4), more viable cells than dead cells were detected in the Andro groups, thus implying that Andro was able to better support cell growth compared with the control group. Among the Andro groups, treatment with 3.125 µM exhibited the best effects, as evidenced by an increase in the number of viable cells.

S100β secretion

The present study detected Schwann cell-specific protein S100β expression using immunohistochemical staining (Fig. 6). Positive S100β staining was increased in the Andro groups compared with the control group at the same time points. Among the three doses of Andro tested, 3.125 µM was superior compared with the others in terms of phenotypic maintenance of Schwann cells.

Gene expression

The mRNA expression levels of RSC96 cell-specific genes were determined by RT-qPCR analysis. Nerve growth factor (NGF) and several neurotrophic factors, including BDNF, GDNF and CNTF, have key roles in Schwann cells and the regeneration of peripheral nerves. The mRNA expression levels of BDNF, GDNF and CNTF were significantly increased in the Andro-treated groups compared with the control group (Fig. 7) except for BDNF levels at 6.25 µM concentratio. Furthermore, among all of the groups, 3.125 µM Andro exhibited the best effect on upregulation of BDNF, GDNF and CNTF.

Discussion

Andro is a diterpenoid lactone predominantly extracted from Andrographis paniculata, which is widely used in China and other regions of Asia for the treatment of inflammation-associated diseases. In addition, Andro has been reported to have neuroprotective properties (2931). Previous studies demonstrated that Andro reduced inflammation-mediated dopaminergic neurodegeneration in mesencephalic neuron-glia cultures by inhibiting microglial activation, thus indicating that Andro may have clinical use for the treatment of Parkinson's disease (17,32). The present study suggested that Andro enhanced neuroprotection and regeneration of peripheral nerves following injury, via its effect on the growth and phenotypic maintenance of RSC96 cells in vitro. The results indicated that Andro was able to promote RSC96 cell growth compared with the control group (Fig. 2). In addition, Andro markedly enhanced DNA synthesis and accelerated the proliferation of RSC96 cells (Figs. 3Figure 45).

Consistent with the increased synthesis of DNA in RSC96 cells, Andro was able to upregulate the mRNA expression levels of BDNF, GDNF and CNTF (Fig. 7). NGF and several neurotrophic factors, including BDNF, GDNF and CNTF, have been reported to exert stimulatory effects on specific neuronal populations (33,34). Neurotrophic factor-based molecular therapies have potential for enhancing functional recovery, as well as for increasing nerve regeneration (35), since they affect several important aspects of regeneration, including axon growth, Schwann cell function and myelination (36). In addition, previous studies regarding molecular therapeutics have concentrated primarily on the creation of neurotrophin factor mimetics, particularly NGF, neurotrophin-3 and BDNF mimetics (3739). It was reported that the secretion of neurotrophic factors by Schwann cells which were notably increased by Andro in the present study is necessary to promote axon growth and prevent neurons from initiating apoptosis (23,40). Therefore, the probable underlying mechanism is that Andro promoted RSC96 cell growth and neurotrophic factor secretion, thus inducing phenotypic maintenance via modulation of BDNF, GDNF and CNTF expression.

In the present study, the PCR, biochemical and immunohistochemical analyses demonstrated that S100β, a specific protein of Schwann cells, was effectively increased in Andro groups (Figs. 6 and 7). The S100β family, which consists of specific Schwann cell markers, is a family of low molecular weight proteins characterized by two calcium-binding sites, which is highly conserved among vertebrates (41). Furthermore, S100β, from the S100 protein family, has been identified as a potential important factor that contributes to neuronal development and differentiation (42,43). In addition, S100A4 is capable of stimulating neuronal differentiation in cultures of rat hippocampal neurons (44). In the present study, S100β protein expression was elevated in the Andro-treated cells. The modulation of S100β expression following treatment with Andro suggested that Andro may increase proliferation of RSC96 cells and maintain their phenotype.

The present study is a preliminary exploration regarding the effects of Andro on the proliferation and phenotype maintenance of RSC96 cells. Following treatment with the recommended concentrations of Andro (0.78–12.5 µM), the proliferation of RSC96 cells was accelerated in vitro. However, we cannot confirm whether Andro is suitable for the treatment of Schwann cells from other species, including humans. Further studies are required to elucidate the underlying mechanisms of the effects of Andro on Schwann cells, with the aim of identifying a promising anti-neuroinflammatory and neuroprotective agent. Furthermore, the application of Andro on peripheral nerve injury should be investigated.

In conclusion, Andro, which is the primary active component isolated from A. paniculata, exerted positive effects on the proliferation and phenotypic maintenance of RSC96 cells in vitro. These results suggested that Andro may serve as a promising therapeutic agent for peripheral nerve regeneration and neural tissue engineering. The present study may provide evidence for the clinical application of Andro.

Acknowledgments

The present study was financially supported by the Innovation Project of Guangxi Graduate Education of China (grant no. YCSZ2015124) and the National Natural Science Foundation of China (grant no. 81160221). The study was supported by the Research Center for Regenerative Medicine and Collaborative Innovation Center of Guangxi Biological Medicine.

References

1 

Lundborg G: A 25-year perspective of peripheral nerve surgery: Evolving neuroscientific concepts and clinical significance. J Hand Surg Am. 25:391–414. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Robinson LR: Traumatic injury to peripheral nerves. Suppl Clin Neurophysiol. 57:173–186. 2004. View Article : Google Scholar

3 

Evans GR: Peripheral nerve injury: A review and approach to tissue engineered constructs. Anat Rec. 263:396–404. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Raine CS: Multiple sclerosis: Immune system molecule expression in the central nervous system. J Neuropathol Exp Neurol. 53:328–337. 1994. View Article : Google Scholar : PubMed/NCBI

5 

Rogers J and Shen Y: A perspective on inflammation in Alzheimer's disease. Ann N Y Acad Sci. 924:132–135. 2000. View Article : Google Scholar

6 

Qin L, Liu Y, Wang T, Wei SJ, Block ML, Wilson B, Liu B and Hong JS: NADPH oxidase mediates lipopolysaccharide-induced neurotoxicity and proinflammatory gene expression in activated microglia. J Biol Chem. 279:1415–1421. 2004. View Article : Google Scholar

7 

Zochodne DW: The microenvironment of injured and regenerating peripheral nerves. Muscle Nerve Suppl. 9:S33–S38. 2000. View Article : Google Scholar

8 

Dezawa M: Central and peripheral nerve regeneration by transplantation of Schwann cells and transdifferentiated bone marrow stromal cells. Anat Sci Int. 77:12–25. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Wang L, Sanford MT, Xin Z, Lin G and Lue TF: Role of Schwann cells in the regeneration of penile and peripheral nerves. Asian J Androl. 17:776–782. 2015.PubMed/NCBI

10 

Puri A, Saxena R, Saxena RP, Saxena KC, Srivastava V and Tandon JS: Immunostimulant agents from Andrographis paniculata. J Nat Prod. 56:995–999. 1993. View Article : Google Scholar : PubMed/NCBI

11 

Zhang XF and Tan BK: Antihyperglycaemic and anti-oxidant properties of Andrographis paniculata in normal and diabetic rats. Clin Exp Pharmacol Physiol. 27:358–363. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Shen YC, Chen CF and Chiou WF: Andrographolide prevents oxygen radical production by human neutrophils: Possible mechanism(s) involved in its anti-inflammatory effect. Br J Pharmacol. 135:399–406. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Zhu T, Wang DX, Zhang W, Liao XQ, Guan X, Bo H, Sun JY, Huang NW, He J, Zhang YK, et al: Andrographolide protects against LPS-induced acute lung injury by inactivation of NF-κB. PLoS One. 8:e564072013. View Article : Google Scholar

14 

Bao Z, Guan S, Cheng C, Wu S, Wong SH, Kemeny DM, Leung BP and Wong WS: A novel antiinflammatory role for andrographolide in asthma via inhibition of the nuclear factor-kappaB pathway. Am J Respir Crit Care Med. 179:657–665. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Chen JX, Xue HJ, Ye WC, Fang BH, Liu YH, Yuan SH, Yu P and Wang YQ: Activity of andrographolide and its derivatives against influenza virus in vivo and in vitro. Biol Pharm Bull. 32:1385–1391. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Li J, Luo L, Wang X, Liao B and Li G: Inhibition of NF-kappaB expression and allergen-induced airway inflammation in a mouse allergic asthma model by andrographolide. Cell Mol Immunol. 6:381–385. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Wang T, Liu B, Zhang W, Wilson B and Hong JS: Andrographolide reduces inflammation-mediated dopaminergic neurodegeneration in mesencephalic neuron-glia cultures by inhibiting microglial activation. J Pharmacol Exp Ther. 308:975–983. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Wen L, Xia N, Chen X, Li Y, Hong Y and Liu Y, Wang Z and Liu Y: Activity of antibacterial, antiviral, anti-inflammatory in compounds andrographolide salt. Eur J Pharmacol. 740:421–427. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Ku CM and Lin JY: Anti-inflammatory effects of 27 selected terpenoid compounds tested through modulating Th1/Th2 cytokine secretion profiles using murine primary splenocytes. Food Chem. 141:1104–1113. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Chen YY, Hsu MJ, Sheu JR, Lee LW and Hsieh CY: Andrographolide, a novel NF- κB inhibitor, induces vascular smooth muscle cell apoptosis via a Ceramide-p47phox-ROS signaling cascade. Evid Based Complement Alternat Med. 2013:8218132013.

21 

Talei D, Valdiani A, Maziah M, Sagineedu SR and Saad MS: Analysis of the anticancer phytochemicals in andrographis paniculata Nees. Under salinity stress. Biomed Res Int. 2013:3190472013. View Article : Google Scholar : PubMed/NCBI

22 

Manikam SD and Stanslas J: Andrographolide inhibits growth of acute promyelocytic leukaemia cells by inducing retinoic acid receptor-independent cell differentiation and apoptosis. J Pharm Pharmacol. 61:69–78. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Lehmann HC and Höke A: Schwann cells as a therapeutic target for peripheral neuropathies. CNS Neurol Disord Drug Targets. 9:801–806. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Huang J, Hu X, Lu L, Ye Z, Zhang Q and Luo Z: Electrical regulation of Schwann cells using conductive polypyrrole/chitosan polymers. J Biomed Mater Res A. 93:164–174. 2010.

25 

Rotshenker S: Wallerian degeneration: The innate-immune response to traumatic nerve injury. J Neuroinflammation. 8:1092011. View Article : Google Scholar : PubMed/NCBI

26 

Bosse F: Extrinsic cellular and molecular mediators of peripheral axonal regeneration. Cell Tissue Res. 349:5–14. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Yuan H, Zhang J, Liu H and Li Z: The protective effects of resveratrol on Schwann cells with toxicity induced by ethanol in vitro. Neurochem Int. 63:146–153. 2013. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Chan SJ, Wong WS, Wong PT and Bian JS: Neuroprotective effects of andrographolide in a rat model of permanent cerebral ischaemia. Br J Pharmacol. 161:668–679. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Zaitone SA, Abo-Elmatty DM and Shaalan AA: Acetyl-L-carnitine and α-lipoic acid affect rotenone-induced damage in nigral dopaminergic neurons of rat brain, implication for Parkinson's disease therapy. Pharmacol Biochem Behav. 100:347–360. 2012. View Article : Google Scholar

31 

Jalali-Nadoushan M and Roghani M: Alpha-lipoic acid protects against 6-hydroxydopamine-induced neurotoxicity in a rat model of hemi-parkinsonism. Brain Res. 1505:68–74. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Zhang Z, Lai D, Wang L, Yu P, Zhu L, Guo B, Xu L, Zhou L, Sun Y, Lee SM and Wang Y: Neuroprotective effects of the andrographolide analogue AL-1 in the MPP+/MPTP-induced Parkinson's disease model in vitro and in mice. Pharmacol Biochem Behav. 122:191–202. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Aloe L, Rocco ML, Bianchi P and Manni L: Nerve growth factor: From the early discoveries to the potential clinical use. J Transl Med. 10:2392012. View Article : Google Scholar : PubMed/NCBI

34 

Bothwell M: NGF, BDNF, NT3, and NT4. Handb Exp Pharmacol. 220:3–15. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Daly W, Yao L, Zeugolis D, Windebank A and Pandit A: A biomaterials approach to peripheral nerve regeneration: Bridging the peripheral nerve gap and enhancing functional recovery. J R Soc Interface. 9:202–221. 2012. View Article : Google Scholar

36 

Klimaschewski L, Hausott B and Angelov DN: The pros and cons of growth factors and cytokines in peripheral axon regeneration. Int Rev Neurobiol. 108:137–171. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Xie Y and Longo FM: Neurotrophin small-molecule mimetics. Prog Brain Res. 128:333–347. 2000. View Article : Google Scholar : PubMed/NCBI

38 

Peleshok J and Saragovi HU: Functional mimetics of neurotrophins and their receptors. Biochem Soc Trans. 34:612–617. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Colangelo AM, Bianco MR, Vitagliano L, Cavaliere C, Cirillo G, De Gioia L, Diana D, Colombo D, Redaelli C, Zaccaro L, et al: A new nerve growth factor-mimetic peptide active on neuropathic pain in rats. J Neurosci. 28:2698–2709. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Gordon T: The role of neurotrophic factors in nerve regeneration. Neurosurg Focus. 26:E32009. View Article : Google Scholar : PubMed/NCBI

41 

Zhu H, Wang WJ, Ding WL, Li F and He J: Effect of panaxydol on hypoxia-induced cell death and expression and secretion of neurotrophic factors (NTFs) in hypoxic primary cultured Schwann cells. Chem Biol Interact. 174:44–50. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Donato R: S100: A multigenic family of calcium-modulated proteins of the EF-hand type with intracellular and extracellular functional roles. Int J Biochem Cell Biol. 33:637–668. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Donato R: Intracellular and extracellular roles of S100 proteins. Microsc Res Tech. 60:540–551. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Novitskaya V, Grigorian M, Kriajevska M, Tarabykina S, Bronstein I, Berezin V, Bock E and Lukanidin E: Oligomeric forms of the metastasis-related Mts1 (S100A4) protein stimulate neuronal differentiation in cultures of rat hippocampal neurons. J Biol Chem. 275:41278–41286. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 14 Issue 4

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
Xu F, Wu H, Zhang K, Lv P, Zheng L and Zhao J: Pro‑neurogenic effects of andrographolide on RSC96 Schwann cells in vitro. Mol Med Rep 14: 3573-3580, 2016
APA
Xu, F., Wu, H., Zhang, K., Lv, P., Zheng, L., & Zhao, J. (2016). Pro‑neurogenic effects of andrographolide on RSC96 Schwann cells in vitro. Molecular Medicine Reports, 14, 3573-3580. https://doi.org/10.3892/mmr.2016.5717
MLA
Xu, F., Wu, H., Zhang, K., Lv, P., Zheng, L., Zhao, J."Pro‑neurogenic effects of andrographolide on RSC96 Schwann cells in vitro". Molecular Medicine Reports 14.4 (2016): 3573-3580.
Chicago
Xu, F., Wu, H., Zhang, K., Lv, P., Zheng, L., Zhao, J."Pro‑neurogenic effects of andrographolide on RSC96 Schwann cells in vitro". Molecular Medicine Reports 14, no. 4 (2016): 3573-3580. https://doi.org/10.3892/mmr.2016.5717