Open Access

The protective mechanism of action of plantamajoside on a rat model of acute spinal cord injury

  • Authors:
    • Hua Hu
    • Xiaofei Jian
  • View Affiliations

  • Published online on: February 19, 2021     https://doi.org/10.3892/etm.2021.9809
  • Article Number: 378
  • Copyright: © Hu 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

Acute spinal cord injury (ASCI) is a severe traumatic disease of the central nervous system, characterized by a high incidence and high morbidity, for which there are no effective drug therapies in the clinic. A rat model of ASCI was established to study the effects of plantamajoside (PMS) treatment on the expression of apoptotic factors, including caspase‑3, caspase‑9, poly (ADP‑ribose) polymerase (PARP), Bax and Bcl‑2. The Allen's weight hit rat ASCI model was used for the present study, and the rats were treated with various concentrations of PMS. The behavior of rats was assessed using the Basso‑Beattle‑Bresnahan locomotor rating scale (BBB), the histopathologic changes of spinal cord tissue were observed by hematoxylin and eosin staining, the survival of neurons was assessed by TUNEL staining and the expression levels of apoptotic proteins such as caspase‑3, caspase‑9, PARP, Bcl‑2 and Bax was measured using western blot assays and RT‑qPCR. It was observed that PMS could reverse the decrease in the BBB score after ASCI, improve the morphological characteristics of the spinal cord, reduce the degree apoptosis and affect the expression of caspase‑3, caspase‑9, PARP, Bax and Bcl‑2 in a concentration dependent manner. In conclusion, PMS protected ASCI rats by inhibiting apoptosis; therefore PMS may be a potential candidate for ASCI therapy.

Introduction

Acute spinal cord injury (ASCI) is a fatal central nervous system disease, which usually causes paralysis below the contused spinal cord segment (1). ACSI not only brings great pain to patients, but also causes a serious socio-economic burden (2), with ~23 cases per million occurring every year, globally (3). The underlying pathological mechanisms of action behind ASCI are tissue edema after injury, which eventually lead to inflammation and apoptosis (4). However, the treatment of ACSI remains a major problem for researchers and clinicians, and there is no effective treatment for patients with ASCI (5). ASCI is divided into direct injury and secondary injury. Secondary injuries result from inflammation, altered Ca2+ homeostasis, oxidative stress and apoptosis (6). Apoptosis is one of the most important causes of spinal cord dysfunction and can dramatically impact the recovery for patients with ASCI (7). It has been reported that the expression levels of apoptosis relevant factors, including caspase-3, Bax and Bcl-2, are altered, accompanied with increasing neuronal apoptosis, after ASCI (8).

Plantamajoside (PMS; C29H36O16) belongs to the phenylpropanoid glycosides family, which is a unique component identified in Herba plantaginis (9). PMS has numerous beneficial pharmacological effects. PMS protects advanced glycation end-induced endothelial cells against inflammatory cellular dysfunction (10). In addition, PMS ameliorates lipopolysaccharide (LPS)-induced acute lung injury through improving pulmonary inflammation (11). PMS also inhibits LPS-induced mucin 5AC expression and inflammation through suppressing the PI3K/Akt and NF-κB signaling pathways (12). Moreover, it has been documented that PMS inhibits growth and metastasis of breast cancer by inhibiting the activity of MMP9 and MMP2(13). Together, PMS has been shown to have anti-oxidant, anti-inflammatory, anti-cancer and anti-proliferative activities (14-16). However, to the best of our knowledge, there are no studies that have investigated the effects of PMS on apoptosis in rats after ASCI. Therefore, the present study aimed to investigate whether PMS could protect against apoptosis in ASCI rats and to elucidate the potential anti-apoptosis mechanisms of action that are involved in the expression of the Bcl-2 and Bax, as well as the caspase-3 signaling pathway.

Materials and methods

Experimental animals

A total of 36 adult male Sprague-Dawley rats (weight, 200-250 g; age, 9-11 weeks) were purchased from Hubei Provincial Institute of Science and Technology. Rats were raised in a suitable environment with 24±3˚C and 12-h light/dark cycle in separated cages (relative humidity 55-60%). All rats had free access to food and water and they were allowed to acclimate to the environment for at least for three days before the experimental procedure. All of the study protocols were approved by the Ethics Committee on Animal Experiments of Tongji Medical College, Huazhong University of Science and Technology.

Rat model of spinal cord injury

Rats were randomly assigned into six groups, namely: Sham, model, positive, PMS 80 mg/kg, PMS 40 mg/kg and PMS 20 mg/kg groups. The positive group was treated with methylprednisolone 30 mg/kg as a positive control (17). The ASCI model of rats was established according to Allen's weight hit model (18). Drinking was prohibited until the surgery had been finished. All rats were anesthetized with 3% chloral hydrate (450 mg/kg) by intraperitoneal injection and maintained in the prostrate position for surgery. Fur around the chest and abdomen of these rats was shaved. A 3 cm incision was performed at the position of the eighth thoracic vertebrae and subsequently the dura mater was exposed. A 25 g cm (10 g x 2.5 cm) injury to the spinal cord was set as the injury gravity, which induces a moderate injury (19). Following induction of the injury, the wound was sutured. The following standards were used to evaluate whether a successful rat model was made: i) Spinal cord ischemia and edema around the wound; ii) flicking of the body and legs as well as the appearance of the tail sway reflex, and iii) the above symptoms coupled with sluggish paralysis. To prevent infection of the wound, liquid ampicillin (8 x 105 U/kg; Pureone Bio Technology Co., Ltd.) was injected into the back, exterior muscles once every day, for three days. In order to keep each cage dry, the padding was changed daily. To establish the autonomic urinary reflex of the rats, the bladder was massaged twice per day. Three days after the surgery, all animals were intraperitoneally injected with pentobarbital (200 mg/kg; Beijing Huaye Huanyu Chemical Co., Ltd.) for euthanasia prior to further investigation.

Evaluation of neuronal function recovery

The neuronal function recovery after injury was scored in accordance with the 21-point Basso-Beattie-Bresnahan (BBB) scale, which was scored as 0-21 representing complete paralysis to normal locomotion, respectively. BBB scores categorize combinations of rat hindlimb movements; joint movement; weight support; fore/hindlimb coordination; trunk position and stability; stepping; paw placement; toe clearance; and tail position, representing sequential recovery stages that rats attain after ACSI (20). Rats were allowed to move randomly and scored over 4 min by two independent observers who were blinded to the experiments. The hindlimb movement ability was assessed at 24, 48 and 72 h after surgery. The ability of the hindlimb joints was firstly assessed with scores between 0-7. Subsequently the pace and coordination abilities of the hindlimbs were assessed (0-7 scores) and then the delicate abilities of paws during movement were assessed (0-7 scores).

Hematoxylin and eosin (H&E) staining

A total of 3 days after surgery, 0.9% NaCl solution was obtained to transcardially perfuse the rats, and subsequently followed by 4% paraformaldehyde (PFA) for 30 min. In order to post-fix spinal cords, they were dissected out and placed in 4% PFA for 12 h at 4˚C. The spinal cords were then further embedded in paraffin at room temperature and 5-µm thick, serial transverse sections were made. These slices were subsequently stained with H&E dye for conventional morphological evaluation to evaluate the relative changes. Samples were stained with hematoxylin for 10 min and with eosin for 2 min, both at room temperature.

TUNEL staining

A TUNEL detection kit (Roche Diagnostics) was used to assess DNA fragmentation. The spinal cord specimens were preserved in 4% paraformaldehyde for 12 h at 4˚C and then washed with PBS, embed in paraffin and cut into 5-µm thick sections. The dewaxed sections were then incubated with 1:200 proteinase K for 10 min at 37˚C, followed by rinsing with PBS three times. The slides were then immersed in TUNEL assay reaction mixture and incubated at 37˚C for 1 h. Subsequently, 50 µl DAB substrate was added to the tissue and the reaction was carried out at room temperature for 10 min. Cell nuclei of apoptotic cells were distinguished by the presence of dark brown staining. The positive cells were counted in five arbitrarily selected fields in each slide (magnification, x400) using an optic microscope. Cell apoptosis (%) was calculated using the following formula: (the number of positive cells/the total cells) x 100%.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA from the spinal cord samples was extracted using TRIzol® reagent (Invitrogen, Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. A total of 600 ng of RNA was used for cDNA synthesis, at a temperature of 42˚C for 60 min and 75˚C for 5 min, using a RT First Strand kit (SA Biosciences LLC). The RT-PCR amplification was performed using iTaq™ Universal SYBR® Green Supermix (Bio-Rad Laboratories, Inc.) on an ABI Prism 7500 sequence detection system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The thermocycling profiles were as follow: 95˚C for 1 min; 40 cycles of 95˚C for 15 sec, 60˚C for 30 sec and an extension at 72˚C for 30 sec. The primers used in the present study were as follows: Bcl-2, forward 3'-GCTGGGGATGACTTCTCTCG-5' and reverse 3'-CCACAATCCTCCCCCAGTTC-5'; Bax, forward 3'-CAC CAAGAAGCTGAGCGAGT-5' and reverse 3'-TAGAAA AGGGCAACCACCCG-5'; caspase-3, forward 3'-CGGACC TGTGGACCTGAAAA-5' and reverse 3'-CGTACAGTT TCAGCATGGCG-5'; caspase-9, forward 3'-TCTTGAGAC TCGAGGGAGGC-5' and reverse 3'-GGTCGTTCT TCACCTCCACC-5'; poly (ADP-ribose) polymerase (PARP), forward 3'-AGCCAATGTTCGAGTCGTGT-5' and reverse 3'-ACAGCATCCTCTTTGGACGG-5'; and GAPDH, forward 3'-TTTCGGTACGGTTAGTAG-5' and reverse 3'-TTTGACCTTGCCTTCCAC-5'. The relative expression levels of the genes were normalized to GAPDH. Data were comparatively analyzed using the 2-ΔΔCq method (21).

Western blot analysis

Proteins from the spinal cord samples were extracted using RIPA lysis buffer kit (Omega Bio-Tek, Inc.) and the concentration was detected using a BCA protein assay kit (Bio-Rad Laboratories, Inc.). Equal amounts of proteins (40 µg per lane) were loaded into 10% SDS-polyacrylamide gels and transferred onto a PVDF membrane (EMD Millipore). The membranes were subsequently blocked with 5% skimmed milk for 1 h at room temperature and incubated with primary antibodies overnight at 4˚C, including: Anti-Bcl-2 (sc-56015; 1:1,000), anti-Bax (sc-20067; 1:1,000), anti-caspase-3 (sc-56053; 1:1,000), anti-caspase-9 (sc-81650; 1:1,000), anti-PARP (sc-56197; 1:1,000) and anti-GAPDH (sc-293335; 1:1,000) antibodies, which were purchased from Santa Cruz Biotechnology, Inc. The membranes were then incubated with goat anti-rabbit horseradish peroxidase-conjugated secondary antibodies (Santa Cruz Biotechnology, Inc.) for 1 h at room temperature. Enhanced chemiluminescence (Applygen Technologies, Inc.) was used for visualization. Band intensities were quantified using ImageJ software (v1.52r; National Institutes of Health). GAPDH was used as an endogenic control in all samples.

Statistical analysis

All results were confirmed in at least three independent experiments and analyses were performed using SPSS v14.0 software (SPPS, Inc.). All quantitative data are presented as the mean ± SD. The data graded by the scoring system was analyzed using a Kruskal-Wallis test, with post-hoc Dunn's test. Statistical comparisons with normally distributed data was made using ANOVAs followed by Turkey's post hoc tests among the groups. P<0.05 was considered to indicate a statistically significant difference.

Results

The rat ASCI model assessment

The rat model of ASCI was established using Allen's weight hit model (18). Rat functional deficits were evaluated using the BBB score until 28 days after ASCI. It was found that rats in the model group walked abnormally, with bilateral hind limb paralysis, and the BBB score of the model group was substantially lower than the sham group across the observed timeframe (Fig. 1). These results indicated that the rat ASCI model was established successfully.

PMS improves the behavioral performance of ASCI rats

According to the experimental result, it was found that the BBB score in sham group stayed at ~20 points which was the highest among these groups (Fig. 1). The BBB score of the rest of the groups increased with time, but the rate of increase varied between them. Overall, the groups stayed consistently in order, namely, from highest to lowest: The Sham, positive, PMS 80 mg/kg, PMS 40 mg/kg, PMS 20 mg/kg and lastly, the model group (Fig. 1). This data suggested that PMS can improve the behavioral performance of ASCI rats in a concentration dependent manner.

PMS reduces the apoptosis of spinal cord cells

Histopathological alterations were subsequently investigated in ASCI rats. H&E staining of the spinal cord from rats at 28 days after ASCI showed that the rats in the model group had an unclear boundary between white and gray matter. The central canal displayed an abnormal morphology and some neurons were found with apoptotic bodies (Fig. 2). However, PMS improved the morphology in a concentration dependent manner. The neurons treated with PMS presented a better histologic characteristics relative to the model group, especially in the PMS 80 mg/kg group (Fig. 2), indicated that PMS could partially improve the morphology of the spinal cord structure. TUNEL staining was also performed which showed the extent of apoptosis induced neuronal damage. The number of TUNEL-positive cells notable increased in the model group, compared with the sham group (Fig. 3). Moreover, the number of apoptotic cells in the PMS treated groups had significantly decreased compared with the model group, with further decreases in a concentration dependent manner (Fig. 3). These results indicated that PMS may inhibit apoptosis and protect the spinal cord cells after ASCI.

The mechanisms of action behind the inhibition of apoptosis by PMS

To further confirm that PMS can inhibit apoptosis, the cells lysates of the spinal cords of rats at 28 days after ASCI were assessed by RT-qPCR and western blot assays, to measure the mRNA and protein expression levels of apoptosis related proteins, including caspase-3, caspase-9, PARP, Bax and Bcl-2 (Fig. 4). The RT-qPCR and western blotting data indicated that PMS significantly downregulated the expression levels of caspase-3, caspase-9, PARP and Bax compared with the model group after ASCI. In addition, Bcl-2 levels were significantly upregulated in the PMS treated rat ASCI models at both the mRNA and protein level (Fig. 4). These results suggested that PMS plays a protective role against apoptosis through modulating the expression levels of apoptotic factors.

Discussion

Damage from ASCI is mainly caused by the direct injury itself and subsequent secondary injury (22). The secondary injury results in a pathological change in the normal tissue around the injured tissue (23). Secondary injury is accompanied by a series of changes at the molecular and cellular levels, including an inflammatory reaction, oxidative stress, internal flow of calcium ions and apoptosis, in which apoptosis is an important mechanism of action behind the damage observed from the secondary injury of ASCIs (24). Caspase-3, Bcl-2 and Bax are involved in apoptosis after spinal cord injury. A number of studies have suggested that the secondary injury is the key cause of dysfunction in the central nervous system (25,26). Therefore, inhibition of neuronal apoptosis provides an opportunity for a therapeutic strategy to improve spinal cord function after ASCI (27-29).

Since its discovery, PMS has been reported to possess broad pharmacological effects, which may exert beneficial functions for numerous therapies (14,30). Studies have indicated that PMS can regulate a variety of conditions, such as renal damage and breast cancer (13,31). PMS also possesses anti-oxidant, antibiotic and anti-inflammatory activities which has been found in a number of previous reports (32,33). However, the effect of PMS on apoptosis and its underlying mechanism of action remains unclear.

Apoptosis is a genetically programmed process resulting in cell death (34). It occurs during embryonic development, tissue reconstruction, immune regulation, and tumor degeneration. Apoptosis is critical for the development of multicellular organisms, but abnormal apoptosis can cause a variety of diseases (35). In damaged spinal cords, apoptosis causes neuronal losses (36). Apoptosis can be divided into two types of pathways, the external and internal pathways. The external pathways are induced by death receptors, such as Fas receptors (37). The internal pathways are triggered by various factors, such as DNA damage and endoplasmic reticulum stress (38). The internal pathway of the cell is regulated by the Bcl-2 protein family. The main anti-apoptotic members, Bcl-2 and Bcl-xl, play a key role in the mitochondrial outer membrane to maintain membrane integrity (39). Bcl-2, is an anti-apoptotic protein which can prevent apoptosis through regulating various signaling pathways after spinal cord injury (40). Bax protein is found in the cytoplasm of mitochondria. ASCI stimulation can activate Bax protein and alter the permeability of the mitochondrial membrane, which in turn can induce neuronal apoptosis (41).

The expression of Bcl-2 and Bax directly affects the apoptosis of spinal cord neurons (8). Bax disrupts the integrity of the mitochondrial membrane, causing apoptosis factors such as cytochrome c to leak into the cytoplasm (42). The cytochrome c complex can promote the formation and activation of caspase-9, then the activated caspase-9 is cut off and activates the downstream protease cascade such as caspase-3(8). Caspase-3 is a cysteine protease, which can destroy a variety of proteases, decompose DNA, prevent the normal function of the calcium pump, cause calcium overload and eventually lead to apoptosis (43). It is considered to be the most important protease in the process of apoptosis (44). Studies have shown that caspase-3 plays an important role in ASCI and that caspase-3 positive cells are present in ischemic and traumatic spinal cord injury models (45,46). Therefore, caspase-3 expression levels may reflect the degree of apoptosis in spinal cord injuries. After caspase-3 activation, the ADP ribose polymerase, PARP-1, can also play a role in apoptosis, resulting in cell death (47,48).

In conclusion, the present study demonstrated that PMS promotes the recovery of neurological function and protects the tissue structure of the spinal cord after ASCI. The underlying mechanisms of action may be due to PMS interrupting apoptosis, thus enhancing the resistance to further damage of the spinal cord and rescuing the locomotive activity. Furthermore, it was revealed that PMS can efficiently inhibit apoptosis by regulating the expression levels of apoptotic factors, including caspase-3, caspase-9, PARP, Bax and Bcl-2.

Acknowledgements

The authors would like to thank Professor Xiaofei Jian of the Department of Orthopedics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

HH wrote the manuscript, analyzed the data and revised the manuscript. HH and XJ performed the literature search, designed the study and performed experiments. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All of the study protocols were approved by the Ethics Committee on Animal Experiments of Tongji Medical College, Huazhong University of Science and Technology.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Guan B, Chen R, Zhong M, Liu N and Chen Q: Protective effect of Oxymatrine against acute spinal cord injury in rats via modulating oxidative stress, inflammation and apoptosis. Metab Brain Dis. 35:149–157. 2020.PubMed/NCBI View Article : Google Scholar

2 

Witiw CD and Fehlings MG: Acute spinal cord injury. J Spinal Disord Tech. 28:202–210. 2015.PubMed/NCBI View Article : Google Scholar

3 

Hu W, Wang H, Liu Z, Liu Y, Wang R, Luo X and Huang Y: Neuroprotective effects of lycopene in spinal cord injury in rats via antioxidative and anti-apoptotic pathway. Neurosci Lett. 642:107–112. 2017.PubMed/NCBI View Article : Google Scholar

4 

Chamberlain JD, Meier S, Mader L, von Groote PM and Brinkhof MWG: Mortality and longevity after a spinal cord injury: Systematic review and meta-analysis. Neuroepidemiology. 44:182–198. 2015.PubMed/NCBI View Article : Google Scholar

5 

Rouanet C, Reges D, Rocha E, Gagliardi V and Silva GS: Traumatic spinal cord injury: Current concepts and treatment update. Arq Neuropsiquiatr. 75:387–393. 2017.PubMed/NCBI View Article : Google Scholar

6 

Zhong ZX, Feng SS, Chen SZ, Chen ZM and Chen XW: Inhibition of MSK1 promotes inflammation and apoptosis and inhibits functional recovery after spinal cord injury. J Mol Neurosci. 68:191–203. 2019.PubMed/NCBI View Article : Google Scholar

7 

Varma AK, Das A, Wallace G IV, Barry J, Vertegel AA, Ray SK and Banik NL: Spinal cord injury: A review of current therapy, future treatments, and basic science frontiers. Neurochem Res. 38:895–905. 2013.PubMed/NCBI View Article : Google Scholar

8 

Luo Y, Fu C, Wang Z, Zhang Z, Wang H and Liu Y: Mangiferin attenuates contusive spinal cord injury in rats through the regulation of oxidative stress, inflammation and the Bcl-2 and Bax pathway. Mol Med Rep. 12:7132–7138. 2015.PubMed/NCBI View Article : Google Scholar

9 

Li Y, Gan L, Li GQ, Deng L, Zhang X and Deng Y: Pharmacokinetics of plantamajoside and acteoside from Plantago asiatica in rats by liquid chromatography-mass spectrometry. J Pharm Biomed Anal. 89:251–256. 2014.PubMed/NCBI View Article : Google Scholar

10 

Son WR, Nam MH, Hong CO, Kim Y and Lee KW: Plantamajoside from Plantago asiatica modulates human umbilical vein endothelial cell dysfunction by glyceraldehyde-induced AGEs via MAPK/NF-κB. BMC Complement Altern Med. 17(66)2017.PubMed/NCBI View Article : Google Scholar

11 

Wu H, Zhao G, Jiang K, Chen X, Zhu Z, Qiu C, Li C and Deng G: Plantamajoside ameliorates lipopolysaccharide-induced acute lung injury via suppressing NF-κB and MAPK activation. Int Immunopharmacol. 35:315–322. 2016.PubMed/NCBI View Article : Google Scholar

12 

Ma C and Ma W: Plantamajoside inhibits lipopolysaccharide-induced MUC5AC expression and inflammation through suppressing the PI3K/Akt and NF-κB signaling pathways in human airway epithelial cells. Inflammation. 41:795–802. 2018.PubMed/NCBI View Article : Google Scholar

13 

Pei S, Yang X, Wang H, Zhang H, Zhou B, Zhang D and Lin D: Plantamajoside, a potential anti-tumor herbal medicine inhibits breast cancer growth and pulmonary metastasis by decreasing the activity of matrix metalloproteinase-9 and -2. BMC Cancer. 15(965)2015.PubMed/NCBI View Article : Google Scholar

14 

Liu F, Huang X, He JJ, Song C, Peng L, Chen T and Wu BL: Plantamajoside attenuates inflammatory response in LPS-stimulated human gingival fibroblasts by inhibiting PI3K/AKT signaling pathway. Microb Pathog. 127:208–211. 2019.PubMed/NCBI View Article : Google Scholar

15 

Yin WZ, Xu J, Li C, Dai XK, Wu T and Wen JF: Plantamajoside inhibits the proliferation and epithelial-to-mesenchymal transition in hepatocellular carcinoma cells via modulating hypoxia-inducible factor-1α-dependent gene expression. Cell Biol Int. 44:1616–1627. 2020.PubMed/NCBI View Article : Google Scholar

16 

Wang Y and Yan D: Plantamajoside exerts antifibrosis effects in the liver by inhibiting hepatic stellate cell activation. Exp Ther Med. 18:2421–2428. 2019.PubMed/NCBI View Article : Google Scholar

17 

Li Y, Gu R, Zhu Q and Liu J: Changes of spinal edema and expression of aquaporin 4 in methylprednisolone-treated rats with spinal cord injury. Ann Clin Lab Sci. 48:453–459. 2018.PubMed/NCBI

18 

Wang B, Dai W, Shi L, Teng H, Li X, Wang J and Geng W: Neuroprotection by Paeoniflorin against Nuclear Factor Kappa B-Induced Neuroinflammation on Spinal Cord Injury. BioMed Res Int. 2018(9865403)2018.PubMed/NCBI View Article : Google Scholar

19 

Yune TY, Lee JY, Jung GY, Kim SJ, Jiang MH, Kim YC, Oh YJ, Markelonis GJ and Oh TH: Minocycline alleviates death of oligodendrocytes by inhibiting pro-nerve growth factor production in microglia after spinal cord injury. J Neurosci. 27:7751–7761. 2007.PubMed/NCBI View Article : Google Scholar

20 

Basso DM, Beattie MS and Bresnahan JC: A sensitive and reliable locomotor rating scale for open field testing in rats. J Neurotrauma. 12:1–21. 1995.PubMed/NCBI View Article : Google Scholar

21 

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.PubMed/NCBI View Article : Google Scholar

22 

Oyinbo CA: Secondary injury mechanisms in traumatic spinal cord injury: A nugget of this multiply cascade. Acta Neurobiol Exp (Wars). 71:281–299. 2011.PubMed/NCBI

23 

Tator CH and Benzel EC (eds): Contemporary Management of Spinal Cord Injury: From Impact to Rehabilitation. Thieme for the American Association of Neurological Surgeons, Illinois, 2000.

24 

Dumont RJ, Okonkwo DO, Verma S, Hurlbert RJ, Boulos PT, Ellegala DB and Dumont AS: Acute spinal cord injury, part I: Pathophysiologic mechanisms. Clin Neuropharmacol. 24:254–264. 2001.PubMed/NCBI View Article : Google Scholar

25 

Jin X and Yamashita T: Microglia in central nervous system repair after injury. J Biochem. 159:491–496. 2016.PubMed/NCBI View Article : Google Scholar

26 

Hall ED, Wang JA, Bosken JM and Singh IN: Lipid peroxidation in brain or spinal cord mitochondria after injury. J Bioenerg Biomembr. 48:169–174. 2016.PubMed/NCBI View Article : Google Scholar

27 

Li G, Shen F, Fan Z, Wang Y, Kong X, Yu D, Zhi X, Lv G and Cao Y: Dynasore improves motor function recovery via inhibition of neuronal apoptosis and astrocytic proliferation after spinal cord injury in rats. Mol Neurobiol. 54:7471–7482. 2017.PubMed/NCBI View Article : Google Scholar

28 

Schwartz M and Hauben E: T cell-based therapeutic vaccination for spinal cord injury. Prog Brain Res. 137:401–406. 2002.PubMed/NCBI View Article : Google Scholar

29 

Young W: Spinal cord contusion models. Prog Brain Res. 137:231–255. 2002.PubMed/NCBI View Article : Google Scholar

30 

Han AR, Nam MH and Lee KW: Plantamajoside inhibits UVB and advanced glycation end products-induced MMP-1 expression by suppressing the MAPK and NF-κB pathways in HaCaT cells. Photochem Photobiol. 92:708–719. 2016.PubMed/NCBI View Article : Google Scholar

31 

Jung HY, Seo DW, Hong CO, Kim JY, Yang SY and Lee KW: Nephroprotection of plantamajoside in rats treated with cadmium. Environ Toxicol Pharmacol. 39:125–136. 2015.PubMed/NCBI View Article : Google Scholar

32 

Ravn HW, Mondolot L, Kelly MT and Lykke AM: Plantamajoside-A current review. Phytochem Lett. 12:42–53. 2015.

33 

Xiao D, Yang R, Gong L, Zhang Y, Xie Y and Ni S: Plantamajoside inhibits high glucose-induced oxidative stress, inflammation, and extracellular matrix accumulation in rat glomerular mesangial cells through the inactivation of Akt/NF-κB pathway. J Recept Signal Transduct Res: doi.org/10.1080/10799893.2020.1784939.

34 

Wang C, Zhang L, Ndong JC, Hettinghouse A, Sun G, Chen C, Zhang C, Liu R and Liu CJ: Progranulin deficiency exacerbates spinal cord injury by promoting neuroinflammation and cell apoptosis in mice. J Neuroinflammation. 16(238)2019.PubMed/NCBI View Article : Google Scholar

35 

Thompson CB: Apoptosis in the pathogenesis and treatment of disease. Science. 267:1456–1462. 1995.PubMed/NCBI View Article : Google Scholar

36 

Lu X, Xue P, Fu L, Zhang J, Jiang J, Guo X, Bao G, Xu G, Sun Y, Chen J, et al: HAX1 is associated with neuronal apoptosis and astrocyte proliferation after spinal cord injury. Tissue Cell. 54:1–9. 2018.PubMed/NCBI View Article : Google Scholar

37 

Zhang J, Cui Z, Shen A, Li W, Xu G, Bao G, Sun Y, Wang L, Gu H, Zhou Y, et al: Upregulation of myelin and lymphocyte protein (MAL) after traumatic spinal cord injury in rats. J Mol Histol. 44:125–134. 2013.PubMed/NCBI View Article : Google Scholar

38 

Timmins JM, Ozcan L, Seimon TA, Li G, Malagelada C, Backs J, Backs T, Bassel-Duby R, Olson EN, Anderson ME, et al: Calcium/calmodulin-dependent protein kinase II links ER stress with Fas and mitochondrial apoptosis pathways. J Clin Invest. 119:2925–2941. 2009.PubMed/NCBI View Article : Google Scholar

39 

Lan WB, Lin JH, Chen XW, Wu CY, Zhong GX, Zhang LQ, Lin WP, Liu WN, Li X and Lin JL: Overexpressing neuroglobin improves functional recovery by inhibiting neuronal apoptosis after spinal cord injury. Brain Res. 1562:100–108. 2014.PubMed/NCBI View Article : Google Scholar

40 

Edlich F: BCL-2 proteins and apoptosis: Recent insights and unknowns. Biochem Biophys Res Commun. 500:26–34. 2018.PubMed/NCBI View Article : Google Scholar

41 

Tang P, Hou H and Zhang L, Lan X, Mao Z, Liu D, He C, Du H and Zhang L: Autophagy reduces neuronal damage and promotes locomotor recovery via inhibition of apoptosis after spinal cord injury in rats. Mol Neurobiol. 49:276–287. 2014.PubMed/NCBI View Article : Google Scholar

42 

Hou Q, Cymbalyuk E, Hsu SC, Xu M and Hsu YT: Apoptosis modulatory activities of transiently expressed Bcl-2: Roles in cytochrome C release and Bax regulation. Apoptosis. 8:617–629. 2003.PubMed/NCBI View Article : Google Scholar

43 

Ying X, Tu W, Li S, Wu Q, Chen X, Zhou Y, Hu J, Yang G and Jiang S: Hyperbaric oxygen therapy reduces apoptosis and dendritic/synaptic degeneration via the BDNF/TrkB signaling pathways in SCI rats. Life Sci. 229:187–199. 2019.PubMed/NCBI View Article : Google Scholar

44 

Mueller THJ, Kienle K, Beham A, Geissler EK, Jauch KW and Rentsch M: Caspase 3 inhibition improves survival and reduces early graft injury after ischemia and reperfusion in rat liver transplantation. Transplantation. 78:1267–1273. 2004.PubMed/NCBI View Article : Google Scholar

45 

Yuan B, Pan S and Zhang WW: Effects of gangliosides on expressions of caspase-3 and NGF in rats with acute spinal cord injury. Eur Rev Med Pharmacol Sci. 21:5843–5849. 2017.PubMed/NCBI View Article : Google Scholar

46 

Yuan B, Pan S and Zhang WW: Effects of gangliosides on expressions of caspase-3 and NGF in rats with acute spinal cord injury. Eur Rev Med Pharmacol Sci. 21:5843–5849. 2017.PubMed/NCBI View Article : Google Scholar

47 

Yamasaki K, Setoguchi T, Takenouchi T, Yone K and Komiya S: Stem cell factor prevents neuronal cell apoptosis after acute spinal cord injury. Spine. 34:323–327. 2009.PubMed/NCBI View Article : Google Scholar

48 

Decker P and Muller S: Modulating poly (ADP-ribose) polymerase activity: Potential for the prevention and therapy of pathogenic situations involving DNA damage and oxidative stress. Curr Pharm Biotechnol. 3:275–283. 2002.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2021
Volume 21 Issue 4

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hu H and Hu H: The protective mechanism of action of plantamajoside on a rat model of acute spinal cord injury. Exp Ther Med 21: 378, 2021
APA
Hu, H., & Hu, H. (2021). The protective mechanism of action of plantamajoside on a rat model of acute spinal cord injury. Experimental and Therapeutic Medicine, 21, 378. https://doi.org/10.3892/etm.2021.9809
MLA
Hu, H., Jian, X."The protective mechanism of action of plantamajoside on a rat model of acute spinal cord injury". Experimental and Therapeutic Medicine 21.4 (2021): 378.
Chicago
Hu, H., Jian, X."The protective mechanism of action of plantamajoside on a rat model of acute spinal cord injury". Experimental and Therapeutic Medicine 21, no. 4 (2021): 378. https://doi.org/10.3892/etm.2021.9809