Open Access

Protective effects of extracellular polymeric substances from Aphanizomenon flos‑aquae on neurotoxicity induced by local anesthetics

  • Authors:
    • Xing Xue
    • Ying Lv
    • Yufang Leng
    • Yan Zhang
  • View Affiliations

  • Published online on: July 30, 2018     https://doi.org/10.3892/etm.2018.6540
  • Pages: 3011-3019
  • Copyright: © Xue 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

The neurotoxicity of local anesthetics has received an increasing amount of attention and more effective therapeutic agents are required. Extracellular polymeric substances from Aphanizomenon flos‑aquae (EPS‑A) are high molecular weight polysaccharides. The present study aimed to elucidate the protective effects of EPS‑A on neurotoxicity induced by local anesthetics in an intraperitoneal injection bupivacaine rat model. The results of immunohistochemical staining inicated that following intraperitoneal injection of EPS‑A the levels of apoptosis and caspase‑3 decreased, and the expression levels of microtubule‑associated protein 1A light chain 3 (LC3) and beclin1 increased. In order to further clarify the mechanism of the EPS‑A‑mediated protection, the expression of key proteins associated with autophagy was investigated by western blotting. The results suggested that the ratio of LC3‑II/LC3‑I and the expression level of beclin1 increased. Taken together, the results indicated that EPS‑A induced neuroprotective effects on bupivacaine‑induced neurotoxicity. The underlying mechanism may be associated with the inhibition of apoptosis, upregulation of autophagy and improvement of cell survival. The results suggested that EPS‑A may be a candidate neuroprotective agent against neurotoxicity caused by local anesthetics.

Introduction

At present, the neurotoxicity of local anesthetics is receiving an increasing amount of attention (1), and therefore, it is particularly important to identify safe methods of prevention and treatment. One pharmacological strategy against the neurotoxicity of local anesthetics is the use of drugs to induce autophagy to treat the associated diseases (2,3). Studies have shown that certain drugs can directly or indirectly activate autophagy to prevent cell death (4,5). Based on these observations, it has been hypothesized that the regulation of autophagy could serve as the basis for the development of candidate neuroprotective agents against neurotoxicity caused by local anesthetics. However, the components of the autophagy signaling pathway remains at an initial stage (6), therefore, the major aims of the current study were to explore drugs for the prevention and treatment of nerve injury caused by local anesthetics via the autophagy signaling pathway.

In previous years, polysaccharides have been investigated in numerous research studies and serve roles in controlling cell division and differentiation, regulate cell growth and aging, and aid in maintaining the normal metabolism of an organism (79). Numerous studies have indicated that polysaccharides can activate autophagy via a non-serine/threonine-protein kinase mTOR pathway and serve a protective role in glomerular lesions and neurodegenerative diseases (10,11). It was also thought that polysaccharides, as natural products, were more safe. Therefore, polysaccharides have attracted increasing research attention (12). Aphanizomenon flos-aquae, a filamentous, heterocystous and dominant cyanobacterium, is common in nutrient-rich freshwater cyanobacterial blooms (13). During the process of growth until death, A. flos-aquae continuously secretes extracellular polymeric substances (EPS) into the surrounding environment (13). EPS are high molecular weight polysaccharides consisting of three or four different monosaccharides arranged in groups of £10 to form repeating units (14). Numerous studies have indicated that EPS exhibit numerous bioactivities, including antitumor, immune-adjusting, antithrombotic, ameliorating endothelial cell injury, antioxidant and antiviral (14,15). Although several studies on the biological activities of EPS have been performed, with a particular focus on their anti-apoptotic activity (1618), little is known about their neuroprotective effects in vivo. Therefore, in the present study, rats received intraperitoneal injections of EPS from A. flos-aquae (EPS-A), and apoptosis and autophagy were observed in spinal cord neurons. The aim was to investigate the protective effects of EPS-A on neuronal injury and the possible underlying mechanisms.

Materials and methods

EPS-A

A strain of A.flos-aquae, isolated from Lake Dianchi in China, was obtained from the Freshwater Algae Culture Collection of the Institute of Hydrobiology, Chinese Academy of Sciences in Wuhan city of China. The culture of A. flos-aquae was performed according to the methods of Zhang et al (19), and extraction and purification of EPS-A were performed as previously described by Hu et al (20).

Animal selection and housing

The present in vivo study was approved by the Medical Ethics Committee of the First Hospital of Lanzhou University (Lanzhou, China). A total of 18 healthy adult male Sprague-Dawley rats (age, 5–7 weeks; weight, 180–220 g), were supplied by the Animal Breeding and Research Center of Lanzhou University (Lanzhou City, China). Rats were housed in separate cages with food and water freely available until the time of testing and kept in temperature-controlled rooms (20–24°C; relative humidity, 50–60%) with a 12-h light/dark cycle (6:00 a.m.-6:00 p.m.).

Groups and treatment

A total of 18 rats were randomly divided into groups A, B and C (n=6/group). In groups A and B, rats received intraperitoneal injections of 1 ml 0.9% NaCl, whole group C received an intraperitoneal injection of EPS-A (100 mg/kg), once a day for 3 days. Subsequently, the group A rats were subjected to 2% isoflurane inhalation and intrathecal injection of 0.9% NaCl 50 µl. Groups B and C received 2% isoflurane inhalation and intrathecal injection of 1% bupivacaine 50 µl (2.5 mg/kg animal body weight).

Bupivacaine in lumbar anesthesia

When optimal flexion of the rat lumbar spine was achieved in a prone position, a 27-gauge needle attached to a 100 µl syringe (model, KL-34; Hamilton Medical, Inc., Reno, NV, USA) was inserted into the midline of the lumbar 4–5 (L4-5) intervertebral space and 50 µl of the respective drug was injected. While a tail-flick indicated entrance into the intrathecal space, rats were subsequently observed for paralysis of the hind limbs, indicative of a spinal blockade. During the selection process, rats were excluded if they lacked a healthy appearance or required more than one spinal puncture. A total of 6 rats were included in each group following this.

Spinal cord section specimen

Rats in each group were sacrificed at 6 h following the aforementioned anaesthesia and the spinal cord was rapidly collected, then sections were transported in fixative (10% neutral buffered formalin) at room temperature for 36–48 h. A section of tissue was frozen in liquid nitrogen and stored at −70°C until further use.

Hematoxylin & eosin (H&E) staining

H&E staining is a standard method used for detecting morphological alterations. First, the spinal cord sections were sliced into 5 µm sections using a microtomer, deparaffinized at 40°C in a water bath and rehydrated. Samples were then washed with distilled water and dried. Hematoxylin was added for 5 min and rinsed with water. Subsequently 1% HCl ethanol solution (1 ml HCl added to 99 ml 70% ethanol) was added for 10 sec in triplicate to remove excess haematoxylin. Following this, sections were washed using distilled water for 25 min, 0.5% eosin was added for 2 min and slices were dehydrated with 95 and 100% ethanol. Dimethylbenzene (Absin Bioscience, Inc., Shanghai, China) was added for 5 min twice and incubated at 37°C for 24 h. Finally, sections were kept in a special slide container at room temperature and observed under a light microscope.

Terminal deoxynucleotidyl-transferase-mediated dUTP nick end labelling (TUNEL)

TUNEL staining was performed to examine apoptosis. The spinal cord samples were fixed in 10% neutralized formalin at room temperature for 20 min and embedded in paraffin. Sections were deparaffinized, rehydrated and incubated for 15 min at 37°C with proteinase K working solution (Shanghai Xiangsheng biotechnology Co., Ltd., Shanghai, China). Sections were rinsed twice with PBS (pH 7.4), blocked with hydrogen peroxide for 15 min, and 50 µl TUNEL reaction mixture was added and incubated for 60 min at 37°C in a humidified atmosphere in the dark. Following rinsing with PBS (pH 7.4) three times, sections were incubated for 30 min at 37°C with 50 µl converter peroxidase (Shanghai Xiangsheng biotechnology Co., Ltd.). A total of 50 µl diaminobenzidine [5 µl 20×DAB, 1 µl 30% H2O2, 94 µl PBS (pH 7.4)] was added and incubated for 10 min at 25°C. Sections were rinsed with PBS (pH 7.4) three times and cover slips were mounted onto 50% glycerol treated slides. Samples were analyzed under a light microscope (magnification, ×400). Slices were photographed continuously and six visual fields were randomly selected under an optical microscope. Pictures were numbered and random fields were obtained using random numbers generated by a computer. The final value of a slice was obtained by a mean value from random high-power fields. The optical density was quantified using Image-Pro Plus (version 7.0; Media Cybernetics, Inc., Rockville, MD, USA).

Immunohistochemical (IHC) staining

IHC is widely used in research to understand the distribution and localization of biomarkers and differentially expressed proteins in different parts of cells or tissues. In the present study, spinal cord samples were replaced with ethanol in decreasing concentrations (100, 90 and 70%), washed with PBS (pH 7.4) three times for 5 min each and heated in a water bath (95–98°C) in 0.01 M trisodium citrate (pH 6.0) for 15 min for antigen retrieval. Subsequently, the sections were incubated with a blocking reagent (3% milk and 5% fetal bovine serum; Absin Bioscience, Inc.) for 1 h at room temperature and further incubated with anti-caspase-3 (cat. no. ab20816; dilution 1:500; Abcam, Cambridge, UK), anti-microtubule-associated protein 1A light chain 3 (LC3) polyclonal rabbit antibody (cat. no. L8918; dilution 1:200; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) or anti-beclin1 (cat. no. SAB2103299; dilution 1:500; Sigma-Aldrich; Merck KGaA) at 4°C overnight. Following washing with PBS (pH 7.4), the tissue samples were exposed to biotinylated anti-rabbit immunoglobulin G (cat. no. A0239; dilution 1:1,000; Beyotime Institute of Biotechnology, Fuzhou, China) and horseradish peroxidase conjugated streptavidin (Vector Laboratories, Inc., Burlingame, CA, USA). After the slides were sealed, the sections were imaged with a confocal microscope. The optical density was quantified using Image-Pro Plus version 7.0 (Media Cybernetics, Inc., Rockville, MD, USA).

Western blot analysis

Western blotting is used for the detection and quantification of specific proteins in a sample of tissue. Protein levels of two autophagy markers, LC3 and beclin1, were detected. To investigate autophagy activation, the ratio of LC3-I to LC3-II, a marker of autophagic vacuole formation, was measured. The spinal cord samples were cut into small pieces (2 mm long; 2 mm thick) and homogenized in lysis buffer [50 mM Tris HCl (pH 7.6), 20 mM MgCl2, 150 mM NaCl, 0.5% Triton-X, 5 units.ml−1 aprotinin, 5 µg.ml-1 leupeptin, 5 µg.ml-1 pepstatin, 1 mM benzamidine, 1 mM phenylmethylsulfonyl fluoride]. Lysate protein levels were determined using a BCA protein assay kit (Shanghai Qcbio Science&Technologies Co., Ltd., Shanghai, China). Equal amounts of proteins (~30 µg) were loaded per lane and subjected to 10% SDS-PAGE, and subsequently transferred to a nitrocellulose membrane. Membranes were blocked with 5% nonfat dried milk in Tris-buffered saline with Tween 20 (TBST; 137 mM sodium chloride, 20 mM Tris, 0.1% Tween 20; Absin Bioscience, Inc.) for 1 h at room temperature. Primary antibodies specific to LC3 (cat. no. bsm-33309M; dilution 1:1,000; Biosynthesis Biotechnology Co., Ltd., Beijing, China), beclin-l (cat. no. bs-1353R; dilution 1:200; Biosynthesis Biotechnology Co., Ltd., Beijing, China), caspase-3 (cat. no. ab20816; dilution 1:500; Abcam) and β-actin (cat. no. bsm-33139M; 1:1,000; Beijing Biosynthesis Biotechnology Co., Ltd., Beijing, China) were diluted in TBST overnight at 4°C. Membranes were incubated with Goat-anti-mouse immunoglobulin G secondary antibodies conjugated to alkaline phosphatase (cat. no. A32723; dilution 1:1,000; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) for 1 h at room temperature. Reactive bands were detected by incubating with nitro blue tetrazolium and 5-bromo-4-chloro-3-indolyl phosphate (Sigma-Aldrich; Merck KGaA) for 5 min. Band densities were detected with an imaging densitometer (GS-800; Bio-Rad Laboratories, Inc., Hercules, CA, USA) and the optical density was quantified using Image-Pro Plus (version 7.0; Media Cybernetics, Inc.), which was calibrated against β-actin.

Statistical analysis

Statistical analysis was performed using SPSS software (verison 20.0; IBM Corp., Armonk, NY, USA). Data were presented as the mean ± standard deviation. All experiments were performed in triplicate. The differences between groups were evaluated by one-way analysis of variance and multiple group comparisons were performed by using Dunnett's tests. P<0.05 was considered to indicate a statistically significant difference.

Results

Pathological alterations

As demonstrated by light microscopy, in group A, spinal cord neurons in the rats were uniformly distributed, the morphology was normal, nissl bodies were clear, cell membranes were intact, the neural fibers of the white matter were arranged in an orderly manner and the intercellular matrix was uniform (Fig. 1A). However, following bupivacaine injection, the number of spinal cord neurons was reduced, neurons were shrunken and darkly stained and the nuclei were condensed (Fig. 1B). However, following administration of EPS-A, injury was ameliorated markedly (Fig. 1C). Therefore, the rat models were successfully established.

Alterations in apoptosis

TUNEL staining was used to observe neuronal apoptosis under a light microscope. The results indicated that neuronal apoptosis in the spinal cord was altered. The apoptosis level in group B increased compared with group A. Compared with group B, the level of apoptosis in group C decreased significantly (Table I; Fig. 2).

Table I.

Comparison of apoptosis and autophagy of spinal cord neurons in rats treated with bupivacaine and extracellular polymeric substances from Aphanizomenon flos-aquae as determined by terminal deoxynucleotidyl-transferase-mediated dUTP nick end labelling and immunohistochemistry.

Table I.

Comparison of apoptosis and autophagy of spinal cord neurons in rats treated with bupivacaine and extracellular polymeric substances from Aphanizomenon flos-aquae as determined by terminal deoxynucleotidyl-transferase-mediated dUTP nick end labelling and immunohistochemistry.

GroupApoptosisCasepase-3Beclin1LC3
A0.137±0.0040.157±0.0020.182±0.0060.141±0.008
B 0.164±0.005a 0.194±0.004a 0.199±0.003a 0.178±0.010a
C 0.146±0.006a,b 0.157±0.002b 0.238±0.003a,b 0.208±0.012a,b
Sum0.149±0.0120.169±0.0180.206±0.0250.175±0.030
F41.06314.47260.8768.27
P0.0000.0000.0000.000

a P<0.05 vs. group A

b P<0.05 vs. group B. LC3, microtubule-associated protein 1A light chain 3; Sum, sum of the mean ± standard deviations. F and P represent the differences between groups which were evaluated using one-way analysis of variance; a and b represent the multiple group comparisons that were confirmed using the Dunnett's test.

Alterations in the expression levels of caspase-3

The expression of casepase-3 was observed by immunohistochemical staining. Caspase-3-positive neurons were stained brown. The expression of caspase-3 in group B increased compared with group A. Compared with group B, the expression of caspase-3 in group C significantly decreased (Table I; Fig. 3).

Alterations in the expression levels of LC3 and beclin1

Immunohistochemical staining was performed to investigate alterations in the expression levels of LC3 and beclin1. The LC3- and beclin1-positive neurons were stained brown. LC3 and beclin1 levels in group B increased compared with group A. Compared with group B, the LC3 and beclin1 levels in group C significantly increased (Table I; Figs. 4 and 5).

Western blotting

In order to further determine the level of autophagy, western blot analysis was performed to observe the expression levels of LC3 and beclin1. The results of the present study indicated that the expression level of beclin1 and the ratio of LC3-II/LC3-I differed between the groups. Compared with group A, the level of beclin1 and the ratio of LC3-II/LC3-I increased significantly in group B. Compared with group B, the level of beclin1 and the ratio of LC3-II/LC3-I increased even further in group C, and these results were consistent with those of the immunohistochemical staining (Table II; Fig. 6).

Table II.

Comparison of the expression levels of beclin1 and LC3 of spinal cord neurons in rats treated with bupivacaine and extracellular polymeric substances from Aphanizomenon flos-aquae as determined by western blotting.

Table II.

Comparison of the expression levels of beclin1 and LC3 of spinal cord neurons in rats treated with bupivacaine and extracellular polymeric substances from Aphanizomenon flos-aquae as determined by western blotting.

GroupBeclin1LC3
A0.428±0.0990.141±0.008
B 0.712±0.064a 0.178±0.010a
C 0.849±0.129a,b 0.208±0.012a,b
Sum0.663±0.2030.175±0.030
F27.1121005.334
P0.0000.000

a P<0.05 vs. group A

b P<0.05 vs. group B. LC3, microtubule-associated protein 1A light chain 3; Sum, sum of the mean ± standard deviations. F and P represent the differences between groups which were evaluated using one-way analysis of variance; a and b represent the multiple group comparisons that were confirmed using the Dunnett's test.

Discussion

Local anesthetics are widely used in clinical anesthesia and pain treatment (21). When their effects end, in general, neurological function can be restored; however, their neurotoxicity has attracted an increasing amount of attention from clinicians (22,23). Identification of novel drugs which serve a protective role in neuronal injury may provide theoretical basis for further research to protect damaged neurons. Previously, studies have shown that polysaccharides can reduce the swelling and shrinking of neurons, promote nervous regeneration, reduce neurological defects and exert protective effects on damaged neurons (2426). Therefore, the present study aimed to evaluate the beneficial effects of EPS-A on neurotoxicity induced by local anesthetics in an intraperitoneal injection bupivacaine rat model.

Morphological alteration is the primary way to detect injury. H&E is one of principal histological stains and the most widely used staining method in medical diagnosis (27). The results of the present study indicated that following injection of bupivacaine, the number of spinal cord neurons in rats was reduced. However, upon administration of EPS-A, injury was ameliorated markedly. The results suggested that EPS-A markedly attenuated neuronal injury compared with bupivacaine induced neuronal injury. However, EPS-A was able to attenuate these pathological alterations and, therefore, the effect of EPS-A on the level of pathological alterations may be involved in the reduction of neuronal injury in rats treated with bupivacaine.

Apoptosis leads to the loss of a large number of neurons, which is an important mechanism of secondary spinal cord injury (28,29). It was also hypothesized to be one mechanism of neurotoxicity caused by local anesthetics (30). A previous study suggested that apoptosis is more common in the neurotoxicity of local anesthesia as induced by local anesthetics (31). In the present study, the results indicated that the apoptosis rate in spinal cord neurons increased following intrathecal injection of bupivacaine, however, the pre-administration of EPS-A resulted in a significant decrease in apoptosis. Furthermore, the results indicated that EPS-A could ameliorate the increase of apoptosis rate, thus the neuroprotective effects of EPS-A may be mediated by its capacity to reduce neuronal apoptosis.

The expression of caspase-3 is closely associated with the level of neuronal apoptosis following spinal cord injury (32). The expression of caspase-3 may also reflect the degree of neuronal damage to some extent (33). In the present study, the results indicated that the injection of bupivacaine significantly increased the expression of caspase-3. However, following treatment with EPS-A, the expression of caspase-3 significantly decreased. The results suggested that the neurotoxic effects caused by bupivacaine were associated with casepase3-dependent apoptosis. EPS-A pre-administration was able to alleviate the neurotoxic effects caused by bupivacaine and significantly reduced casepase3-dependent apoptosis, which was consistent with the results of the TUNEL staining. Therefore, the role of EPS-A in the expression of caspase-3 may be involved in the attenuation of apoptosis in rats treated with bupivacaine. The results implied that EPS-A may affect the level of apoptosis, and this may aid in understanding the beneficial neuroprotective effects of EPS-A against local anesthetics.

Autophagy serves an important role in death and survival of neuronal cells, which may influence several neurodegenerative disorders (34,35). Basal autophagy acts as one of the cytoprotective mechanisms and participates in maintaining homeostasis (36). Additionally, in disease conditions of myocardial ischemia-reperfusion and neuronal ischemic anoxia, autophagy enhancement serves a role as a protective mechanism (37). Recent studies (38,39) have shown that drug-induced autophagy is characterized by the altered expression of autophagy-associated proteins LC3 and beclin1. LC3 and beclin1 are two pacemakers in the autophagic cascade. LC3, exists in cytosolic (LC3-I) and membrane bound forms (LC3-II) (40). The ratio of conversion from LC3-I to LC3-II is closely associated with the extent of autophagosome formation (41). Beclin1 is essential for the recruitment of other autophagic proteins during the expansion of the pre-autophagosomal membrane (42,43). The results of the present study indicated that bupivacaine increased the protein expression levels of beclin1 and LC3; however, with pre-administration of EPS-A, the LC3 and beclin1 protein expression levels improved further. The present study also suggested that the induction of autophagy was associated with the alteration of the LC3II/LC3I ratio. In addition, further data confirming the protective effects of EPS-A were obtained from the western blot analysis of beclin1 and LC3. The results demonstrated that bupivacaine increased the ratio of LC3II/LC3I and the expression levels of beclin1; however, following pre-administration of EPS-A, the ratio of LC3II/LC3I and the beclin1 expression level increased further. The results implied that EPS-A may affect the autophagy level, and this may aid in understanding the beneficial neuroprotective effects of EPS-A against local anesthetics by inducing the expression of autophagy.

Autophagy is associated with apoptosis in a number of ways, which are as complex as the roles of autophagy in cell survival and death (44). It has been suggested that autophagy may be a trigger for apoptotic cell death (45), while others have argued that autophagy protects against apoptosis and inflammation; apoptosis and autophagy may cooperate, coexist or antagonize each other to balance death and survival signaling (46). In the present study, the results suggested that autophagy and apoptosis exhibited cross-inhibitory interactions; the activation of autophagy could inhibit apoptosis and autophagy could promote cell survival against apoptosis, which was consistent with a previous report (47).

In conclusion, EPS-A exhibited neuroprotective effects against bupivacaine-induced neurotoxicity in an intraperitoneal injection bupivacaine rat model. The mechanisms may be attributed to inhibiting apoptosis, inducing autophagy and improving cell survival. These results suggested that EPS-A may be a candidate neuroprotective agent against the neurotoxicity of local anesthetics. The promotion and utilization of autophagy induced by drugs has attracted an increasing amount of attention in the field of medicine. However, further details on the beneficial mechanisms of EPS-A in apoptosis and autophagy in an intraperitoneal injection model are required, and this remains to be evaluated in the future.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Gansu Province Health Industry Scientific Research Plan (grant no. GSWSKY2017-18).

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

XX wrote the manuscript. XX, YLv and YLe conceived and designed the present study. XX and YaZ performed the majority of the experimental procedures. YLv analyzed the data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Medical Ethics Committee of the First Hospital of Lanzhou University in (Lanzhou, China).

Patient consent for publication

Not applicable.

Competing interests

All authors declare that they have no competing interests.

References

1 

Verlinde M, Hollmann MW, Stevens MF, Henning H, Werdehausen R and Lirk P: Local anesthetic-induced neurotoxicity. Int J Mol Sci. 17:3392016. View Article : Google Scholar : PubMed/NCBI

2 

Chen HC, Fong TH, Hsu PW and Chiu WT: Multifaceted effects of rapamycin on functional recovery after spinal cord injury in rats through autophagy promotion, anti-inflammation, and neuroprotection. J Surg Res. 179:e203–e210. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Xiong J, Kong Q, Dai L, Ma H, Cao X, Li L and Ding Z: Autophagy activated by tuberin/mTOR/p70S6K suppression is a protective mechanism against local anaesthetics neurotoxicity. J Cell Mol Med. 21:579–587. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Klionsky DJ and Emr SD: Autophagy as a regulated pathway of cellular degradation. Science. 290:1717–1721. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Levine B and Klionsky DJ: Development by self-digestion: Molecular mechanisms and biological functions of autophagy. Dev Cell. 6:463–477. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Mizushima N and Komatsu M: Autophagy: Renovation of cells and tissues. Cell. 147:728–741. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Schepetkin IA and Quinn MT: Botanical polysaccharides: Macrophage immunomodulation and therapeutic potential. Int Immunopharmacol. 6:317–333. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Zhang M, Cui SW, Cheung PCK and Wang Q: Antitumor polysaccharides from mushrooms: A review on their isolation process, structural characteristics and antitumor activity. Trends Food Sci Tech. 18:4–19. 2007. View Article : Google Scholar

9 

Jaehrig SC, Rohn S, Kroh LW, Wildenauer FX, Lisdat F, Fleischer LG and Kurz T: Antioxidative activity of (1->3), (1->6)-beta-D-glucan from Saccharomyces cerevisiae grown on different media. LWT-Food Sci Tech. 41:868–877. 2008. View Article : Google Scholar

10 

Kang YL, Saleem MA, Chan KW, Yung BY and Law HK: Trehalose, an mTOR independent autophagy inducer, alleviates human podocyte injury after puromycin aminonucleoside treatment. PLoS One. 9:e1135202014. View Article : Google Scholar : PubMed/NCBI

11 

Emanuele E: Can trehalose prevent neurodegeneration? Insights from experimental studies. Curr Drug Targets. 15:551–557. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Zhao F, Zhao J, Song L, Zhang YQ, Guo Z and Yang KH: The induction of apoptosis and autophagy in human hepatoma SMMC-7721 cells by combined treatment with vitamin C and polysaccharides extracted from Grifola frondosa. Apoptosis. 22:1461–1472. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Lv Y, Xue X, Tao L, Zhang D, Hu C, Liu Y and Ren JF: Effect of extracellular polymeric substances from aphanizomenon flos-aquae (EPS-A) on human epidermoid carcinoma (A431 cells). Fresen Environ Bull. 24:3307–3314. 2015.

14 

Jiao G, Yu G, Zhang J and Ewart HS: Chemical structures and bioactivities of sulfated polysaccharides from marine algae. Mar Drugs. 9:196–223. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Wei W, Wang SX and Guan HS: The antiviral activities and mechanisms of marine polysaccharides: An overview. Mar Drugs. 10:2795–2816. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Xue X, Lv Y, Liu Q, Zhang X, Zhao Y, Zhang L and Xu S: Extracellular polymeric substance from Aphanizomenon flos-aquae induces apoptosis via the mitochondrial pathway in A431 human epidermoid carcinoma cells. Exp Ther Med. 10:927–932. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Imatani T, Kato T, Okuda K and Yamashita Y: Histatin 5 inhibits apoptosis in human gingival fibroblasts induced by porphyromonas gingivalis cell-surface polysaccharide. Eur J Med Res. 9:528–532. 2004.PubMed/NCBI

18 

Zhang CX and Huang KX: Mechanism of apoptosis induced by a polysaccharide, from the loach Misgurnus anguillicaudatus (MAP) in human hepatocellular carcinoma cells. Toxicol Appl Pharmacol. 210:236–245. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Zhang D, Hu C, Wang G, Li D, Li G and Liu Y: Zebrafish neurotoxicity from aphantoxins-cyanobacterial paralytic shellfish poisons (PSPs) from Aphanizomenon flos-aquae DC-1. Environ Toxicol. 28:239–254. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Hu C, Liu Y, Paulsen BS, Petersen D and Klaveness D: Extracellular carbohydrate polymers from five desert soil algae with different cohesion in the stabilization of fine sand grain. Carbohyd Polym. 54:33–42. 2003. View Article : Google Scholar

21 

Sigirci A: Pain management in total knee arthroplasty by intraoperative local anesthetic application and one-shot femoral block. Indian J Orthop. 51:280–285. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Rigler ML, Drasner K, Krejcie TC, Yelich SJ, Scholnick FT, DeFontes J and Bohner D: Cauda equina syndrome after continuous spinal anesthesia. Anesth Analg. 72:275–281. 1991. View Article : Google Scholar : PubMed/NCBI

23 

Sun Z, Liu H, Guo Q, Xu X, Zhang Z and Wang N: In vivo and in vitro evidence of the neurotoxic effects of ropivacaine: the role of the Akt signaling pathway. Mol Med Rep. 6:1455–1459. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Gao Y, Li C, Yin J, Shen J, Wang H, Wu Y and Jin H: Fucoidan, a sulfated polysaccharide from brown algae, improves cognitive impairment induced by infusion of Aβ peptide in rats. Environ Toxicol Pharmacol. 33:304–311. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Xiao J, Liong EC, Ching YP, Chang RC, Fung ML, Xu AM, So KF and Tipoe GL: Lycium barbarum polysaccharides protect rat liver from non-alcoholic steatohepatitis-induced injury. Nutr Diabetes. 3:e812013. View Article : Google Scholar : PubMed/NCBI

26 

Qi B, Ji Q, Wen Y, Liu L, Guo X, Hou G, Wang G and Zhong J: Lycium barbarum polysaccharides protect human lens epithelial cells against oxidative stress-induced apoptosis and senescence. PLoS One. 9:e1102752014. View Article : Google Scholar : PubMed/NCBI

27 

Laine L, Lewin DN, Naritoku W and Cohen H: Prospective comparison of H&E, Giemsa, and Genta stains for the diagnosis of Helicobacter pylori. Gastrointest Endosc. 45:463–467. 1997. View Article : Google Scholar : PubMed/NCBI

28 

Yune TY, Kim SJ, Lee SM, Lee YK, Oh YJ, Kim YC, Markelonis GJ and Oh TH: Systemic administration of 17beta-estradiol reduces apoptotic cell death and improves functional recovery following traumatic spinal cord injury in rats. J Neurotrauma. 21:293–306. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Whalley K, O'Neill P and Ferretti P: Changes in response to spinal cord injury with development: Vascularization, hemorrhage and apoptosis. Neuroscience. 137:821–832. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Park CJ, Park SA, Yoon TG, Lee SJ, Yum KW and Kim HJ: Bupivacaine induces apoptosis via ROS in the Schwann cell line. J Dent Res. 84:852–857. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Werdehausen R, Fazeli S, Braun S, Hermanns H, Essmann F, Hollmann MW, Bauer I and Stevens MF: Apoptosis induction by different local anaesthetics in a neuroblastoma cell line. Brit J Anaesth. 103:711–718. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Boselli E, Duflo F, Debon R, Allaouchiche B, Chassard D, Thomas L and Portoukalian J: The induction of apoptosis by local anesthetics: A comparison between lidocaine and ropivacaine. Anesth Analg. 96:755–756. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Ananth C, Dheen Thameem S, Gopalakrishnakone P and Kaur C: Domoic acid-induced neuronal damage in the rat hippocampus: Changes in apoptosis related genes (bcl-2, bax, caspase-3) and microglial response. J Neurosci Res. 66:177–190. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Nixon RA: The role of autophagy in neurodegenerative disease. Nat Med. 19:983–997. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Wang ZY, Lin JH, Muharram A and Liu WG: Beclin-1-mediated autophagy protects spinal cord neurons against mechanical injury-induced apoptosis. Apoptosis. 19:933–945. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Kim HJ, Kim J, Kang KS, Lee KT and Yang HO: Neuroprotective effect of chebulagic acid via autophagy induction in SH-SY5Y cells. Biomol Ther (Seoul). 22:275–281. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Rautou PE, Mansouri A, Lebrec D, Durand F, Valla D and Moreau R: Autophagy in liver diseases. J Hepatol. 53:1123–1134. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Hamurcu Z, Delibaşı N, Geçene S, Şener EF, Dönmez-Altuntaş H, Özkul Y, Canatan H and Ozpolat B: Targeting LC3 and Beclin-1 autophagy genes suppresses proliferation, survival, migration and invasion by inhibition of Cyclin-D1 and uPAR/Integrin β1/Src signaling in triple negative breast cancer cells. J Cancer Res Clin Oncol. 144:415–430. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Niu Y, Zhang Y, Li K, Xing C and Sun C: miR-124-3p and miR-140-3p.2 act as negative regulators of Beclin1 and LC3 expression in the liver of rat model with hepatic impact injury. Bio Res. 29:2018.(ISSN: 0970-938X).

40 

Liu C, Xu P, Chen D, Fan X, Xu Y, Li M, Yang X and Wang C: Roles of autophagy-related genes Beclin-1 and LC3 in the development and progression of prostate cancer and benign prostatic hyperplasia. Biomed Rep. 1:855–860. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Miracco C, Cevenini G, Franchi A, Luzi P, Cosci E, Mourmouras V, Monciatti I, Mannucci S, Biagioli M, Toscano M, et al: Beclin 1 and LC3 autophagic gene expression in cutaneous melanocytic lesions. Hum Pathol. 41:503–512. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Gao L, Jiang T, Guo J, Liu Y, Cui G, Gu L, Su L and Zhang Y: Inhibition of autophagy contributes to ischemic postconditioning-induced neuroprotection against focal cerebral ischemia in rats. PLoS One. 7:e460922012. View Article : Google Scholar : PubMed/NCBI

43 

Liu C, Xu P, Chen D, Fan X, Xu Y, Li M, Yang X and Wang C: Roles of autophagy-related genes Beclin-1 and LC3 in the development and progression of prostate cancer and benign prostatic hyperplasia. Biomed Rep. 1:855–860. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Xiao M, Li L, Li C, Zhang P, Hu Q, Ma L and Zhang H: Role of autophagy and apoptosis in wound tissue of deep second-degree burn in rats. Acad Emerg Med. 21:383–391. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Shrivastava A, Kuzontkoski PM, Groopman JE and Prasad A: Cannabidiol induces programmed cell death in breast cancer cells by coordinating the cross-talk between apoptosis and autophagy. Mol Cancer Ther. 10:1161–1172. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Zeng R, He J, Peng J, Chen Y, Yi S, Zhao F and Cui G: The time-dependent autophagy protects against apoptosis with possible involvement of Sirt1 protein in multiple myeloma under nutrient depletion. Ann Hematol. 91:407–417. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Maiuri MC, Zalckvar E, Kimchi A and Kroemer G: Self-eating and self-killing: Crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 8:741–752. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 16 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
Xue X, Lv Y, Leng Y and Zhang Y: Protective effects of extracellular polymeric substances from Aphanizomenon flos‑aquae on neurotoxicity induced by local anesthetics. Exp Ther Med 16: 3011-3019, 2018
APA
Xue, X., Lv, Y., Leng, Y., & Zhang, Y. (2018). Protective effects of extracellular polymeric substances from Aphanizomenon flos‑aquae on neurotoxicity induced by local anesthetics. Experimental and Therapeutic Medicine, 16, 3011-3019. https://doi.org/10.3892/etm.2018.6540
MLA
Xue, X., Lv, Y., Leng, Y., Zhang, Y."Protective effects of extracellular polymeric substances from Aphanizomenon flos‑aquae on neurotoxicity induced by local anesthetics". Experimental and Therapeutic Medicine 16.4 (2018): 3011-3019.
Chicago
Xue, X., Lv, Y., Leng, Y., Zhang, Y."Protective effects of extracellular polymeric substances from Aphanizomenon flos‑aquae on neurotoxicity induced by local anesthetics". Experimental and Therapeutic Medicine 16, no. 4 (2018): 3011-3019. https://doi.org/10.3892/etm.2018.6540