α‑lipoic acid protects against cerebral ischemia/reperfusion‑induced injury in rats

  • Authors:
    • Houliang Deng
    • Xialin Zuo
    • Jingjing Zhang
    • Xiaoxia Liu
    • Li  Liu
    • Qian Xu
    • Zhuomin Wu
    • Aimin Ji
  • View Affiliations

  • Published online on: January 9, 2015     https://doi.org/10.3892/mmr.2015.3170
  • Pages: 3659-3665
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It is well established that the brain is sensitive to ischemia/reperfusion (I/R)‑induced injury. α‑lipoic acid (LA), a free radical scavenger and antioxidant, has a neuroprotective effect against cerebral I/R‑induced injury, however, the underlying mechanisms remain to be elucidated. Therefore, the present study was undertaken to evaluate whether LA was able to protect against cerebral I/R‑induced injury and to examine the potential mechanisms. The neuroprotective effects of LA were investigated in a rat model of transient focal ischemia induced by middle cerebral artery occlusion (MCAO) followed by reperfusion. Adult male Sprague‑Dawley rats were randomly assigned into the sham, cerebral I/R injury model and model plus LA groups. Cerebral I/R injury was induced by 90 min MCAO followed by reperfusion for 24 h. Cerebral infarct size was detected by 2,3,5‑triphenyltetrazolium chloride staining. Neurological deficit score (NDS), brain water content and oxidative parameters, including malondialdehyde (MDA), nitric oxide (NO), total antioxidant capacity (T‑AOC) and superoxide dismutase (SOD) were measured. The expression of cleaved caspase‑3, brain‑derived neurotrophic factor (BDNF), phosphatidylinositol‑4,5‑bisphosphate 3‑kinase (PI3K), p‑Akt and phosphorylated extracellular signal‑regulated kinase 1/2 (p‑ERK1/2) were also analyzed using western blotting. The present study demonstrated that pretreatment with LA significantly decreased the infarction size, brain water content and improved NDS. LA reversed the levels of oxidative parameters, including MDA, NO, T‑AOC and SOD to their normal state in rat brains following cerebral I/R. Furthermore, the expression of cleaved caspase‑3 markedly decreased and the expression of BDNF, PI3K, p‑Akt and p‑ERK1/2 significantly increased following administration of LA. On the basis of these findings, it was concluded that LA protected the brain from cerebral I/R damage by attenuation of oxidative stress and caspase‑dependent apoptosis. Furthermore, LA exerts its neuroprotective effects potentially through activation of the BDNF‑PI3K/Akt‑ERK1/2 pathway.

Introduction

Cerebral ischemic injury is one of the leading causes of human mortality and disability worldwide (1). Restoration of blood flow to the ischemic brain is often used to treat patients in clinical experiments. However, reperfusion itself also has the potential to produce additional injuries in the ischemic brain due to overproduction of reactive oxygen species (ROS). The potential pathological mechanisms of ischemia/reperfusion (I/R) injury include glutamate excitotoxicity, calcium overload, nitric oxide (NO) production, oxidative stress, inflammation and apoptosis, which eventually lead to cell death (2). Oxidative stress and apoptosis following cerebral I/R are the two major processes that induce neuronal injury (3,4). For this reason, multifunctional molecules with anti-oxidative and anti-apoptotic properties are ideal neuroprotective agents.

α-lipoic acid (LA), an endogenous short-chain fatty acid, is a cofactor for multiple mitochondrial dehydrogenase enzymes, including pyruvate dehydrogenase (5). It is an ROS scavenger, which is able to stimulate the insulin signaling pathway, chelate metal and regenerate endogenous natural antioxidants (6). In previous decades, using several different experimental models of I/R, studies have demonstrated the protective effects of LA against I/R-induced injury, including myocardial injury (7), peripheral nerve injury (8), testicular injury (9) and retinal injury (10). In addition, it has been reported that LA can activate phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt and extracellular signal-regulated kinase 1/2 (ERK1/2) pathways to induce protection against I/R injury in other organs (1113). However, to the best of our knowledge, few studies were reported to address whether LA has neuroprotective effects against cerebral I/R-induced injury and its potential mechanisms.

In the present study, the neuroprotective effects of LA in rats were investigated with 90 min middle cerebral artery occlusion (MCAO)/24 h reperfusion-induced injury. Furthermore, the hypothesis that the neuroprotective effect of LA is associated with a reduction in oxidative stress and inhibition of apoptosis though activation of brain-derived neurotrophic factor (BDNF), PI3K/Akt and ERK1/2 in rats was assessed. Therefore, the effect of LA on infarct size, neurological deficit score (NDS) and brain water content were investigated. In addition, to examine the mechanisms activated by LA, oxidative parameters, including malondialdehyde (MDA), NO and total antioxidant capacity (T-AOC), superoxide dismutase (SOD) and the expression of cleaved caspase-3, BDNF, PI3K, p-Akt and p-ERK1/2 proteins were measured in rat brains.

Materials and methods

Animals and drug administration

Adult male Sprague-Dawley rats, 10–12 weeks old, weighing 250–280 g, were purchased from the Animal Center of Southern Medical University (Guangzhou, China). All protocols were approved by the animal care committee of Southern Medical University and undertaken according to the guidelines for the Care and Use of Laboratory Animals of the National Institutes of Health (Bethesda, MD, USA). The rats were kept under constant laboratory conditions (20–25°C, 60±5% humidity) and a 12 h light/dark cycle. They were allowed free access to food and water up until 12 h prior to surgery at which point only water was available.

LA powder (Shanghai Modern Pharmaceutical Co., Ltd., Shanghai, China) was mixed with sterile saline. Subsequently, 1 M NaOH was added until the suspension dissolved. The pH was lowered to 7.4 using hydrochloric acid (14). Rats were administered with LA or saline via subcutaneous injection 2 h prior to MCAO (15).

Experimental groups

In preliminary experiments, different dosages of LA were administered to determine the optimal dosage. Rats were randomly assigned to the following groups (n=6): i) Sham group; ii) I/R group, wherein the animals received I/R and a vehicle treatment; iii) LA group, in which the rats received I/R and LA (50 mg/kg); iv) LA group, in which the rats received I/R and LA (70 mg/kg); v) LA group, in which the rats received I/R and LA (100 mg/kg). Following determining 100 mg/kg as the optimal dosage, rats were randomly divided into the following groups (n=9): i) Sham group; ii) I/R group, in which the rats received I/R and a vehicle treatment; iii) LA group, wherein the rats received I/R and LA (100 mg/kg).

Establishment of the MCAO model

Rats fasted overnight, but were allowed free access to water prior to the MCAO procedure. MCAO was induced as described previously (16). Briefly, rats were anesthetized with 10% chloral hydrate (3.5 ml/kg) by intraperitoneal injection. The skin and surrounding fur were disinfected with 75% ethyl alcohol. Following incision to the skin, the left common carotid artery (CCA) was exposed and carefully separated from the vagal nerves. The left external carotid artery (ECA) and the left internal carotid artery (ICA) were carefully isolated and the ECA was ligated. In addition, the CCA was temporally occluded with 3-0 silk thread. The left MCA was occluded for 90 min with a paraffin-coated nylon filament (17), which was introduced into the ICA for 18–20 mm until resistance was detected. After 90 min of occlusion, the ECA was permanently occluded. The silk thread occluding the CCA was removed for reperfusion for 24 h. A heating lamp was used to maintain body temperature at 37°C during surgery.

Determination of infarct size and neurological function

After 90 min of ischemia followed by 24 h of reperfusion animals were anesthetized with 10% chloral hydrate (3.5 ml/kg) and decapitated. The brains were quickly removed and frozen at −20°C for 20 min, dissected into 2 mm coronal slices and immediately incubated in 2% 2,3,5-triphenyltetrazolium chloride (TTC; Mbchem, Shanghai, China) at 37°C for 10 min as described previously (16). Following TTC staining, the normal brain tissue was dark red, whereas infarcted tissue was unstained. Following TTC staining, the tissues were then fixed in 4% paraformaldehyde (Mbchem) for 24 h and scanned with a digital camera (Canon, Inc., Tokyo, Japan). Infarct size was calculated using Image J software (National Institutes of Health, Bethesda, MD, USA) by an individual blinded to the identity of the experimental groups and the result was expressed as a percentage of infarct area to total brain area.

Neurological deficit evaluations were performed at the end of the experiment by an observer masked to the identity of experimental groups using the following criteria as previously described (16): 0, no neurologic deficit or normal function; 1, failure to extend right forepaw fully; 2, circling to right; 3, leaning to right; 4, absence of spontaneous motor activity. Therefore, a higher score was associated with poorer neurological function.

Measurement of brain water content

Brain water content was measured as described previously (18). Briefly, after 24 h of reperfusion animals were anesthetized with 10% chloral hydrate (3.5 ml/kg) and decapitated. The brains were rinsed with saline and separated into ischemic and non-ischemic hemispheres, then immediately weighed to gain the wet weight (WW). The brains were placed in an oven at 100°C for 24 h and weighed to obtain the dry weight (DW). The brain water content (%) was measured using the following formula: (WW − DW)/WW × 100%.

Measurement of the oxidative parameters MDA, NO, T-AOC and SOD

After 24 h of reperfusion, animals were anesthetized with 10% chloral hydrate (3.5 ml/kg) and decapitated. The rat brains were quickly removed and detached, then rinsed with cold saline. The brains were then homogenized in cold saline (1:10, wt/vol) and centrifuged at 3,000 × g for 10 min to prepare tissue homogenate. The levels of MDA, NO and the activities of T-AOC and SOD were determined using commercially available assay kits (Nanjing Jiancheng Bioengineering Institute, Nanjing, China) according to the manufacturer’s instructions.

Western blotting

Animals were anesthetized with 10% chloral hydrate (3.5 ml/kg) and decapitated 24 h after reperfusion. The rat brains were quickly removed and detached, then rinsed with cold saline. Tissue samples were lysed in ice-cold RIPA buffer (Nanjing Keygen Biotech, Co., Ltd., Nanjing, China) containing 1% phenylmethylsulfonyl fluoride, 1% phosphatase inhibitors and 0.1% protease inhibitor with a glass homogenizer on ice, repeated for 5 min and incubated on ice for 10 min, then centrifuged at 13,000 × g for 20 min at 4°C. The supernatant was aliquoted and stored at −80°C. Protein concentration was measured using the BCA kit (Pierce Biotechnology, Inc., Rockford, IL, USA). The protein (30 μg) was then separated on 8–15% polyacrylamide SDS gels and transferred onto a polyvinylidene difluoride membrane. The membranes were treated with blocking solution (5% non-fat dry milk or 5% bovine serum albumin in Tris-buffered saline with Tween® 20 and incubated at room temperature for 2 h. Following this, the membranes were incubated with a rabbit anti-rat polyclonal cleaved caspase-3 antibody (1:1,000; Cell Signaling Technology, Inc., Beverly, MA, USA), a rabbit anti-rat polyclonal BDNF antibody (1:1,000; Chemicon, Temecula, CA, USA), a rabbit anti-rat polyclonal PI3K antibody (1:1,000; Cell Signaling Technology, Inc.), a rabbit anti-rat polyclonal p-Akt antibody (1:2,000; Cell Signaling Technology, Inc.) and a rabbit anti-rat polyclonal p-ERK1/2 antibody (1:2,000; Cell Signaling Technology, Inc.) at 4°C overnight, followed by incubation with the corresponding goat anti-rabbit secondary antibodies (Wuhan Boster Bio-Engineering Co., Ltd., Wuhan, China) at room temperature for 1 h. GADPH was used as a control to ensure equal protein loading. The blots were visualized with ECL-Plus reagent (Pierce Biotechnology, Inc.) and the exposures were transferred to radiographic films. The radiographic films were scanned by a cannon scanner (Canon, Inc.) and analyzed using Image J software (National Institutes of Health).

Statistical analysis

All data were analyzed using SPSS 13.0 software (SPSS, Inc., Chicago, IL, USA) and are expressed as the mean ± standard deviation. Statistical analysis was performed by one-way analysis of variance and P<0.05 was considered to indicate a statistically significant difference.

Results

LA attenuates infarct size

The experimental procedure is shown in Fig. 1A. In order to investigate the effect of LA on infarct size, the brain infarct area was analyzed by TCC staining after 24 h of reperfusion. There was no infarct area in the sham rat brain (Fig. 1B). Pretreatment with 100 mg/kg LA significantly reduced total infarct size by 51.3% compared with ischemic rats with vehicle treatment from 26.9±1.7% to 13.8±1.4% (P<0.05), while 50 mg/kg and 70 mg/kg did not reveal a significant effect (26.9±2.1% and 24.1±2.3%, respectively; Fig. 1C). Therefore, 100 mg/kg LA was selected as the optimum dosage for further investigation.

LA improves neurological function

As shown in Fig. 2A, 24 h after reperfusion, the neurological score was 3.17±0.41 in the I/R group, while this significantly decreased following treatment with LA (100 mg/kg) to 1.83±0.41 (P<0.05), however, 50 and 70 mg/kg LA did not have a significant effect (3.0±0.63 and 2.5±0.55, respectively).

LA ameliorates brain edema

Brain water content was quantified using the dry-wet weight method and the result is shown in Fig. 2B. The brain water content in the ischemic area of the I/R group was significantly higher than that of the sham group. Pretreatment of rats with 100 mg/kg LA significantly reduced brain edema in the rats that underwent I/R (82.1±0.5% vs. 80.3±0.6%; P<0.05).

LA improves the oxidative parameters MDA, NO, T-AOC and SOD

As shown in Fig. 3, the content of MDA and NO significantly increased in the I/R group (P<0.05) compared with that in the sham group and decreased in the LA-pretreated group (P<0.05). The activities of T-AOC and SOD were significantly higher in the LA-treated group (P<0.05) than in the I/R group.

LA suppresses caspase-independent apoptosis

Cleaved caspase-3, activated from caspase-3, has been identified as a key mediator of apoptosis and is considered to be one of the final steps in cell apoptosis (19,20). As shown in Fig. 4, LA significantly suppressed the level of cleaved caspase-3 compared with the I/R group (P<0.05).

LA upregulates the expression of BDNF, PI3K, p-Akt and p-ERK1/2

The expression of BDNF, PI3K, p-Akt and p-ERK1/2 were examined using western blotting (Fig. 5A). As shown in Fig. 5B, compared with the I/R group, LA significantly increased the level of BDNF and p-Akt (P<0.05). The results demonstrated that the levels of PI3K and p-ERK1/2 were significantly downregulated following cerebral I/R, however, pretreatment with LA was able to reverse this effect and increase the expression of these proteins (Fig. 5C–E; P<0.05).

Discussion

The present study demonstrated that pretreatment with LA has protective effects against neuronal injury caused by cerebral I/R in rats. Therefore, as a preventive agent, LA is beneficial to cerebral I/R-induced injury. This finding is consistent with previous studies on the beneficial effects of LA against cerebral ischemia (21,22).

The results demonstrated that the optimal dose of LA was 100 mg/kg. This was not in accordance with a previous study in which only 5 mg/kg LA had the capacity to protect against cerebral I/R-induced injury (22). This difference may be due to the fact that the time period of cerebral I/R is longer in the present study (30 min of ischemia followed by 5.5 h of reperfusion vs. 90 min of ischemia followed by 24 h of reperfusion), leading to a more severe neuronal injury. Therefore, a higher dose of LA (100 mg/kg) is required to protect against cerebral I/R-induced injury.

Oxidative stress, is the main pathological process of cerebral I/R injury due to the balance between the formation and elimination of ROS, including superoxide, hydrogen peroxide and peroxynitrite (23). The increased concentration of ROS can cause cellular damage and subsequent cell death since ROS may oxidize crucial cellular components, including lipids, proteins and DNA (24). The levels of MDA and NO increased indicating that severe oxidative damage was caused by I/R (25). It has been reported that LA has the capability to scavenge MDA and NO within brain tissue (26). In the present study, pretreatment with LA decreased the levels of MDA and NO in rat brains. The endogenous antioxidant enzymes, including SOD and glutathione (GSH) peroxidase and low-molecular weight ROS scavengers, including GSH, are critical in attenuating the injury induced by I/R (25,27). In the present study, LA enhanced the activities of T-AOC and SOD in rat brains. These data provided further evidence that LA could inhibit I/R-induced oxidative stress, which may contribute to the attenuation of I/R-induced injury.

Apoptosis has a critical pathogenic role in I/R injury. Cleaved caspase-3, which is activated from caspase-3, is an important molecule involved in apoptosis in I/R injury (28). It is reported that activation of caspase-3 at the final execution phase of apoptosis leads to DNA fragmentation and cell death (29). It was observed that the level of cleaved caspase-3 markedly increased in the brain following cerebral I/R indicating extensive apoptosis caused by I/R. The present data indicated that pretreatment with LA reversed the increased level of cleaved caspase-3. The result suggested that LA had anti-apoptotic capacity, which was beneficial to cerebral I/R-induced injury.

BDNF is a member of the neurotrophin family and is important in neuronal survival, differentiation, axon growth, dendritic spine development and synaptic plasticity. When a neuron is damaged, BDNF performs a variety of biological effects, including preventing cell death in damaged neurons, improving the pathological state of neurons and promoting the regeneration of damaged neurons (30). In the case of cerebral I/R, BDNF may protect neurons (3133). BDNF can activate the TrkB receptor, leading to the activation of several intracellular signaling pathways, including PI3K/Akt and RAS/ERK pathways (34). The PI3K/Akt and ERK1/2 signaling pathways are important in regulating cell growth, proliferation and apoptosis (3537). Numerous studies have reported that the activation of the PI3K/Akt and ERK1/2 pathways are markedly associated with protection from cerebral I/R injury (38,39). Akt, which is a direct downstream target of PI3K when phosphorylated can suppress apoptosis by activation of anti-apoptotic substrates, including B-cell lymphoma 2 family members and inhibition of pro-apoptotic substrates, including cytochrome c (40). When cells suffer ischemic insults, the phosphorylation of Akt may have a protective effect (41). ERK1/2 is a member of the mitogen-activated protein kinase superfamily that can mediate cell proliferation and apoptosis (42). The activation of ERK1/2 and subsequent downstream signaling targets, is important in ROS-mediated cell death (43). In the present study, it was demonstrated that LA significantly increased the level of BDNF. The results revealed that expression of PI3K and p-Akt was significantly downregulated in brains following cerebral I/R, while LA could reverse this situation and increase the levels of PI3K and p-Akt. In addition, the phosphorylation of ERK1/2 increased with administration of LA. The results suggested that LA pretreatment provided protective effects against cerebral I/R-induced injury possibly by promoting the BDNF-PI3K/Akt-ERK1/2 signaling pathway.

In conclusion, the present study demonstrated that LA could afford protection against cerebral I/R-induced injury by attenuation of oxidative stress and caspase-dependent apoptosis. Furthermore, the results suggest that administration of LA induces a neuroprotective effect in association with the activation of the BDNF-PI3K/Akt-ERK1/2 signaling pathway.

Acknowledgements

This study was supported by the National Science Foundation of China (grant no. 81370449), the Industry, Education and Research of Guangdong Province (grant no. 2011B090400015) and the Science and Technology Development project of Guangzhou (grant no. 2010UI-E00531-7).

References

1 

Tu Q, Wang R, Ding B, Zhong W and Cao H: Protective and antioxidant effect of Danshen polysaccharides on cerebral ischemia/reperfusion injury in rats. Int J Biol Macromol. 60:268–271. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Sahota P and Savitz SI: Investigational therapies for ischemic stroke: neuroprotection and neurorecovery. Neurotherapeutics. 8:434–451. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Mattson MP, Duan W, Pedersen WA and Culmsee C: Neurodegenerative disorders and ischemic brain diseases. Apoptosis. 6:69–81. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Zhao ZQ: Oxidative stress-elicited myocardial apoptosis during reperfusion. Curr Opin Pharmacol. 4:159–165. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Perham RN: Swinging arms and swinging domains in multifunctional enzymes: catalytic machines for multistep reactions. Annu Rev Biochem. 69:961–1004. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Smith AR, Shenvi SV, Widlansky M, Suh JH and Hagen TM: Lipoic acid as a potential therapy for chronic diseases associated with oxidative stress. Curr Med Chem. 11:1135–1146. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Wang X, Yu Y, Ji L, Liang X, Zhang T and Hai CX: Alpha-lipoic acid protects against myocardial ischemia/reperfusion injury via multiple target effects. Food Chem Toxicol. 49:2750–2757. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Mitsui Y, Schmelzer JD, Zollman PJ, Mitsui M, Tritschler HJ and Low PA: Alpha-lipoic acid provides neuroprotection from ischemia-reperfusion injury of peripheral nerve. J Neurol Sci. 163:11–16. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Ozbal S, Ergur BU, Erbil G, Tekmen I, Bagrıyanık A and Cavdar Z: The effects of α-lipoic acid against testicular ischemia-reperfusion injury in rats. ScientificWorldJournal. 2012:4892482012. View Article : Google Scholar

10 

Chidlow G, Schmidt KG, Wood JP, Melena J and Osborne NN: Alpha-lipoic acid protects the retina against ischemia-reperfusion. Neuropharmacology. 43:1015–1025. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Deng C, Sun Z, Tong G, et al: α-Lipoic acid reduces infarct size and preserves cardiac function in rat myocardial ischemia/reperfusion injury through activation of PI3K/Akt/Nrf2 pathway. PLoS One. 8:e583712013. View Article : Google Scholar

12 

Oh SK, Yun KH, Yoo NJ, et al: Cardioprotective effects of alpha-lipoic acid on myocardial reperfusion injury: suppression of reactive oxygen species generation and activation of mitogen-activated protein kinase. Korean Circ J. 39:359–366. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Xie R, Li X, Ling Y, et al: Alpha-lipoic acid pre- and post-treatments provide protection against in vitro ischemia-reperfusion injury in cerebral endothelial cells via Akt/mTOR signaling. Brain Res. 1482:81–90. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Cameron NE, Cotter MA, Horrobin DH and Tritschler HJ: Effects of alpha-lipoic acid on neurovascular function in diabetic rats: interaction with essential fatty acids. Diabetologia. 41:390–399. 1998. View Article : Google Scholar : PubMed/NCBI

15 

Wolz P and Krieglstein J: Neuroprotective effects of alpha-lipoic acid and its enantiomers demonstrated in rodent models of focal cerebral ischemia. Neuropharmacology. 35:369–375. 1996. View Article : Google Scholar : PubMed/NCBI

16 

Longa EZ, Weinstein PR, Carlson S and Cummins R: Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke. 20:84–91. 1989. View Article : Google Scholar : PubMed/NCBI

17 

Zuo XL, Wu P and Ji AM: Nylon filament coated with paraffin for intraluminal permanent middle cerebral artery occlusion in rats. Neurosci Lett. 519:42–46. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Lan R, Xiang J, Wang GH, et al: Xiao-Xu-Ming decoction protects against blood-brain barrier disruption and neurological injury induced by cerebral ischemia and reperfusion in rats. Evid Based Complement Alternat Med. 2013:6297822013.PubMed/NCBI

19 

Namura S, Zhu J, Fink K, et al: Activation and cleavage of caspase-3 in apoptosis induced by experimental cerebral ischemia. J Neurosci. 18:3659–3668. 1998.PubMed/NCBI

20 

D’Amelio M, Cavallucci V and Cecconi F: Neuronal caspase-3 signaling: not only cell death. Cell Death Differ. 17:1104–1114. 2010. View Article : Google Scholar

21 

Clark WM, Rinker LG, Lessov NS, Lowery SL and Cipolla MJ: Efficacy of antioxidant therapies in transient focal ischemia in mice. Stroke. 32:1000–1004. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Connell BJ, Saleh M, Khan BV and Saleh TM: Lipoic acid protects against reperfusion injury in the early stages of cerebral ischemia. Brain Res. 1375:128–136. 2011. View Article : Google Scholar

23 

Yabuki Y and Fukunaga K: Oral administration of glutathione improves memory deficits following transient brain ischemia by reducing brain oxidative stress. Neuroscience. 250:394–407. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Zhou XQ, Zeng XN, Kong H and Sun XL: Neuroprotective effects of berberine on stroke models in vitro and in vivo. Neurosci Lett. 447:31–36. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Wang PR, Wang JS, Zhang C, Song XF, Tian N and Kong LY: Huang-Lian-Jie-Du-Decotion induced protective autophagy against the injury of cerebral ischemia/reperfusion via MAPK-mTOR signaling pathway. J Ethnopharmacol. 149:270–280. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Packer L, Tritschler HJ and Wessel K: Neuroprotection by the metabolic antioxidant alpha-lipoic acid. Free Radic Biol Med. 22:359–378. 1997. View Article : Google Scholar : PubMed/NCBI

27 

Yang G, Chan PH, Chen J, et al: Human copper-zinc superoxide dismutase transgenic mice are highly resistant to reperfusion injury after focal cerebral ischemia. Stroke. 25:165–170. 1994. View Article : Google Scholar : PubMed/NCBI

28 

Haylor JL, Harris KP, Nicholson ML, Waller HL, Huang Q and Yang B: Atorvastatin improving renal ischemia reperfusion injury via direct inhibition of active caspase-3 in rats. Exp Biol Med (Maywood). 236:755–763. 2011. View Article : Google Scholar

29 

Thornberry NA and Lazebnik Y: Caspases: enemies within. Science. 281:1312–1316. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Binder DK and Scharfman HE: Brain-derived neurotrophic factor. Growth Factors. 22:123–131. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Ferrer I, Krupinski J, Goutan E, Marti E, Ambrosio S and Arenas E: Brain-derived neurotrophic factor reduces cortical cell death by ischemia after middle cerebral artery occlusion in the rat. Acta Neuropathol. 101:229–238. 2001.PubMed/NCBI

32 

Muller HD, Hanumanthiah KM, Diederich K, Schwab S, Schabitz WR and Sommer C: Brain-derived neurotrophic factor but not forced arm use improves long-term outcome after photothrombotic stroke and transiently upregulates binding densities of excitatory glutamate receptors in the rat brain. Stroke. 39:1012–1021. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Ploughman M, Windle V, MacLellan CL, White N, Doré JJ and Corbett D: Brain-derived neurotrophic factor contributes to recovery of skilled reaching after focal ischemia in rats. Stroke. 40:1490–1495. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Patapoutian A and Reichardt LF: Trk receptors: mediators of neurotrophin action. Curr Opin Neurobiol. 11:272–280. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Burgering BM and Coffer PJ: Protein kinase B (c-Akt) in phosphatidylinositol-3-OH kinase signal transduction. Nature. 376:599–602. 1995. View Article : Google Scholar : PubMed/NCBI

36 

Franke TF, Yang SI, Chan TO, et al: The protein kinase encoded by the Akt proto-oncogene is a target of the PDGF-activated phosphatidylinositol 3-kinase. Cell. 81:727–736. 1995. View Article : Google Scholar : PubMed/NCBI

37 

Xia Z, Dickens M, Raingeaud J, Davis RJ and Greenberg ME: Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science. 270:1326–1331. 1995. View Article : Google Scholar : PubMed/NCBI

38 

Arslan F, Lai RC, Smeets MB, et al: Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 10:301–312. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Zhou L and Miller CA: Mitogen-activated protein kinase signaling, oxygen sensors and hypoxic induction of neurogenesis. Neurodegener Dis. 3:50–55. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Franke TF, Hornik CP, Segev L, Shostak GA and Sugimoto C: PI3K/Akt and apoptosis: size matters. Oncogene. 22:8983–8998. 2003. View Article : Google Scholar : PubMed/NCBI

41 

Liu H, Liu X, Wei X, et al: Losartan, an angiotensin II type 1 receptor blocker, ameliorates cerebral ischemia-reperfusion injury via PI3K/Akt-mediated eNOS phosphorylation. Brain Res Bull. 89:65–70. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Mebratu Y and Tesfaigzi Y: How ERK1/2 activation controls cell proliferation and cell death: Is subcellular localization the answer? Cell Cycle. 8:1168–1175. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Dong J, Ramachandiran S, Tikoo K, Jia Z, Lau SS and Monks TJ: EGFR-independent activation of p38 MAPK and EGFR-dependent activation of ERK1/2 are required for ROS-induced renal cell death. Am J Physiol Renal Physiol. 287:F1049–F1058. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 11 Issue 5

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
Deng H, Zuo X, Zhang J, Liu X, Liu L, Xu Q, Wu Z and Ji A: α‑lipoic acid protects against cerebral ischemia/reperfusion‑induced injury in rats. Mol Med Rep 11: 3659-3665, 2015
APA
Deng, H., Zuo, X., Zhang, J., Liu, X., Liu, L., Xu, Q. ... Ji, A. (2015). α‑lipoic acid protects against cerebral ischemia/reperfusion‑induced injury in rats. Molecular Medicine Reports, 11, 3659-3665. https://doi.org/10.3892/mmr.2015.3170
MLA
Deng, H., Zuo, X., Zhang, J., Liu, X., Liu, L., Xu, Q., Wu, Z., Ji, A."α‑lipoic acid protects against cerebral ischemia/reperfusion‑induced injury in rats". Molecular Medicine Reports 11.5 (2015): 3659-3665.
Chicago
Deng, H., Zuo, X., Zhang, J., Liu, X., Liu, L., Xu, Q., Wu, Z., Ji, A."α‑lipoic acid protects against cerebral ischemia/reperfusion‑induced injury in rats". Molecular Medicine Reports 11, no. 5 (2015): 3659-3665. https://doi.org/10.3892/mmr.2015.3170