Inhibition of cerebral ischemia/reperfusion injuryinduced apoptosis: nicotiflorin and JAK2/STAT3 pathway

2017-03-30 04:44GuangqiangHuXiDuYongjieLiXiaoqingGaoBiqiongChenLuYu

Guang-qiang Hu, Xi Du, Yong-jie Li, Xiao-qing Gao, Bi-qiong Chen, Lu Yu,

1 Department of Anatomy, Southwest Medical University, Luzhou, Sichuan Province, China

2 Department of Chemistry, Southwest Medical University, Luzhou, Sichuan Province, China

3 Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan Province, China

4 Department of Anatomy and Neurobiology, Southwest Medical University, Luzhou, Sichuan Province, China

Inhibition of cerebral ischemia/reperfusion injuryinduced apoptosis: nicotiflorin and JAK2/STAT3 pathway

Guang-qiang Hu1,*, Xi Du2, Yong-jie Li3, Xiao-qing Gao4, Bi-qiong Chen2, Lu Yu2,*

1 Department of Anatomy, Southwest Medical University, Luzhou, Sichuan Province, China

2 Department of Chemistry, Southwest Medical University, Luzhou, Sichuan Province, China

3 Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan Province, China

4 Department of Anatomy and Neurobiology, Southwest Medical University, Luzhou, Sichuan Province, China

How to cite this article:Hu GQ, Du X, Li YJ, Gao XQ, Chen BQ, Yu L (2017) Inhibition of cerebral ischemia/reperfusion injury-induced apoptosis: nicoti fl orin and JAK2/STAT3 pathway. Neural Regen Res 12(1):96-102.

Open access statement:is is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

Graphical Abstract

Nicoti fl orin protects against cerebral ischemia/reperfusion injury-induced apoptosisviathe JAK2/SA3 pathway

orcid: 0000-0002-8873-9138 (Lu Yu)

Nicoti fl orin is a fl avonoid extracted from Carthamus tinctorius. Previous studies have shown its cerebral protective e ff ect, but the mecha‐nism is unde fi ned. In this study, we aimed to determine whether nicoti fl orin protects against cerebral ischemia/reperfusion injury‐induced apoptosis through the JAK2/STAT3 pathway.e cerebral ischemia/reperfusion injury model was established by middle cerebral artery occlusion/reperfusion. Nicoti fl orin (10 mg/kg) was administered by tail vein injection. Cell apoptosis in the ischemic cerebral cortex was examined by hematoxylin‐eosin staining and terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Bcl‐2 and Bax expres‐sion levels in ischemic cerebral cortex were examined by immunohistochemial staining. Additionally, p‐JAK2, p‐STAT3, Bcl‐2, Bax, and caspase‐3 levels in ischemic cerebral cortex were examined by western blot assay. Nicoti fl orin altered the shape and structure of injured neurons, decreased the number of apoptotic cells, down‐regulates expression of p‐JAK2, p‐STAT3, caspase‐3, and Bax, decreased Bax immunoredactivity, and increased Bcl‐2 protein expression and immunoreactivity.ese results suggest that nicoti fl orin protects against cerebral ischemia/reperfusion injury‐induced apoptosisviathe JAK2/STAT3 pathway.

nerve regeneration; brain injury; nicoti fl orin; ischemic stroke; cerebral ischemia/reperfusion injury; treatment; cell apoptosis; terminal deoxynucleotidyl transferase dUTP nick end labeling; JAK2/STAT3 pathway; Bcl-2; Bax; caspase-3; neural regeneration

Introduction

Stroke is a serious leading cause of death that causes fi nan‐cial burden, especially in low‐income and middle‐income countries (Feigin et al., 2014; Levine et al., 2015; Banerjee and Das, 2016). Ischemic stroke accounts for 75% of all stroke patients (Zevallos et al., 2015). Within a certain time window, thrombolysis is thought to be the most effective treatment method, but many people cannot arrive at hospital within 4.5—6.0 hours, therefore systemic recombinant tissue plasminogen activator is limited (Zaidat et al., 2012; Akbik et al., 2016). Due to developments in pathophysiological stroke research, di ff erent mechanisms provide varied treatment op‐portunities (Lo et al., 2003; Hachinski et al., 2010; Liu et al., 2014).

In recent decades, studies have shown that significant blood fl ow reductions within the ischemic core accompany irreversible nerve cell necrosis, while programmed cell death (namely apoptosis) appears within the ischemic penumbra, and is reversible until a few hours aer cerebral ischemia (Xu and Zhang, 2011; Ghosh et al., 2012; Kalogeris et al., 2012). Hence, saving apoptotic cells is an important strategy in stroke treatment.

Cerebral ischemia/reperfusion (I/R) injury triggers mul‐tiple cell apoptotic pathways (Li et al., 1997; Polster and Fiskum, 2004; Wang et al., 2013; Feng et al., 2016). Indeed, there is overwhelming evidence that ischemia‐induced oxi‐dative stress and in fl ammation are principally responsible for subsequent cell death by necrotic or apoptotic mechanisms (Nita et al., 2001; Jin et al., 2013). Reactive oxygen species regulate cell survival/death by activating various cell signal‐ing pathways, such as p38, c‐Jun N‐terminal kinases, nuclear factor‐kappa B, and Janus kinase/signal transducers and acti‐vators of transcription JAK/STAT (Nakka et al., 2008; Wang et al., 2014; Hou et al., 2016). Studies show that JAK2/STAT3 activation contributes to cell apoptosis following transient focal cerebral ischemia (Satriotomo et al., 2006; Xie et al., 2007).

Traditional Chinese medicines are e ff ective and have less clinical side‐e ff ects for cerebral ischemia patients, but their mechanisms and targets need further investigation (Murphy, 2003; Sun et al., 2015). Nicoti fl orin, kaempferol‐3‐O‐rutino‐side, a fl avonoid glycoside extracted from Carthamus tinc‐torius, has shown protective effects against brain injury in a multi‐infarct dementia model (Xie et al., 2007). Similarly, other studies have shown that nicoti fl orin improves ischemic brain damage after transient focal cerebral ischemia (Li et al., 2006; Huang et al., 2007). Nicoti fl orin is neuroprotective against hypoxia‐, glutamate‐ or oxidative stress‐induced ret‐inal ganglion cell death (Nakayama et al., 2011). In addition, we have previously shown protective effects of nicotiflorin in brain injury and neuroin fl ammation by inhibiting STAT3 activation (Yu et al., 2013). As already noted, JAK2/STAT3 activation contributes to cell apoptosis following transient focal cerebral ischemia. However, it remains poorly under‐stood whether nicoti fl orin protects against cerebral I/R‐in‐duced cell apoptosis through the JAK2/STAT3 pathway. Ac‐cordingly, this is the focus of our present study, and indeed has not previously been investiagted.us, we examined the anti‐apoptopic effect and underlying nicotiflorin signaling pathway in rats following transient ischemia induced by I/R.

Materials and Methods

Animals

Twenty‐four male specific‐pathogen‐free Sprague‐Dawley rats weighing 260—310 g and aged 13—15 weeks were provid‐ed by the Experimental Animal Center, Southwest Medical University, China (license No. SCXK(Chuan)2013‐17).e rats were equally and randomly allocated to three groups: sham, I/R, and nicotiflorin. Rats were maintained under standardized temperature and humidity with a 12‐hour light/dark cycle, and free access to food and water. All pro‐cedures were performed in accordance with China Animal Welfare Legislation. Protocols were approved by the Ethics Committee of Southwest Medical University, China.

Surgical operation

Transient middle cerebral artery occlusion (MCAO) was performed on the right side using a nylon fi lament, as pre‐viously described (Longa et al., 1989). Briefly, rats were anesthetized with 10% chloral hydrate (350 mg/kg, intraper‐itoneally). The internal carotid artery and external carotid artery were carefully detached and a prepared segment of 4‐0 monofilament fiber was inserted from the external carotid artery into the internal carotid artery to block the origin of the middle cerebral artery. The sham operation involved a similar surgical procedure except for MCAO. Aer 2 hours of ischemia, the fiber was gently removed to enable reper‐fusion of the middle cerebral artery. Body temperature was maintained at 37°C throughout the operation. Rats with the following symptoms were assumed to be successful models: failure to fully extend left forepaw, circling to the left, or falling to the le. Aer 24 hours of reperfusion, rats were ex‐ecuted.

Drug administration

Nicotiflorin (Shanghai Winherb Medical Technology Co., Ltd., Shanghai, China) was dissolved in 25% polyethylene glycol 400. Nicotiflorin (10.0 mg/kg) was administered by tail vein injection to the nicoti fl orin group at the beginning of reperfusion. Vehicle was administered to the I/R and sham groups in the same manner. Rats were executed aer 24 hours of nicoti fl orin treatment.

Brain tissue extraction

Brain tissue was removed after reperfusion for 24 hours, then fi xed, embedded in para ffi n blocks, and cut into 5 μm‐thick coronal sections (0.26—0.51 mm anterior to bregma) for conventional hematoxylin‐eosin staining, terminal de‐oxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, and immunohistochemical staining.

Hematoxylin-eosin staining

Figure 1 Representative hematoxylin-eosin (HE) stained micrographs of the cortical ischemic penumbra.

Figure 2 E ff ect of nicoti fl orin on cell apoptosis in the I/R model.

Figure 3 Nicoti fl orin e ff ect on expression of JAK2 and SA3 phosphorylation in the ischemic cerebral cortex of I/R rats.

Figure 4 Nicoti fl orin e ff ect on Bcl-2 and Bax immunreactivity in the ischemic cerebral cortex of I/R rats (immunohistochemical staining).

Figure 5 Nicoti fl orin e ff ect on Bcl-2, Bax, and caspase-3 expression in the ischemic cerebral cortex of I/R rats (western blot assay).

Briefly, slices were deparaffinized, hydrated in water, then stained with hematoxylin for 15 minutes, and washed in running tap water for 20 minutes. Next, slices were counter‐stained with eosin for 2 minutes, dehydrated in 95% absolute alcohol until excess eosin was removed, permeabilized in xylene, and mounted. Pathological changes were observed under a light microscope.

TUNEL staining was used to detect fragmented nuclear DNA during apoptosis (ArunaDevi et al., 2010; Bahmani et al., 2011), according to standard protocols for the TUNEL assay kit (Boster, Wuhan, China). Brie fl y, slices were depa‐raffinized, rehydrated, and then incubated in proteinase K for 15 minutes to digest DNA. Aer washing in a Tris bu ff er, sections were incubated in labeling buffer mixed with TdT and digoxigenin (DIG)‐d‐UTP at 37°C for 2 hours. Block‐ing reagent was added at room temperature for 30 minutes. Next, sections were incubated in anti‐DIG‐biotin for 30 min‐utes, with Strept Avidin Biotin Complex (SABC)‐FITC used for final detection. Stained cells were counted from three di ff erent views per section using a fl uorescence microscope (AMG EVos FL Microscopy, Seattle, WA, USA).e average count number was used for quanti fi cation and comparison between groups.

Immunohistochemical staining

Immunohistochemistry of coronal sections was performed as described previously (Yu et al., 2013). Rabbit anti‐B‐cell lymphoma 2 (Bcl‐2) associated X (Bax) polyclonal antibody (Boster; 1:200), and rabbit anti‐Bcl‐2 polyclonal antibody (Boster; 1:100) were used. Briefly, sections were dewaxed, rinsed with 3% H2O2for 20 minutes, and incubated in 5% bovine serum albumin for 30 minutes to block nonspeci fi c binding. Sections were then separately incubated in Bax and Bcl‐2 antibodies overnight at 4°C. Next, ready‐to‐use goat anti‐rabbit IgG secondary antibody (Boster) was incubated at 37°C for 1 hour. Sections were then stained with 3,3′‐di‐aminobenzidine and counterstained with hematoxylin. For each antibody staining, immunoreactive cells per 1 mm2of the cortex were counted using an optical microscope (Leica DM750, Solms, Germany) (Chu et al., 2006). The average count number was used for quanti fi cation and comparison between groups.

Western blot assay

Statistical analysis

All data are presented as the mean ± SEM, and were ana‐lyzed using SPSS 16.0 software (SPSS, Chicago, IL, USA). Di ff erences among groups were analyzed by one‐way analy‐sis of variance followed bypost hocTukey test. In all analyses, values ofP< 0.05 were considered statistically signi fi cant.

Results

Nicoti fl orin inhibited neuronal pathological changes

Hematoxylin‐eosin staining showed that normal neurons in the sham group exhibited regulatory round and bright blue nuclei (Figure 1). While in the I/R group, neurons showed apparent disorder, and part of them presented apoptosis fea‐tures: nuclear chromatin pyknosis, side scatter, or fracture. Nicotiflorin reduced pathological neuronal injury and the number of dead neurons (necrosis and apoptosis) induced by cerebral I/R.

Nicoti fl orin decreased cerebral I/R-induced cell apoptosis

In the sham group, there were only a few TUNEL‐positive cells (bright green fl uorescence) in the cerebral cortex (Figure 2A). Further, in the I/R group, the TUNEL‐positive cell number markedly increased after 2 hours of MCAO and 24 hours of reperfusion. In contrast, compared with the I/R group, TUNEL‐positive cells signi fi cantly decreased aer nicoti fl orin treatment (P< 0.01; Figure 2B).

Nicoti fl orin inhibited JAK2 and SA3 phosphorylation

Phosphorylation of JAK2 and STAT3 is thought to play an important role in apoptosis regulation.erefore, both pro‐teins were assessed by western blot assay. Activited pJAK2 and pSTAT3 both increased aer ischemic injury in the I/R group compared with the sham group (P< 0.01). However, compared with the I/R group, nicoti fl orin inhibited pJAK2 and pSTAT3 activation (Figure 3A, B).

Nicoti fl orin altered caspase-3, Bcl-2, and Bax immunoreactivity

Bcl‐2 family members play important regulatory roles in cerebral I/R injury‐induced apoptosis (Hardwick et al., 2012; Kalogeris et al., 2012; Kvansakul and Hinds, 2013; Troy and Jean, 2015). Therefore, we examined changes in Bcl‐2 and Bax immunoreactivity. Immunohistochemical staining showed increased Bax immunoreactivity and lower Bcl‐2 immunoreactivity in the cerebral cortex of the I/R group (Figure 4A). In contrast, nicotiflorin significantly reduced Bax immunoreactivity, but increased Bcl‐2 immunoreactivi‐ty (P< 0.05 orP< 0.01; Figure 4B).

To further examine this anti‐apoptotic e ff ect of nicoti fl o‐rin, caspase‐3, Bax, and Bcl‐2 expression was examined by western blot assay in brain tissue aer ischemia. Bax and Bcl‐2 con fi rmed the changes observed by immunohistochemis‐try. Following transient MCAO and 24 hours of reperfusion in the I/R group, increased Bax and caspase‐3 expression, and relatively less Bcl‐2 expression was detected. However, in the nicoti fl orin group, decreased Bax and caspase‐3, and increased Bcl‐2 expression was observed (P< 0.05 orP< 0.01,n= 3; Figure 5).

Discussion

Neuronal damage includes both apoptosis and necrosis (Charriaut‐Marlangue et al., 1996; Sugawara et al., 2004; Poon et al., 2010). Cerebral ischemia leads to irreversible neuronal injury in the ischemic core area within minutes of onset (Lavrik et al., 2005; Uyttenboogaart et al., 2009; Murray et al., 2010; Jiang et al., 2016). While in the infarct penumbra area, reversible apoptosis is the main cell death mechanism, and is closely related to the final infarct area (Olsen et al., 1983; Dirnagl et al., 1999; Lo, 2008; Popp et al., 2009; Deng et al., 2016). Promisingly, reversible apoptosis provides multiple opportunities for therapeutic intervention in ischemic stroke (Nakka et al., 2008; Broughton et al., 2009). Consequently, we examined the e ff ect of nicoti fl orin on neuronal apoptosis in I/R rats. TUNEL staining was chosen as an indicator of apoptotic neurons. Our results show significantly decreased apoptosis in the nicotiflorin group. In addition, hematoxy‐lin‐eosin staining showed that nicoti fl orin reduces pathologi‐cal neuronal injury and the number of dead neurons induced by cerebral I/R.is is the fi rst time the e ff ect of nicoti fl orin on neuronal apoptosis has been studied.

Apoptotic signal transduction mechanisms play a vital role (Chong et al., 2010; Wang et al., 2015).e JAK/STAT pathway is a critical human disease target (Ghoreschi et al., 2009; Babon et al., 2014; Tao et al., 2015; Chai et al., 2016). Within this fam‐ily, JAK2/STAT3 is closely associated with ischemia‐induced neuronal apoptosis and cancer cell apoptosis (Satriotomo et al., 2006; Xie et al., 2007; Du et al., 2012; Zhang et al., 2015). AG490, a potent JAK2 phosphorylation inhibitor, blocks post‐ischemic JAK2 and STAT3 phosphorylation, and signi fi‐cantly decreases cerebral infarction, cell apoptosis, and neu‐rological dysfunction (Satriotomo et al., 2006). We previously found that nicoti fl orin is protective against brain injury and neuroin fl ammation by inhibiting STAT3 activation (Yu et al., 2013). Whether nicoti fl orin prevents apoptosis and in fl uences ischemic outcomeviathe JAK2/STAT3 pathway is not known. In the present study, we examined JAK2 and STAT3 phos‐phorylation in post‐ischemic rat brain by western blot assay, in accordance with previous studies (Gorina et al., 2005; Satri‐otomo et al., 2006). Consistent with previous AG490 fi ndings, we found nicoti fl orin blocked post‐ischemic JAK2 and STAT3 phosphorylation.erefore, our results suggest that the protec‐tive e ff ect of nicoti fl orin against brain damage is related to the JAK2/STAT3 pathway.

To further understand the downstream anti‐apoptotic mechanism of nicotiflorin in cerebral I/R injury, we also examined Bax, Bcl‐2, and caspase‐3 expression. Undoubt‐edly, both extrinsic and intrinsic pathways are responsible for activation of apoptosis (Gorina et al., 2005; Venderova and Park, 2012). Internally, cerebral ischemia increases in‐tracellular calcium levels, activates calpains, and mediates cleavage of Bid to truncated Bid. Truncated Bid interacts with apoptotic proteins, such as Bax, which generally maintains balance with the anti‐apoptotic protein, Bcl‐2 (Hassan et al., 2014). Mitochondrial transition pores areopened and release cytochrome c or apoptosis‐inducing factor to activate caspase‐9, and subsequently caspase‐3, which leads to nuclear DNA damage and apoptosis (Mattson and Kroemer, 2003; Mishra and Kumar, 2005; Taylor et al., 2008; Broughton et al., 2009). Our results show signi fi cantly increased expression of the main pro‐apoptotic proteins, Bax and caspase‐3, in the ipsilateral cortical penumbra. Encour‐agingly, nicoti fl orin strongly inhibits their expression. Alter‐natively, the anti‐apoptotic protein, Bcl‐2, was signi fi cantly increased in the nicotiflorin group compared with the I/R group. These findings provide improved understanding of the cerebral protective mechanism of nicoti fl orin.

In conclusion, nicotiflorin ameliorates cortical neuronal shape and structure, and also decreases the number of apop‐totic neurons. Further, this protective mechanism involves the JAK2/STAT3 signaling pathway. Our fi ndings o ff er fur‐ther theoretical evidence for nicoti fl orin as a potential ther‐apeutic drug for ischemic stroke. Of course, other possible mechanisms need to be further studied.

Acknowledgments:We would like to thank Drug Discovery Research Center of Southwest Medical University of China for providing technical platform.

Author contributions:LY designed the experiments and wrote the paper. GQH performed the experiments. XD and BQC were responsible for data analysis. YJL and XQG provided technical support. All authors approved the fi nal version of the paper.

Con fl icts of interest:None declared.

Plagiarism check:This paper was screened twice using CrossCheck to verify originality before publication.

Peer review:

Akbik F, Hirsch JA, Cougo‐Pinto PT, Chandra RV, Simonsen CZ, Les‐lie‐Mazwi T (2016)e evolution of mechanical thrombectomy for acute Stroke. Curr Treat Options Cardiovasc Med 18:32.

ArunaDevi R, Ramteke VD, Kumar S, Shukla MK, Jaganathan S, Ku‐mar D, Sharma AK, Tandan SK (2010) Neuroprotective effect of s‐methylisothiourea in transient focal cerebral ischemia in rat. Nitric Oxide 22:1‐10.

Babon JJ, Lucet IS, Murphy JM, Nicola NA, Varghese LN (2014)e molecular regulation of Janus kinase (JAK) activation. Biochem J 462:1‐13.

Bahmani P, Schellenberger E, Klohs J, Steinbrink J, Cordell R, Zille M, Muller J, Harhausen D, Hofstra L, Reutelingsperger C, Farr TD, Dirnagl U, Wunder A (2011) Visualization of cell death in mice with focal cerebral ischemia using fluorescent annexin A5, propidium iodide, and TUNEL staining. J Cereb Blood Flow Metab 31:1311‐1320.

Banerjee TK, Das SK (2016) Fiy years of stroke researches in India. Ann Indian Acad Neurol 19:1‐8.

Broughton BR, Reutens DC, Sobey CG (2009) Apoptotic mechanisms aer cerebral ischemia. Stroke 40:e331‐339.

Chai HT, Yip HK, Sun CK, Hsu SY, Leu S (2016) AG490 suppresses EPO‐mediated activation of JAK2‐STAT but enhances blood flow recovery in rats with critical limb ischemia. J In fl amm (Lond) 13:18.

Charriaut‐Marlangue C, Margaill I, Represa A, Popovici T, Plotkine M, Ben‐Ari Y (1996) Apoptosis and necrosis after reversible focal ischemia: an in situ DNA fragmentation analysis. J Cereb Blood Flow Metab 16:186‐194.

Chong ZZ, Shang YC, Hou J, Maiese K (2010) Wnt1 neuroprotection translates into improved neurological function during oxidant stress and cerebral ischemia through AKT1 and mitochondrial apoptotic pathways. Oxid Med Cell Longev 3:153‐165.

Chu K, Lee ST, Koo JS, Jung KH, Kim EH, Sinn DI, Kim JM, Ko SY, Kim SJ, Song EC, Kim M, Roh JK (2006) Peroxisome proliferator‐ac‐tivated receptor‐gamma‐agonist, rosiglitazone, promotes angiogene‐sis aer focal cerebral ischemia. Brain Res 1093:208‐218.

Deng YH, He HY, Yang LQ, Zhang PY (2016) Dynamic changes in neu‐ronal autophagy and apoptosis in the ischemic penumbra following permanent ischemic stroke. Neural Regen Res 11:1108‐1114.

Dirnagl U, Iadecola C, Moskowitz MA (1999) Pathobiology of isch‐aemic stroke: an integrated view. Trends Neurosci 22:391‐397.

Du W, Hong J, Wang YC, Zhang YJ, Wang P, Su WY, Lin YW, Lu R, Zou WP, Xiong H, Fang JY (2012) Inhibition of JAK2/STAT3 signalling induces colorectal cancer cell apoptosis via mitochondrial pathway. J Cell Mol Med 16:1878‐1888.

Feigin VL, Forouzanfar MH, Krishnamurthi R, Mensah GA, Connor M, Bennett DA, Moran AE, Sacco RL, Anderson L, Truelsen T, O’Donnell M, Venketasubramanian N, Barker‐Collo S, Lawes CM, Wang W, Shinohara Y, Witt E, Ezzati M, Naghavi M, Murray C, et al. (2014) Global and regional burden of stroke during 1990‐2010: findings from the Global Burden of Disease Study 2010. Lancet 383:245‐254.

Feng YH, Zhu ZH, Wu CX, Zhou GP (2016) E ff ects of electroacupunc‐ture at points selected by orthogonal experiment on the extracellular signal regulated kinase signal pathway in a rat model of cerebral ischenia‐reperfusion injury. Zhongguo Zuzhi Gongcheng Yanjiu 20:5953‐5958.

Ghoreschi K, Laurence A, O’Shea JJ (2009) Janus kinases in immune cell signaling. Immunol Rev 228:273‐287.

Ghosh N, Yuan X, Turenius CI, Tone B, Ambadipudi K, Snyder EY, Obenaus A, Ashwal S (2012) Automated core‐penumbra quantifi‐cation in neonatal ischemic brain injury. J Cereb Blood Flow Metab 32:2161‐2170.

Gorina R, Petegnief V, Chamorro A, Planas AM (2005) AG490 prevents cell death after exposure of rat astrocytes to hydrogen peroxide or proin fl ammatory cytokines: involvement of the Jak2/STAT pathway. J Neurochem 92:505‐518.

Hachinski V, Donnan GA, Gorelick PB, Hacke W, Cramer SC, Kaste M, Fisher M, Brainin M, Buchan AM, Lo EH, Skolnick BE, Furie KL, Hankey GJ, Kivipelto M, Morris J, Rothwell PM, Sacco RL, Smith SC Jr, Wang Y, Bryer A, et al. (2010) Stroke: working toward a prioritized world agenda. Stroke 41:1084‐1099.

Hardwick JM, Chen YB, Jonas EA (2012) Multipolar functions of BCL‐2 proteins link energetics to apoptosis. Trends Cell Biol 22:318‐328.

Hassan M, Watari H, AbuAlmaaty A, Ohba Y, Sakuragi N (2014) Apoptosis and molecular targeting therapy in cancer. Biomed Res Int 2014:150845.

Hou SK, Hao LN, Wei J, Zhou YJ (2016) Propofol pretreatment com‐bined with umbilical blood mesenchymal stem cell transplantation improves cerebral ischemia‐reperfusion injuries. Zhongguo Zuzhi Gongcheng Yanjiu 20:2810‐2816.

Huang JL, Fu ST, Jiang YY, Cao YB, Guo ML, Wang Y, Xu Z (2007) Pro‐tective e ff ects of Nicoti fl orin on reducing memory dysfunction, ener‐gy metabolism failure and oxidative stress in multi‐infarct dementia model rats. Pharmacol Biochem Behav 86:741‐748.

Jiang CJ, Wang ZJ, Zhao YJ, Zhang ZY, Tao JJ, Ma JY (2016) Erythro‐poietin reduces apoptosis of brain tissue cells in rats after cerebral ischemia/reperfusion injury: a characteristic analysis using magnetic resonance imaging. Neural Regen Res 11:1450‐1455.

Jin R, Liu L, Zhang S, Nanda A, Li G (2013) Role of in fl ammation and its mediators in acute ischemic stroke. J Cardiovasc Transl Res 6:834‐851.

Kalogeris T, Baines CP, Krenz M, Korthuis RJ (2012) Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol 298:229‐317.

Kvansakul M, Hinds MG (2013) Structural biology of the Bcl‐2 family and its mimicry by viral proteins. Cell Death Dis 4:e909.

Lavrik I, Golks A, Krammer PH (2005) Death receptor signaling. J Cell Sci 118:265‐267.

Levine DA, Galecki AT, Langa KM, Unverzagt FW, Kabeto MU, Gior‐dani B, Wadley VG (2015) Trajectory of cognitive decline aer inci‐dent stroke. JAMA 314:41‐51.

Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X (1997) Cytochrome c and dATP‐dependent formation of Apaf‐1/caspase‐9 complex initiates an apoptotic protease cascade. Cell 91:479‐489.

Li R, Guo M, Zhang G, Xu X, Li Q (2006) Nicoti fl orin reduces cerebral ischemic damage and upregulates endothelial nitric oxide synthase in primarily cultured rat cerebral blood vessel endothelial cells. J Ethnopharmacol 107:143‐150.

Liu J, Wang Y, Akamatsu Y, Lee CC, Stetler RA, Lawton MT, Yang GY (2014) Vascular remodeling aer ischemic stroke: mechanisms and therapeutic potentials. Prog Neurobiol 115:138‐156.

Lo EH (2008) A new penumbra: transitioning from injury into repair aer stroke. Nat Med 14:497‐500.

Lo EH, Dalkara T, Moskowitz MA (2003) Neurological diseases: Mech‐anisms, challenges and opportunities in stroke. Nat Rev Neurosci 4:399‐415.

Longa EZ, Weinstein PR, Carlson S, Cummins R (1989) Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke 20:84‐91.

Mattson MP, Kroemer G (2003) Mitochondria in cell death: novel targets for neuroprotection and cardioprotection. Trends Mol Med 9:196‐205.

Mishra NC, Kumar S (2005) Apoptosis: a mitochondrial perspective on cell death. Indian J Exp Biol 43:25‐34.

Murphy J (2003) Pharmacological treatment of acute ischemic stroke. Crit Care Nurs Q 26:276‐282.

Murray V, Norrving B, Sandercock PA, Terent A, Wardlaw JM, Wester P (2010)e molecular basis of thrombolysis and its clinical applica‐tion in stroke. J Intern Med 267:191‐208.

Nakayama M, Aihara M, Chen YN, Araie M, Tomita‐Yokotani K, Iwashina T (2011) Neuroprotective e ff ects of fl avonoids on hypoxia‐, glutamate‐, and oxidative stress‐induced retinal ganglion cell death. Mol Vis 17:1784‐1793.

Nakka VP, Gusain A, Mehta SL, Raghubir R (2008) Molecular mech‐anisms of apoptosis in cerebral ischemia: multiple neuroprotective opportunities. Mol Neurobiol 37:7‐38.

Nita DA, Nita V, Spulber S, Moldovan M, Popa DP, Zagrean AM, Zagrean L (2001) Oxidative damage following cerebral ischemia depends on reperfusion‐a biochemical study in rat. J Cell Mol Med 5:163‐170.

Olsen TS, Larsen B, Herning M, Skriver EB, Lassen NA (1983) Blood fl ow and vascular reactivity in collaterally perfused brain tissue. Evi‐dence of an ischemic penumbra in patients with acute stroke. Stroke 14:332‐341.

Polster BM, Fiskum G (2004) Mitochondrial mechanisms of neural cell apoptosis. J Neurochem 90:1281‐1289.

Poon IK, Hulett MD, Parish CR (2010) Molecular mechanisms of late apoptotic/necrotic cell clearance. Cell Death Di ff er 17:381‐397.

Popp A, Jaenisch N, Witte OW, Frahm C (2009) Identi fi cation of isch‐emic regions in a rat model of stroke. PLoS One 4:e4764.

Satriotomo I, Bowen KK, Vemuganti R (2006) JAK2 and STAT3 activa‐tion contributes to neuronal damage following transient focal cere‐bral ischemia. J Neurochem 98:1353‐1368.

Sugawara T, Fujimura M, Noshita N, Kim GW, Saito A, Hayashi T, Narasimhan P, Maier CM, Chan PH (2004) Neuronal death/survival signaling pathways in cerebral ischemia. NeuroRx 1:17‐25.

Sun K, Fan J, Han J (2015) Ameliorating e ff ects of traditional Chinese medicine preparation, Chinese materia medica and active com‐pounds on ischemia/reperfusion‐induced cerebral microcirculatory disturbances and neuron damage. Acta Pharm Sin B 5:8‐24.

Tao Z, Cheng M, Wang SC, Lv W, Hu HQ, Li CF, Cao BZ (2015) JAK2/ STAT3 pathway mediating in fl ammatory responses in heatstroke‐in‐duced rats. Int J Clin Exp Pathol 8:6732‐6739.

Taylor RC, Cullen SP, Martin SJ (2008) Apoptosis: controlled demoli‐tion at the cellular level. Nat Rev Mol Cell Biol 9:231‐241.

Troy CM, Jean YY (2015) Caspases: therapeutic targets in neurologic disease. Neurotherapeutics 12:42‐48.

Uyttenboogaart M, De Keyser J, Luijckx GJ (2009) Thrombolysis for acute ischemic stroke. Curr Top Med Chem 9:1285‐1290.

Venderova K, Park DS (2012) Programmed cell death in Parkinson’s disease. Cold Spring Harb Perspect Med 2 doi: 10.1101/cshperspect. a009365.

Wang DB, Kinoshita C, Kinoshita Y, Morrison RS (2014) p53 and mi‐tochondrial function in neurons. Biochim Biophys Acta 1842:1186‐1197.

Wang JP, Yang ZT, Liu C, He YH, Zhao SS (2013) L‐carnosine inhibits neuronal cell apoptosis through signal transducer and activator of transcription 3 signaling pathway aer acute focal cerebral ischemia. Brain Res 1507:125‐133.

Wang PF, Xiong XY, Chen J, Wang YC, Duan W, Yang QW (2015) Function and mechanism of toll‐like receptors in cerebral ischemic tolerance: from preconditioning to treatment. J Neuroin fl ammation 12:80.

Xie HF, Xu RX, Wei JP, Jiang XD, Liu ZH (2007) P‐JAK2 and P‐STAT3 protein expression and cell apoptosis following focal cerebral isch‐emia‐reperfusion injury in rats. Nan Fang Yi Ke Da Xue Xue Bao 27:208‐211, 218.

Xu M, Zhang HL (2011) Death and survival of neuronal and astrocytic cells in ischemic brain injury: a role of autophagy. Acta Pharmacol Sin 32:1089‐1099.

Yu L, Chen C, Wang LF, Kuang X, Liu K, Zhang H, Du JR (2013) Neu‐roprotective e ff ect of kaempferol glycosides against brain injury and neuroinflammation by inhibiting the activation of NF‐kappaB and STAT3 in transient focal stroke. PLoS One 8:e55839.

Zaidat OO, Lazzaro MA, Liebeskind DS, Janjua N, Wechsler L, Noguei‐ra RG, Edgell RC, Kalia JS, Badruddin A, English J, Yavagal D, Kir‐mani JF, Alexandrov AV, Khatri P (2012) Revascularization grading in endovascular acute ischemic stroke therapy. Neurology 79:S110‐116.

Zevallos J, Santiago F, Gonzalez J, Rodriguez A, Pericchi L, Rodri‐guez‐Mercado R, Nobo U (2015) Burden of stroke in puerto rico. Int J Stroke 10:117‐119.

Zhang HH, Kuang S, Wang Y, Sun XX, Gu Y, Hu LH, Yu Q (2015) Bi‐gelovin inhibits STAT3 signaling by inactivating JAK2 and induces apoptosis in human cancer cells. Acta Pharmacol Sin 36:507‐516.

Copyedited by James R, Hindle A, Wang J, Qiu Y, Li CH, Song LP, Zhao M

10.4103/1673-5374.198992

Accepted: 2016-10-22

*Correspondence to: Guang-qiang Hu or Lu Yu, hgq863@sina.com or yulu863@sina.com.