trimetazidine protects retinal ganglion cells from acute ... · trimetazidine protects retinal...

31
Trimetazidine protects retinal ganglion cells from acute glaucoma via the Nrf2/Ho-1 pathway Peixing Wan, MD *1 ; Wenru Su, MD, PhD *1 ; Yingying Zhang, MD 1 ; Zhidong Li, MD 1 3 Caibin Deng, BD 1 ; Yehong Zhuo, MD, PhD 1# 4 1 5 6 7 State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China. * These authors contributed equally to this work. 8 # Correspondence to: 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 Yehong Zhuo, MD,PhD, Phone: +86-020-87331382, Fax: +86-020-87333271, Email: [email protected] Wenru Su, MD, PhD, Phone: +86-020-87330402, Fax: +86-020-87333271, Email: [email protected] Author contributions: P.W.: conception and design, data collection and assembly, manuscript writing, and final manuscript approval; W.S.: conception and design, data collection and assembly, manuscript writing, final manuscript approval, and financial support; Y.Zhang, Z.L. and C.D.: data collection and assembly; Y.Zhuo: conception and design, manuscript writing, final manuscript approval. Conflicts of interest: The authors declare that there are no competing financial conflicts of interest. Financial support: This study is supported by NSFC (81470627). The sponsor of the study had no role in the design of the original study protocol, in the collection, analysis and interpretation of the data, in writing the report, or in the decision to submit the manuscript for publication. Key words: Trimetazidine; Retinal ganglion cells; Acute glaucoma; Oxidation; Inflammation 25 ACCEPTED MANUSCRIPT 10.1042/CS20171182 . Please cite using the DOI 10.1042/CS20171182 http://dx.doi.org/ up-to-date version is available at encouraged to use the Version of Record that, when published, will replace this version. The most this is an Accepted Manuscript, not the final Version of Record. You are : Clinical Science ). http://www.portlandpresspublishing.com/content/open-access-policy#Archiving Archiving Policy of Portland Press ( which the article is published. Archiving of non-open access articles is permitted in accordance with the Use of open access articles is permitted based on the terms of the specific Creative Commons Licence under

Upload: hoangdung

Post on 15-Aug-2019

238 views

Category:

Documents


0 download

TRANSCRIPT

Trimetazidine protects retinal ganglion cells from acute glaucoma via the

Nrf2/Ho-1 pathway

Peixing Wan, MD*1

; Wenru Su, MD, PhD*1

; Yingying Zhang, MD1; Zhidong Li, MD

1

3

Caibin Deng, BD1; Yehong Zhuo, MD, PhD

1#

4

1

5

6

7

State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen

University, Guangzhou 510060, China.

*

These authors contributed equally to this work.

8

# Correspondence to:

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

Yehong Zhuo, MD,PhD, Phone: +86-020-87331382, Fax: +86-020-87333271, Email:

[email protected]

Wenru Su, MD, PhD, Phone: +86-020-87330402, Fax: +86-020-87333271, Email:

[email protected]

Author contributions: P.W.: conception and design, data collection and assembly, manuscript

writing, and final manuscript approval; W.S.: conception and design, data collection and assembly,

manuscript writing, final manuscript approval, and financial support; Y.Zhang, Z.L. and C.D.: data collection

and assembly; Y.Zhuo: conception and design, manuscript writing, final manuscript approval.

Conflicts of interest: The authors declare that there are no competing financial conflicts of interest.

Financial support: This study is supported by NSFC (81470627). The sponsor of the study had no role in

the design of the original study protocol, in the collection, analysis and interpretation of the data, in

writing the report, or in the decision to submit the manuscript for publication.

Key words: Trimetazidine; Retinal ganglion cells; Acute glaucoma; Oxidation; Inflammation

25

AC

CE

PT

ED

MA

NU

SC

RIP

T

10.1042/CS20171182. Please cite using the DOI 10.1042/CS20171182http://dx.doi.org/up-to-date version is available at

encouraged to use the Version of Record that, when published, will replace this version. The most this is an Accepted Manuscript, not the final Version of Record. You are:Clinical Science

). http://www.portlandpresspublishing.com/content/open-access-policy#ArchivingArchiving Policy of Portland Press (which the article is published. Archiving of non-open access articles is permitted in accordance with the Use of open access articles is permitted based on the terms of the specific Creative Commons Licence under

ABSTRACT 26

Acute glaucoma is one of the leading causes of irreversible vision impairment characterized 27

by the rapid elevation of intraocular pressure and consequent retinal ganglion cell (RGC) 28

death. Oxidative stress and neuro-inflammation have been considered critical for the 29

pathogenesis of RGC death in acute glaucoma. Trimetazidine (TMZ), an anti-ischemic drug, 30

possesses anti-oxidative and anti-inflammatory properties, contributing to its therapeutic 31

potential in tissue damage. However, the role of TMZ in acute glaucoma and the underlying 32

molecular mechanisms remain elusive. Here, we report that treatment with TMZ significantly 33

attenuated retinal damage and RGC death in mice with acute glaucoma, with a significant 34

decrease in reactive oxygen species (ROS) and inflammatory cytokine production in the retina. 35

Furthermore, TMZ treatment directly decreased ROS production and rebalanced the 36

intracellular redox state, thus contributing to the survival of RGCs in vitro. TMZ treatment 37

also reduced the production of inflammatory cytokines in vitro. Mechanistically, the 38

TMZ-mediated inhibition of apoptosis and inflammatory cytokine production in RGCs 39

occurred via the inhibition of the nuclear factor erythroid 2-related factor 2/heme oxygenase 40

1/caspase-8 pathway. Moreover, the TMZ-mediated neuroprotection in acute glaucoma was 41

abrogated when an HO-1 inhibitor, SnPP, was used. Our findings identify potential 42

mechanisms of RGC apoptosis and proposea novel therapeutic agent, TMZ, which exerts a 43

precise neuroprotective effect against acute glaucoma. 44

CLINICAL PERSPECTIVE 45

Acute glaucoma jeopardizes normal vision due to substantially high intraocular pressure46

(IOP) and consequent retinal ganglion cell (RGC) death; currently, an attractive47

alternative for next-generation glaucoma therapy is to select an effective drug to prevent48

RGC apoptosis in IOP-induced retinal damage.49

This study demonstrated that trimetazidine (TMZ) significantly ameliorated high50

IOP-induced retinal damage and RGC apoptosis, by exerting therapeutic efficacy through51

its anti-oxidative and anti-inflammatory properties via the Nrf2/HO-1 pathway.52

Our findings suggested that TMZ can protect against acute glaucoma and emerge as a53

promising candidate in the treatment of acute glaucoma and other neurological disorders.54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

INTRODUCTION

Acute glaucoma is one of the main causes of irreversible visual impairment and

blindness, especially among people of Asian descent [1,2]. Acute glaucoma is

characterized by considerable high intraocular pressure (IOP), pain and vision loss [3].

The rapid increase and decrease of IOP causes retinal ischemic/reperfusion (I/R)

injury, thus resulting in apoptosis of retinal ganglion cells (RGCs). Although the

pathogenesis of RGC death is not fully understood, oxidative stress and neuro-

inflammation have been considered critical for the pathogenesis for RGC death in retinal

I/R injury during acute glaucoma [4]. Current treatments that lower IOP through

medication and surgery cannot completely prevent the progressive death of RGCs in acute

glaucoma. Therefore, safe and effective novel alternatives are needed for acute glaucoma

treatment.

Trimetazidine (TMZ), a piperazine-derived antianginal agent [5,6], has traditionally

been used as an anti-ischemic drug for coronary artery disease. Recently, studies have

shown that TMZ possesses anti-oxidative and anti-inflammatory properties and plays

cytoprotective roles in various tissues, including nervous tissue, pancreatic tissue and

renal tissue [7,8]. Therefore, TMZ may be a potential therapeutic alternative for acute

glaucoma treatment. However, the role of TMZ in acute glaucoma has not yet been

explored. Moreover, the mechanisms by which TMZ mediates anti-oxidative and anti-

inflammatory effects remain elusive. Thus, in this study, we investigated the therapeutic

effect of TMZ on experimental acute glaucoma and the anti-oxidative and anti-inflammatory

mechanisms of TMZ.

74

75

MATERIALS AND METHODS 76

Establishment of the retinal I/R model 77

Six- to eight-week-old C57BL/6J male mice were purchased from Guangdong Medical 78

Laboratory Animal Center. The feeding and administration of mice strictly complied with the 79

Association for Research in Vision and Ophthalmology Statement for the Use of Animals in 80

Ophthalmic and Vision Research. 81

The mice were anesthetized with 100 mg/kg pentobarbital sodium by intraperitoneal injection. 82

Before operation, mice corneas were topically anesthetized with 0.5% Alcaine eye drops, and 83

pupils were dilated with 1% Tropicamide. We then conducted cannulation into the anterior 84

chamber of the right eye using a 30-gauge needle supplied with balanced salt solution to 85

maintain the IOP at 70 mmHg for 60 minutes. Sham operation was performed in the 86

contralateral eye without elevating the IOP to serve as the control. After 60 minutes, the IOP 87

was normalized by withdrawing the needle, and then tobramycin ointment was used to 88

prevent bacterial infection. 89

Preparation of Trimetazidine 90

TMZ with purity higher than 95% was purchased from Sigma Chemical Co. (St Louis, USA), 91

dissolved in sterile PBS to a stock concentration of 0.1 mol/L, and stored at 4°C in the dark to 92

be used within 2 days after preparation. 93

Experiment Grouping Design 94

The mice were randomly divided into five groups: normal control group (Normal), Tin 95

protoporphyrin IX dichloride (SnPP) group, I/R group, TMZ (100 μM) group and TMZ + 96

SnPP group. The mice in I/R, TMZ and TMZ + SnPP groups were subjected to experimental 97

I/R. The normal control and SnPP groups had sham cannulation performed without elevating 98

the IOP as described above. The eyes of mice in the SnPP or TMZ groups were intravitreally 99

injected with 1 μL TMZ [9] or SnPP solution, respectively, before the onset of reperfusion. 100

The eyes of mice in the TMZ + SnPP group were intravitreally injected with 1 μL TMZ and 101

SnPP solution together before the onset of reperfusion. The eyes of mice in the normal control 102

group were injected with 1 μL of sterile PBS into the vitreous cavity. 103

RGC Labeling and Survival Quantification. 104

Mice were anesthetized with 100 mg/kg pentobarbital sodium by intraperitoneal injection and 105

placed in a stereotactic apparatus (Stoelting, USA). The skull was exposed and cleaned with 106

PVP-J. Bilateral holes were drilled at the surface of superior colliculi. Approximately 1 µl of 107

4% Fluorogold (FG, hydroxystilbamidine; Fluorochrome, USA) solution was injected into 108

both superior colliculis. After injection, the micro-syringe was kept still for 30 seconds and 109

then slowly removed. Finally, the dissected scalp was sutured with topical application of 110

tobramycin. 111

To ensure proper RGC labeling, the animals were allowed 7 days for retrograde transport of 112

FG before sacrifice. FG-positive RGCs were identified with a fluorescence microscope 113

(AxioImager; Carl Zeiss MicroImaging Inc., USA) in retinal flat mount. Surviving RGCs 114

(green dots) were counted automatically using ImageJ (LOCI, University of 115

Wisconsin-Madison). 116

Histological examination 117

Seven days after I/R treatment, mouse eyes were enucleated and embedded in paraffin. Every 118

paraffin block was sectioned to 4-µm thickness through the optic nerve. Three sections of 119

each eye were cut and stained with hematoxylin and eosin (H&E). The inner plexiform layer 120

(IPL) thickness was measured within 1 mm to the optic nerve center to quantify retinal 121

damage by Axiovision software (Carl Zeiss MicroImaging Inc.). The data from three sections 122

per eye were averaged. 123

Primary culture of RGCs 124

Primary RGCs were obtained according to the protocol published by Alissa Winzeler and 125

Jack T. Wang in 2013 [10]. In brief, retinal samples were separated from neonatal mice to 126

prepare single cell suspensions. The retinal suspension was incubated in rabbit anti-mouse 127

macrophage antibody-coated flasks (Cedarlane, USA) and goat anti-mouse macrophage 128

antibody-coated flasks (Jackson Immuno Research, USA) to remove the adherent 129

macrophages. The non-adherent cells were transferred to Thy1.2 monoclonal antibody-coated 130

flasks (Millipore Chemicon, USA) to collect adherent cells. The adherent RGCs were 131

incubated at 37°C in 5% CO2 with RGC Growth Medium containing supplements as 132

described. 133

Mix culture with BV2 cells 134

Mouse microglia cell line BV2 (ATCC, USA) was co-cultured with primary RGCs in DMEM 135

(High Glucose 4.5 g/ml, Gibco, USA) supplemented with 10% fetal bovine serum (FBS) at 136

37°C with 5% CO2 and 95% air. RGCs were placed on the upper permeable membrane of a 137

transwell chamber, with BV2 cells grown in the lower well of the plate for 24 hours before 138

treatment. 139

Establishment of the Oxygen-glucose deprivation and reperfusion (OGD/R) model 140

The OGD/R model was established as follows: the culture medium was replaced with 141

glucose-free DMEM (Gibco) after washing the cells twice with PBS, and then the cells were 142

placed in a modular incubator chamber (Billups-Rothenberg, Inc., Del Mar, CA) filled with a 143

gas mixture of 5% CO2 and 95% N2 at 37°C for 3 hours. The control cells were incubated in 144

serum-free medium with 4.5 g/L D-glucose under normoxic conditions (5% CO2 and 95% air) 145

for the same duration. At the end of the exposure period, the cells were returned to normoxic 146

conditions with glucose and incubated for 12 hours. 147

Cell treatment with TMZ 148

Primary RGCs were cultured in 6-well plates or transwell plates before treatment. TMZ was 149

added at indicated concentrations into cell supernatants 6 hours prior to the onset of OGD/R 150

or other assays. TMZ was diluted in DMEM and added at concentrations ranging from 0.1 to 151

100 μM. 152

Enzyme-linked Immunosorbent Assay (ELISA) 153

The cytokines TNFα and IL1β were measured in the culture supernatant of BV2 cells. 154

Immunochemical analyses were conducted using commercially available ELISA kits 155

(eBioscience, Vienna, Austria). For measuring the level of 3-nitrotyrosine proteins in retinal 156

homogenates, a commercial kit was used (Oxiselect Nitrotyrosine kit; Cell Biolabs, Inc., 157

USA), following the manufacturer’s instructions. 158

Viability and Apoptosis Assays in RGCs 159

Cell viability was measured using the CCK8 Assay Kit (Beyotime Biotechnology, China) 160

according to the manufacturer’s protocol. CCK8-reduction activity was presented as the 161

percentage of the unexposed control cells (100%). Flow cytometry was also performed to 162

measure OGD/R-induced apoptosis of RGCs by using a propidium iodide (PI) and Annexin 163

V-FITC detection kit (BD Biosciences, New York, USA) according to the manufacturer’s 164

instructions. Flow cytometric analysis using FlowJo 7.6.2 (FlowJo, LLC) was performed 165

following standard protocols. 166

Inhibition of Ho-1 Activity 167

Ho-1 activity was inhibited in vivo through an intraperitoneal injection of SnPP (40 mol/kg, 168

Tocris Bioscience, USA) 0.5 h prior to I/R and once daily for 3 days after I/R. SnPP was 169

dissolved in 0.1 N NaOH and diluted with PBS (pH = 7.4). 170

Cells were seeded in a 6-well plate at approximately 70% confluency. Ten micromolar SnPP 171

or vehicle was added to the culture medium for 6 hours. The cells were then treated with TMZ 172

or vehicle for 6 hours and subjected to OGD/R. 173

Detection of Intracellular ROS Levels 174

Quantification of intracellular reactive oxygen species (ROS) accumulation was performed by 175

fluorescence detection as well as flow cytometry using the fluorescent probe 176

2′,7′-dichlorofluorescein diacetate (DCFH-DA, KeyGEN, China). Primary RGCs were 177

subjected to the appropriate treatments and then incubated for 20 minutes in the dark at 37°C 178

with 10 µM DCFH-DA solutions. After incubation, the cells were analyzed within 30 minutes. 179

Mean fluorescence intensity of ROS was measured using a fluorescence microscope, and flow 180

cytometry was performed using a Fortessa system (BD). The data were analyzed using 181

ImageJ and FlowJo software. 182

Analysis on MMP (JC-1 staining) 183

The changes in mitochondrial membrane potential (MMP) in RGCs were explored using the 184

5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethyl benzimidazolyl carbocyanine iodide (JC-1) probe as 185

described by the manufacturer and then visualized using an inverted fluorescence microscope. 186

The average result of three plates of cells or mice were obtained for each condition. 187

Quantitative Real-time PCR 188

Total RNA was extracted from the retinal samples and cultured cells by TRIzol reagent 189

(Invitrogen, USA) following the manufacturer’s protocol. cDNA synthesis was conducted 190

with PrimeScript RT Master Mix (TaKaRa, China). Quantitative analysis was performed with 191

a Light Cycler 480 Real-Time PCR System. The expression level of target mRNA was 192

measured and normalized to Gapdh. The primer sequences are as follows: GAPDH-forward 193

primer CCGGGAAACTGTGGCGTGATGG; GAPDH-reverse primer 194

AGGTGGAGGAGTGGGTGTCGCTGTT; TNFα-forward primer 195

GCACCACCATCAAGGACTCAA; TNFα-reverse primer TCGAGGCCCAGTGAATTCG; 196

IL1β -forward primer GGGCCTCAAAGGAAAGAATC; IL1β-reverse primer 197

CTCTGCTTGTGAGGTGCTGA. 198

Western Blot 199

Total protein was isolated from the retinal samples and cultured cells according to standard 200

procedures. Nuclear protein was extracted using ProteoExtract® Subcellular Proteome 201

Extraction Kit (Merck Millipore, German) following the manufacturer’s protocol. Proteins 202

were run on 12% polyacrylamide gels following standard protocol. The expression of total 203

protein and nuclear protein was normalized to Gapdh and Histone H3, respectively, and then 204

quantified using ImageJ. 205

The primary antibodies and dilutions used were as follows: anti-Nrf2 (CST, USA, 1:400), 206

anti-Ho-1 (CST, 1:200), anti-cleaved-Caspase-8 (CST, 1:400), anti-Gapdh (CST, 1:400), 207

anti-Histone H3 (Abcam, UK, 1:400). 208

Caspase-8 Activity Assay 209

The Caspase-8 activity of retinal tissues and RGCs were performed using the CaspGLOWTM

210

Fluorescein Active Caspase-8 Staining Kit (BioVision, Milpitas, USA) according to the 211

manufacturer’s instructions. One microliter of FITC-IETD-FMK was added into each tube 212

containing 300 µl of cell suspension and incubated for 1 hour at 37°C in 5% CO2. Then, the 213

cells were resuspended, and the fluorescence intensity was measured at Ex/Em = 485/535 nm. 214

Statistical Analysis 215

The data were presented as the mean ± standard deviation (SD). One-way analysis of variance 216

(ANOVA) was performed, followed by Bonferroni’s post hoc test using SPSS software, 217

version 17 (SPSS Inc., Chicago, IL, USA). All statistical tests were two-tailed, and a p-value 218

below 0.05 was considered statistically significant. 219

220

RESULTS 221

Neuroprotective effects of intravitreal TMZ in IOP-induced retinal damage 222

To evaluate the role of TMZ in acute glaucoma, TMZ was administered intravitreally before 223

acute elevation of IOP. We first investigated the change in retinal thickness in three 224

independent groups of mice. Retinas exposed to I/R displayed a significant decrease in IPL 225

thickness compared with normal retinas 7 days after reperfusion. However, the damage to the 226

retina, especially to the IPL, was significantly ameliorated by TMZ (Fig. 1A and 1B). The 227

irreversible apoptosis of RGCs is another important indicator for the functional damage of the 228

retina. FG analysis demonstrated that the number of RGCs was decreased in the experimental 229

mice at 7 days after reperfusion compared with that observed in control mice (Fig. 1C and 230

1D). Intravitreal TMZ injections significantly decreased the severity of retinal damage and 231

extent of RGC death (Fig. 1C and 1D). Previous studies have shown that over-production and 232

accumulation of neurotoxic mediators, including ROS, TNF-α and IL-1β, play important roles 233

in the primary and secondary waves of RGC apoptosis [11-13]. As shown in Fig. 1E, the 234

production of nitrotyrosine (a protein derivative of ROS) was significantly increased in the 235

whole retina at 24 hours after reperfusion compared with that in control retinas, and it 236

declined in the retinas treated with TMZ. In addition, intravitreal injection of TMZ 237

significantly suppressed the expression of TNFα and IL1β mRNA (Fig. 1F and 1G). 238

Collectively, these data show that TMZ exerts neuroprotective effects against IOP-induced 239

retinal damage. 240

TMZ directly protects RGC apoptosis induced by OGD/R 241

Next, we performed a series of in vitro studies to explore whether TMZ is capable of directly 242

inhibiting the apoptosis of RGCs and the underlying mechanisms. We first identified primary 243

cultured RGCs by characteristic markers. Immunostaining results indicated that primary 244

RGCs indeed expressed Brn-3a, β3-tubulin and RBPMS (Fig. 2A). CCK8 assay was used to 245

test whether TMZ can influence cellular metabolism or proliferation. The results showed that 246

cellular metabolism was affected when TMZ concentration reached 50 μM (Fig. 2B). 247

Therefore, we selected the concentration range from 0.1 to 10 μM for subsequent in vitro 248

studies. We then exposed primary RGCs to OGD/R to effectively mimic the IOP-induced I/R 249

damage in vivo [4,14]. OGD/R decreased cell viability, and TMZ (0.1-10 μM) treatment 250

significantly increased the number of surviving RGCs (Fig. 2C). The flow cytometry results 251

showed that the number of apoptotic cells marked by PI and AnnexinV was increased after 252

OGD/R exposure, whereas TMZ treatment significantly decreased the number of 253

PI+Annexin+ cells (Fig. 2D and 2E). The TMZ concentration range from 1 to 10 μM was 254

found to be the most effective in exerting cytoprotective effects against OGD/R. In addition, 255

TMZ significantly decreased caspase-8 activation as measured by Western blotting and 256

caspase-8 activity assay (Fig. 2F and 2G). These results demonstrate that TMZ decreases 257

OGD/R-induced RGC apoptosis in vitro. 258

The anti-oxidant effect of TMZ is involved in the TMZ-mediated neuroprotection in 259

RGCs 260

Primary RGCs in the OGD/R group display a greater extent of ROS production than the cells 261

in the control group. TMZ was found to significantly attenuate the over-production of ROS in 262

RGCs exposed to OGD/R as marked by the reduction in green fluorescence in a 263

concentration-dependent manner (Fig. 3A and 3B). The diminished ROS production in 264

primary RGCs by TMZ could also be observed by flow cytometry. The peak area occupied by 265

FITC+ cells, representing cells with excess ROS, was found to be significantly decreased by 266

TMZ treatment (Fig. 3C and 3D). OGD/R also resulted in a decrease in the MMP in RGCs, 267

which was marked by fluorescence shift from red to green. However, TMZ treatment 268

protected mitochondrial function by restoring the MMP (Fig. 3F and 3G). These results 269

further support the anti-oxidant effect of TMZ on RGCs in vitro. 270

TMZ conferred protection against apoptosis in RGCs via Nrf2/Ho-1 signaling 271

The above results showed that the anti-oxidant effects of TMZ are actively involved in the 272

TMZ-mediated protection of RGCs. Nrf2, a transcription factor, is regarded as a master 273

regulator of cellular responses to oxidative stress. Thus, we next determined whether Nrf2 and 274

its downstream signaling factor, Ho-1, is associated with the TMZ-mediated protection 275

against RGC apoptosis. The Western blotting results showed that TMZ (10 μM) treatment 276

significantly induced the nuclear translocation of Nrf2 in RGCs under OGD/R conditions (Fig. 277

4A). TMZ treatment also increased Ho-1 protein expression compared with vehicle treatment 278

(Fig. 4B). To further confirm the role of Nrf2/Ho-1 signaling in the TMZ-mediated 279

neuroprotection of RGCs, SnPP, a Ho-1 inhibitor was used. Our results showed that SnPP 280

treatment significantly but incompletely reversed the TMZ-mediated neuroprotection of 281

RGCs (Fig. 4C and 4D). The reduction of intracellular ROS (Fig. 4E and 4F) and alterations 282

in MMP (Fig. 4G and 4H) induced by TMZ were partially inhibited by SnPP. Taken together, 283

these data suggest that TMZ confers neuroprotection through an Nrf2/Ho-1-dependent 284

mechanism. 285

Nrf2/Ho-1 signaling is essential for TMZ-mediated anti-inflammatory effects in vitro 286

We also performed a series of in vitro experiments to further evaluate the anti-inflammatory 287

effects of TMZ and to identify the underlying mechanisms. For this purpose, BV2 cells, an 288

established mouse microglial cell line was cultured under OGD/R conditions. BV2 cells were 289

plated in medium in the presence or absence of TMZ for 6 h before OGD/R. We used ELISA 290

kits to examine the concentration of TNFα and IL1β in the supernatant of BV2 cells after 291

OGD/R. The OGD/R-induced over-production of TNFα and IL1β could be inhibited by TMZ 292

(Fig. 5A). The real-time PCR results further confirmed the anti-inflammatory effects of TMZ 293

(Fig. 5B). Nrf2/Ho-1 signaling plays an important role in the TMZ-mediated neuroprotection 294

of RGCs. Thus, we next assessed whether this pathway is also involved in the 295

anti-inflammatory effects of TMZ. As shown in Fig. 5C and 5D, TMZ significantly induced 296

the nuclear translocation of Nrf2 and increased the protein expression of Ho-1 in BV2 cells 297

under OGD/R conditions. Our results demonstrated that SnPP significantly but incompletely 298

reversed the TMZ-mediated inhibition of TNFα and IL1β production (Fig. 5E and 5F). In 299

addition, the inhibitory effect of TMZ on caspase-8 activation was significantly decreased 300

after SnPP treatment (Fig. 5G). These results indicate that TMZ is capable of regulating 301

Nrf2/Ho-1 signaling to exert anti-inflammatory effects in vitro. 302

TMZ protects RGCs from IOP-induced damage via Nrf2/Ho-1 signaling 303

Our in vitro studies showed that Nrf2/Ho-1 signaling plays a critical role in TMZ-mediated 304

anti-oxidant and anti-inflammatory effects. Therefore, we determined whether Nrf2/Ho-1 305

signaling was also involved in the TMZ-mediated neuroprotection against IOP-induced retinal 306

damage. The Western blotting results showed that TMZ treatment significantly induced the 307

nuclear translocation of Nrf2 and increased the expression of Ho-1 in the retinas (Fig. 6A and 308

B). SnPP was intraperitoneally injected (40 mol/kg) 0.5 h prior to I/R and once daily for 3 309

days after I/R. As shown in Fig. 6C-F, treatment with the Ho-1 inhibitor significantly blocked 310

the TMZ-mediated neuroprotection against IOP-induced retinal damage. The Ho-1 inhibitor 311

also significantly decreased the inhibitory effect of TMZ on ROS accumulation and 312

inflammatory cytokine production (Fig 6G-I). These findings suggest that the upregulation of 313

Nrf2/Ho-1 signaling may contribute, at least in part, to the TMZ-mediated neuroprotection 314

against IOP-induced retinal damage. 315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

DISCUSSION

Acute glaucoma jeopardizes normal vision due to substantially high IOP. The progressive loss

of RGCs caused by rapid increase of IOP is mainly responsible for irreversible blindness in

acute glaucoma [15]. Currently, selecting effective drugs to prevent RGC apoptosis in

IOP-induced retinal damage has become an attractive alternative for next-generation

glaucoma therapy. TMZ, which improves energy metabolism in ischemic myocytes, was first

developed as an anti-angina drug [16]. However, a growing number of studies suggest that

TMZ possesses numerous non-antianginal functions, including immunosuppression [17],

anti-oxidation [18], anti-ischemia [19,20] and anti-apoptosis [21]. This study

demonstrated that treatment with TMZ significantly ameliorated high IOP-induced retinal

damage and RGC apoptosis, with a significant decrease in ROS and inflammatory cytokine

production in the retina. TMZ primarily exerts its therapeutic efficacy through its

anti-oxidative and anti-inflammatory effects. These results are consistent with those of

previous studies, showing that TMZ treatment attenuates acute inflammatory responses and

oxidative stress in experimental cardiac remodeling [22] and pancreatitis [23]. Next, we

found that the anti-oxidative and anti-inflammatory effects of TMZ depended primarily on the

Nrf2/HO-1 pathway. Interestingly, TMZ treatment in IOP-induced retinal damage resulted

in an obvious activation of the Nrf2/HO-1 pathway, which plays a crucial role in TMZ-

mediated protection against IOP-induced retinal damage. These findings suggest that

TMZ exerts neuroprotective effects against acute glaucoma mainly through anti-

oxidative and anti-inflammatory properties.

The transcription factor Nrf2 is a central regulator of cellular responses to oxidative stress

stimulation [24]. Under normal conditions, Nrf2 is an inactive complex in the cytoplasm with

Kelch-like ECH-associated protein 1. When cells undergo oxidative stress, the complex is

disrupted, and, subsequently, Nrf2 translocates to the nucleus, leading to target gene

transcription, including Ho-1, a key heme-degrading enzyme [25,26]. In addition to its

well-known anti-oxidative properties, the Nrf2/Ho-1 signaling pathway also exhibits

anti-inflammatory and anti-apoptotic proprieties [27,28]. In this study, we demonstrated that

TMZ treatment promoted the nuclear translocation of Nrf2 and subsequently increased the 345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

expression of HO-1 in primary RGCs and BV2 cells. Furthermore, HO-1 blockade

significantly but incompletely reversed the TMZ-mediated anti-oxidative and

anti-inflammatory effects. Importantly, the HO-1 inhibitor also blocked the TMZ-mediated

neuroprotection in RGCs in acute glaucoma. Therefore, these compelling findings suggest

that Nrf2/HO-1 signaling contributes, at least in part, to the TMZ-mediated neuroprotection in

acute glaucoma.

Caspase-8, a well-characterized initiator of apoptotic signaling, has been shown to have

multiple non-apoptotic functions. Burguillos et al. reported that caspase-8 promotes

microglial activation and neuroinflammation. Our recent study indicated that caspase-8

mediates the production of cytokines, including IL1β and TNFα in immunocytes, which play

important roles in high-IOP-induced retinal damage [4]. In this study, we observed that TMZ

inhibited caspase-8-mediated RGC apoptosis. Furthermore, TMZ reduced caspase-8-

dependent inflammatory reactions both in vivo and in vitro. Therefore, combining our results

on the protective role of TMZ in acute glaucoma with existing evidence enables us to propose

a mechanistic model that is described in Fig. 7 Proposed model for the neuroprotective

mechanisms of TMZ in acute glaucoma.

The unique pharmacological properties of TMZ enhance its efficacy and enable it to reach its

peak concentration in less than 2 h. Timely treatment is closely associated with efficacy of

neuroprotection [1,2]. Hence, TMZ could be used to halt the early stages of RGCs apoptosis

in acute glaucoma, which is attributable to its fast mechanism of action. Due to its efficacy

and safety, TMZ could be used a promising candidate to treat acute glaucoma and other

related ocular diseases.

In summary, our study initially demonstrated the neuroprotective effects of TMZ on

experimental acute glaucoma. Our results showed that TMZ inhibited RGC apoptosis and

inflammation in vitro and in vivo, possibly by inhibiting caspase-8 signaling. Importantly, we

further determined the neuroprotective mechanisms of TMZ by demonstrating that

Nrf2-HO-1 signaling is essential for TMZ-mediated neuroprotective effects in vitro and in

vivo. These findings provide compelling evidence that TMZ can protect against acute

glaucoma and emerge as a promising candidate in the treatment of acute glaucoma. 374

References375

376

1 Guo, L., Moss, S. E., Alexander, R. A., Ali, R. R., Fitzke, F. W. and Cordeiro, M. F. (2005) Retinal ganglion cell 377

apoptosis in glaucoma is related to intraocular pressure and IOP-induced effects on extracellular matrix. Invest. 378

Ophthalmol. Vis. Sci. 46,175-182 379

2 Guo, L., Salt, T. E., Maass, A., Luong, V., Moss, S. E., Fitzke, F. W. and Cordeiro, M. F. (2006) Assessment of 380

neuroprotective effects of glutamate modulation on glaucoma-related retinal ganglion cell apoptosis in vivo. Invest. 381

Ophthalmol. Vis. Sci. 47,626-633 382

3 Shakya, S.and Gupta, H. R. (2006) Angle closure glaucoma: a cause for bilateral visual threat. Nepal Med Coll J 383

8,153-155 384

4 Chi, W., Li, F., Chen, H., Wang, Y., Zhu, Y., Yang, X., Zhu, J., Wu, F., Ouyang, H., Ge, J., Weinreb, R. N., Zhang, K. 385

and Zhuo, Y. (2014) Caspase-8 promotes NLRP1/NLRP3 inflammasome activation and IL-1β production in acute 386

glaucoma. Proc. Natl. Acad. Sci. U.S.A. 111,11181-11186 387

5 Lopaschuk, G. D., Barr, R., Thomas, P. D. and Dyck, J. R. (2003) Beneficial effects of trimetazidine in ex vivo 388

working ischemic hearts are due to a stimulation of glucose oxidation secondary to inhibition of long-chain 3-ketoacyl 389

coenzyme a thiolase. Circ. Res. 93,e33-37 390

6 MacInnes, A., Fairman, D. A., Binding, P., Ja, R., Wyatt, M. J., Phelan, A., Haddock, P. S. and Karran, E. H. (2003) 391

The antianginal agent trimetazidine does not exert its functional benefit via inhibition of mitochondrial long-chain 392

3-ketoacyl coenzyme A thiolase. Circ. Res. 93,e26-32393

7 Atilgan, D., Parlaktas, B. S., Uluocak, N., Erdemir, F., Markoc, F., Saylan, O. and Erkorkmaz, U. (2014) The effects 394

of trimetazidine and sildenafil on bilateral cavernosal nerve injury induced oxidative damage and cavernosal fibrosis 395

in rats. ScientificWorldJournal 2014,970363 396

8 Serarslan, Y., Bal, R., Altug, M. E., Kontaş, T., Keleş, O. N., Unal, D. and Unal, B. (2009) Effects of trimetazidine on 397

crush injury of the sciatic nerve in rats: a biochemical and stereological study. Brain Res. 1247,11-20 398

9 Kay, L., Finelli, C., Aussedat, J., Guarnieri, C. and Rossi, A. (1995) Improvement of long-term preservation of the 399

isolated arrested rat heart by trimetazidine: effects on the energy state and mitochondrial function. Am. J. Cardiol. 400

76,45B-49B 401

10 Winzeler, A.and Wang, J. T. (2013) Purification and culture of retinal ganglion cells from rodents. Cold Spring Harb 402

Protoc 2013,643-652 403

11 Li, S. Y., Fu, Z. J., Ma, H., Jang, W. C., So, K. F., Wong, D. and Lo, A. C. (2009) Effect of lutein on retinal neurons 404

and oxidative stress in a model of acute retinal ischemia/reperfusion. Invest. Ophthalmol. Vis. Sci. 50,836-843 405

12 Liu, L., Sun, Q., Wang, R., Chen, Z., Wu, J., Xia, F. and Fan, X. Q. (2016) Methane attenuates retinal 406

ischemia/reperfusion injury via anti-oxidative and anti-apoptotic pathways. Brain Res. 1646,327-333 407

13 Yang, C., Lafleur, J., Mwaikambo, B. R., Zhu, T., Gagnon, C., Chemtob, S., Di, P. A. and Hardy, P. (2009) The role 408

of lysophosphatidic acid receptor (LPA1) in the oxygen-induced retinal ganglion cell degeneration. Invest. 409

Ophthalmol. Vis. Sci. 50,1290-1298 410

14 Osborne, A., Aldarwesh, A., Rhodes, J. D., Broadway, D. C., Everitt, C. and Sanderson, J. (2015) Hydrostatic 411

pressure does not cause detectable changes in survival of human retinal ganglion cells. PLoS ONE 10,e0115591 412

15 Almasieh, M., Wilson, A. M., Morquette, B., Cueva, V. J. L. and Di, P. A. (2012) The molecular basis of retinal 413

ganglion cell death in glaucoma. Prog Retin Eye Res 31,152-181 414

16 Mehrotra, T. N.and Bassadone, E. T. (1967) Trimetazidine in the treatment of angina pectoris. The British journal of 415

clinical practice 21,553-554 416

17 Chen, J., Lai, J., Yang, L., Ruan, G., Chaugai, S., Ning, Q., Chen, C. and Wang, D. W. (2016) Trimetazidine prevents 417

macrophage-mediated septic myocardial dysfunction via activation of the histone deacetylase sirtuin 1. Br. J. 418

Pharmacol. 173,545-561 419

18 Tselepis, A., Doulias, P., Lourida, E., Glantzounis, G., Tsimoyiannis, E. and Galaris, D. (2001) Trimetazidine protects 420

low-density lipoproteins from oxidation and cultured cells exposed to H(2)O(2) from DNA damage. Free Radic. Biol. 421

Med. 30,1357-1364 422

19 Demir, T., Turgut, B., Ozercan, I., Gul, F. C., Ilhan, N. and Celiker, U. (2010) Trimetazidine for prevention of 423

induced ischemia and reperfusion of guinea pig retina. Clin Ophthalmol 4,21-26 424

20 Ozden, S., Kildaci, B., Muftuoglu, S., Cakar, N. and Yildirim, C. (2001) Effect of trimetazidine on retinal 425

ischemia/reperfusion injury in rats. Ophthalmologica. Journal international d'ophtalmologie. International journal of 426

ophthalmology. Zeitschrift fur Augenheilkunde 215,309-317 427

21 Gong, X., Fan, G., Wang, W. and Wang, G. (2014) Trimetazidine protects umbilical cord mesenchymal stem cells 428

against hypoxia and serum deprivation induced apoptosis by activation of Akt. Cell. Physiol. Biochem. 34,2245-2255 429

22 Zhou, X., Li, C., Xu, W. and Chen, J. (2012) Trimetazidine protects against smoking-induced left ventricular 430

remodeling via attenuating oxidative stress, apoptosis, and inflammation. PLoS ONE 7,e40424 431

23 Tanoglu, A., Yazgan, Y., Kaplan, M., Berber, U., Kara, M., Demırel, D. and Ipcioglu, O. M. (2015) Trimetazidine 432

significantly reduces cerulein-induced pancreatic apoptosis. Clin Res Hepatol Gastroenterol 39,145-150 433

24 Ma, Q. (2013) Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 53,401-426 434

25 Loboda, A., Damulewicz, M., Pyza, E., Jozkowicz, A. and Dulak, J. (2016) Role of Nrf2/HO-1 system in 435

development, oxidative stress response and diseases: an evolutionarily conserved mechanism. Cell. Mol. Life Sci. 436

73,3221-3247 437

26 Na, H. K.and Surh, Y. J. (2014) Oncogenic potential of Nrf2 and its principal target protein heme oxygenase-1. Free 438

Radic. Biol. Med. 67,353-365 439

27 Li, F., Lu, S., Zhu, R., Zhou, Z., Ma, L., Cai, L. and Liu, Z. (2011) Heme oxygenase-1 is induced by thyroid hormone 440

and involved in thyroid hormone preconditioning-induced protection against renal warm ischemia in rat. Mol. Cell. 441

Endocrinol. 339,54-62 442

28 Paine, A., Eiz-Vesper, B., Blasczyk, R. and Immenschuh, S. (2010) Signaling to heme oxygenase-1 and its 443

anti-inflammatory therapeutic potential. Biochem. Pharmacol. 80,1895-1903 444

445

446

Figure legends 447

448

449

RG

C s

ur

viv

al

(%

)

No

r ma

l

I / R

TM

Z

0

5 0

1 0 0 * *

C D

Normal I/R TMZ

Me

an

th

ick

ne

ss

of

IPL

(μM

)

Norm

a lI /

R

TM

Z

1 0

2 0

3 0

4 0

**

GCLIPL

INL

ONL

A B

Figure 1

Nit

roty

rosin

e

(

M)

Norm

a lI /

R

TM

Z

0

3 0

6 0

9 0

**

TN

F

mR

NA

(F

old

ch

an

ge

)

No

r ma

l

I / R

TM

Z

0

2

4

6* *

IL

1

mR

NA

(F

old

ch

an

ge

)

No

r ma

l

I / R

TM

Z

0

2

4

6

* *

E F G

Figure 1

Normal I/R TMZ

Figure 1. Neuroprotective effects of intravitreal TMZ against IOP-induced retinal 450

damage 451

A and B. H&E staining of retinal cross-sections showing that intravitreal TMZ injection 452

significantly inhibited the attenuation of total retinal thickness and IPL thickness in response 453

to I/R damage 7 days after reperfusion. 454

C and D. FG labeling was performed 7 days after reperfusion. The results indicated that TMZ 455

significantly increased the number of surviving cells in the RGC layer compared with that in 456

the untreated I/R group. 457

E. The production of nitrotyrosine was significantly decreased by TMZ treatment in the whole 458

retina 24 hours after reperfusion compared with that in the untreated retinas. 459

F and G. As measured by real-time PCR, intravitreal injection of TMZ significantly 460

suppressed the expression of TNFα and IL1β mRNA in retinas. 461

GCL, ganglion cell layer; INL, inner nuclear layer; IPL, inner plexiform layer; ONL, outer 462

nuclear layer. Scale bar = 50 μm. 463

The data represents the means ± SD (n=6). *p < 0.05,

**p < 0.01. 464

465

466

467

Annexin

PI

0 0.1 1 10 μM

T M Z ( M )

Ce

ll

Su

rv

iva

l

(%

)

0

0. 1 1

10

50

10

0

5 0

7 5

1 0 0

*

RBMPS β3-tublin

Brn

3a

/DA

PI

Ce

ll s

urv

iva

l

(%)

0 00.1 1 1

0

2 5

5 0

7 5

1 0 0

O G D /R + T M Z (M )

**

**

A B C

D E

Figure 2

Re

la

tiv

e d

en

sity

00

. 1 11

00 . 0

0 . 2

0 . 4

0 . 6* *

* *

T M Z ( M )

CleavedCasp8

Gapdh

Ca

sp

8 a

ctiv

ity

(F

old

ch

an

ge

)

00

. 1 11

00 . 0 0

0 . 2 5

0 . 5 0

0 . 7 5

1 . 0 0

* *

* *

T M Z ( M )

F G

PI

+A

nn

ex

in+

Ce

lls

(%

)

00

. 1 11

00

5

1 0

1 5

2 0 * *

*

T M Z ( M )

15.8% 4.17% 3.15% 2.93%

43kDa

37kDa

Figure 2. TMZ directly protects RGCs against apoptosis induced by OGD/R 468

A. Immunostaining results indicated that primary RGCs indeed expressed characteristic 469

markers of retinal ganglion cells including Brn-3a (red), RBPMS (green, left) and β3-tubulin 470

(green, right). 471

B. CCK8 assay showed that cellular metabolism was affected when TMZ concentration 472

reached 50 µM. 473

C. CCK8 assay showed that cell survival was significantly increased by TMZ treatment after 474

OGD/R. 475

D and E. The results of flow cytometry showed that the proportion of apoptotic cells was 476

increased to 15.8% (± 3.1%) after OGD/R exposure. TMZ significantly decreased the 477

proportion of PI+Annexin+ cells to an average of 2.93%. 478

F. Western blot analysis demonstrated that TMZ significantly suppressed the level of 479

cleaved-caspase-8. 480

G. The reduction of caspase-8 activity was confirmed by caspase-8 activity assay. 481

Scale bar = 20 μm. The data represent the means ± SD (n=8). *p < 0.05, **p < 0.01. 482

483

484

485

ROS

Cel

l C

ou

nt

TMZ 0 0.1 1 10 μM

86.1% 57.1% 45.4% 28.3%

C D

RO

S+

Ce

lls

(%

)

00

. 1 11

00

3 0

6 0

9 0* *

* *

T M Z ( M )

Figure 3

TMZ 0 0.1 1 10 μM

TMZ 0 0.1 1 10 μM

RO

S+

Ce

lls

(N

um

be

r/m

m2

)

00

. 1 11

00

2 0 0

4 0 0

6 0 0 * *

* *

T M Z ( M )

PE

/F

IT

C

00

. 1 11

00

1

2

3

4

* *

* *

T M Z ( M )

A B

E F

Figure 3. The anti-oxidant effect of TMZ is involved in the TMZ-mediated 486

neuroprotective effect on RGCs 487

A and B. After OGD/R exposure, primary RGCs were subjected to ROS over-production 488

(green fluorescence). TMZ could significantly decreasing ROS levels in RGCs. 489

C and D. As marked by the peak area of FITC+ population, the proportion of cells with 490

excess ROS was significantly decreased from 80% (± 6.1%) to 30% (± 1.7%) in the OGD/R 491

+TMZ RGCs. 492

E and F. OGD/R decreased the MMP, which was marked by a fluorescence shift from red to 493

green in RGCs. TMZ pretreatment elevated the PE/FITC ratio. 494

Scale bar = 50 μm. The data represent the means ± SD (n=8). *p < 0.05, **p < 0.01. 495

496

497

16.92% 18.12 % 3.52% 14.36%

Annexin

PI

OGD/R SnPP TMZ TMZ+SnPP

PI+

An

ne

xin

+(%

)

OG

D/R

SnP

P

TM

Z

0

1 0

2 0

3 0

**

**

T M Z+

S n P P

Figure 4

76.1% 80.34% 22.52 % 58.36 %

ROS

Cel

l co

un

t

OGD/R SnPP TMZ TMZ+SnPP

PE

/FIT

C

OG

D/R

SnP

P

TM

Z

0

1

2

3

****

T M Z+

S n P P

OGD/R SnPP TMZ TMZ+SnPP

RO

S+

Ce

lls (

%)

OG

D/R

SnP

P

TM

Z

0

2 5

5 0

7 5

1 0 0

**

**

T M Z+

S n P P

A B C

D E F

G H

Nrf2

Histone H3

Re

la

tiv

e d

en

sity

OG

D/ R

TM

Z

0 . 0

0 . 3

0 . 6

0 . 9

* *

GapdhR

ela

tiv

e d

en

sity

OG

D/ R

TM

Z

0 . 0

0 . 3

0 . 6

0 . 9* *

Ho-197kDa

17kDa

28kDa

37kDa

Figure 4. TMZ protects RGCs from apoptosis via Nrf2/Ho-1 signaling 498

A and B. Western blot analysis demonstrated that TMZ elevated the nuclear translocation of 499

Nrf2 and total expression of Ho-1 in primary RGCs under OGD/R conditions. 500

C and D. TMZ suppressed OGD/R-induced apoptosis in primary RGCs. SnPP treatment 501

significantly but incompletely reversed TMZ-mediated cytoprotective effect on RGCs. 502

E and F. Flow cytometry assay of ROS accumulation (FITC+) in RGCs. SnPP resulted in an 503

increase in the proportion of FITC+ cells and inhibited the TMZ-induced anti-oxidative 504

effect. 505

G and H. The decreasing of PE/FITC ratio indicated significant disruption of the MMP in the 506

SnPP-treated RGCs, which could not be rescued by TMZ treatment. 507

Scale bar = 50 μm. The data represent the means ± SD (n=8). *p < 0.05, **p < 0.01. 508

509

510

511

nNrf2Histone

H3 Gapdh

Cleaved- casp8

(a) (b) (c)

pg

/ml

OG

D/ R

TM

Z

OG

D/ R

TM

Z

0

5 0

1 0 0

1 5 0 * *

* *

T N F

I L 1

mR

NA

(F

old

ch

an

ge

)

OG

D/ R

TM

Z

OG

D/ R

TM

Z

0 . 0 0

0 . 2 5

0 . 5 0

0 . 7 5

1 . 0 0* * * *

T N F I L 1

Re

la

tiv

e d

en

sity

OG

D/ R

TM

Z

0 . 0

0 . 3

0 . 6

0 . 9

* *

Nrf2

Histone H3 Gapdh

Re

la

tiv

e d

en

sity

OG

D/ R

TM

Z

0 . 0

0 . 3

0 . 6

0 . 9

* *

pg

/ml

OG

D/R

SnP

P

TM

Z

TM

Z+

SnP

P

OG

D/R

SnP

P

TM

Z

TM

Z+

SnP

P0

5 0

1 0 0

1 5 0

**

**

T N F

IL 1

mR

NA

(Fo

ld c

ha

ng

e)

OG

D/R

SnP

P

TM

Z

TM

Z+

SnP

P

OG

D/R

SnP

P

TM

Z

TM

Z+

SnP

P0 .0

0 .5

1 .0

1 .5

****

T N F IL 1

A B C D

E F G

Figure 5

Ca

sp

8 a

cti

vit

y

(Fo

ld c

ha

ng

e)

OG

D/R

SnP

P

TM

Z

TM

Z+

SnP

P

0 .0

0 .5

1 .0

1 .5

**

Ho-197kDa

17kDa

28kDa

37kDa

Figure 5. Nrf2/Ho-1 signaling is essential for the TMZ-mediated anti-inflammatory 512

effects in vitro 513

A. As measured by ELISA, the concentration of TNFα and IL1β in supernatants was found to 514

be significantly reduced by TMZ. 515

B. Real-time PCR analysis showed that TMZ decreased TNFα and IL1β mRNA in the TMZ 516

group. 517

C and D. TMZ significantly induced the nuclear translocation of Nrf2 and increased the 518

protein expression of Ho-1 in BV2 cells under OGD/R conditions 519

E and F. SnPP significantly but incompletely reversed the TMZ-mediated inhibition on TNFα 520

and IL1β production as measured by ELISA and real-time PCR. 521

G. SnPP treatment significantly decreased the inhibitory effect of TMZ on caspase-8 522

activation. 523

The data represent the means ± SD (n=8). *p < 0.05, **p < 0.01. 524

525

526

Nrf2

Histone H3

Re

la

tiv

e d

en

sity

I / R

TM

Z

0 . 0

0 . 3

0 . 6

0 . 9

* *

GapdhR

ela

tiv

e d

en

sity

I / R

TM

Z

0 . 0

0 . 3

0 . 6

0 . 9* *

Me

an

th

ick

ne

ss

of

IPL

( μM

)

I /R

SnP

P

TM

Z

0

1 0

2 0

3 0**

T M Z+

S n P P

RG

C s

urv

iva

l

(%)

I /R

SnP

P

TM

Z

0

5 0

1 0 0 **

T M Z+

S n P P

I/R SnPP TMZ TMZ+SnPP

Figure 6

Nit

roty

rosin

e

(

M)

I /R

SnP

P

TM

Z

0

4 0

8 0

1 2 0 **

T M Z+

S n P P

TN

F

m

RN

A

(Fo

ld c

ha

ng

e)

I /R

SnP

P

TM

Z

0 .0

0 .5

1 .0

1 .5

**

T M Z+

S n P P

IL1

m

RN

A

(Fo

ld c

ha

ng

e)

I /R

SnP

P

TM

Z

0 .0

0 .5

1 .0

1 .5

**

T M Z+

S n P P

A B C

D E F

G H I

I/R SnPP TMZ TMZ+SnPPHo-197kDa

17kDa

28kDa

37kDa

Figure 6. TMZ protects RGCs from IOP-induced damage via Nrf2/Ho-1 signaling 527

A and B. Western blot analysis demonstrated that TMZ dramatically induced the nuclear 528

translocation of Nrf2 and increased the expression of Ho-1 in the retinas. 529

C and D. Treatment with the SnPP significantly blocked the TMZ-mediated neuroprotection 530

in IOP-induced retinal attenuation, especially IPL thickness. 531

E and F. SnPP dramatically inhibited the TMZ-mediated neuroprotection in IOP-induced 532

RGC apoptosis. 533

G. SnPP also significantly decreased the inhibitory effect of TMZ on nitrotyrosine production. 534

H and I. Real-time PCR results confirmed that SnPP significantly suppressed the 535

TMZ-induced decrease in TNFα and IL1β mRNA levels compared with the TMZ only group. 536

537

538

539

Figure 7. Proposed Model for the Neuroprotective Mechanisms of TMZ in Acute 540

Glaucoma 541

I/R damages the function of mitochondria by disrupting the MMP, which leads to the 542

over-production of ROS. ROS pushes cells toward the apoptotic fate and stimulates 543

neuro-inflammation. Nrf2/Ho-1 pathway can be activated by TMZ to confer protective effects 544

by inhibiting the accumulation of ROS. Immunocyte activation during injury results in 545

caspase-8-dependent inflammation by increasing TNFα and IL1β production, which also 546

induces RGC apoptosis. Pro-inflammatory reactions can be effectively suppressed by TMZ. 547

Therefore, RGC apoptosis can be attenuated by pretreatment with TMZ via anti-oxidative and 548

anti-inflammatory mechanisms in acute glaucoma. 549

I/R, ischemia/reperfusion injury; RGC, retinal ganglion cell; MMP, mitochondrial membrane 550

potential; ROS, reactive oxygen species. 551