Articles in PresS. Am J Physiol Cell Physiol (February 4, 2015). doi:10.1152/ajpcell.00353.2014

1

A Neuroprotective Role of the NMDA Receptor Subunit GluN3A (NR3A) in

2

Ischemic Stroke of the Adult Mouse

3 4

Jin Hwan Lee1, Zheng Z. Wei1,2, Dongdong Chen1,2, Xiaohuan Gu1,2, Ling Wei1,2

5

and Shan Ping Yu1,2

6 7

1

Departments of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322; 2

8

Atlanta VA Medical Center, Center for Center for Visual

9

and Neurocognitive Rehabilitation, Decatur, GA 30033

10 11

Running title: Neuroprotective role of GluN3A in adult ischemic stroke

12 13 14

Corresponding author

15

Shan Ping Yu, M.D., Ph.D.

16

101 Woodruff Circle

17

Woodruff Memorial Research Building, Suite 620

18

Emory University School of Medicine

19

Atlanta, GA 30322

20

E-mail: [email protected]

21

Tel. 404-712-8678

1 Copyright © 2015 by the American Physiological Society.

22

Abstract

23

GluN3A or NR3A is a developmentally regulated N-methyl-D-aspartate receptor

24

(NMDAR) subunit, showing a unique inhibitory role that decreases NMDAR current and

25

the receptor mediated Ca2+ influx. In the neonatal brain, GluN3A is shown to associate

26

with synaptic maturation, spine formation, and plays a neuroprotective role. Its functional

27

role in the adult brain, however, is largely unknown. We tested the hypothesis that,

28

disrespecting the relatively lower expression level of GluN3A in the adult brain, this

29

inhibitory NMDAR subunit shows a protective action against ischemia-induced brain

30

injury. In littermate wild type (WT) and GluN3A knockout (KO) mice, focal cerebral

31

ischemia was induced by permanent occlusion of right distal branches of the middle

32

cerebral artery (MCA) plus 10 min ligation of both common carotid arteries (CCAs).

33

Twenty-four hrs after focal cerebral ischemia, the infarction volume assessed using 2,3,5-

34

triphenyltetrazolium chloride (TTC) staining was significantly larger in GluN3A KO

35

mice compared to WT mice. Terminal deoxynucleotidyl transferase dUTP nick end

36

labeling (TUNEL) staining demonstrated enhanced cell death in GluN3A KO mice.

37

Moreover, the deletion of GluN3A hindered sensorimotor functional recovery after stroke.

38

It is suggested that, although the expression level is relatively lower in the adult brain,

39

GluN3A is still a noteworthy regulator in ischemia-induced excitotoxicity and brain

40

injury.

41 42

Key words: Stroke, NMDA receptors, GluN3A, Adult brain, Functional recovery

43

2

44

Abbreviations

45

NMDA: N-methyl-D-aspartate

46

NMDAR: NMDA receptor

47

MCA: middle cerebral artery

48

CCAs: common carotid arteries

49

WT: wild type

50

KO: knockout

51

GluN1: glutamate NMDA receptor 1 (NR1)

52

GluN2: glutamate NMDA receptor 2A (NR2A)

53

GluN2B: glutamate NMDA receptor 2B (NR2B)

54

GluN3A: glutamate NMDA receptor 3A (NR3A)

55

GluN3B: glutamate NMDA receptor 3B (NR3B)

56

TTC: 2,3,5-triphenyltetrazolium chloride

57

TUNEL: terminal deoxynucleotidyl transferase dUTP nick end labeling

3

58 59

Introduction The N-methyl-D-aspartate subtype of glutamate receptor (NMDAR) serves

60

critical roles in physiological and pathological processes in the central nervous system

61

(CNS) (7, 9, 17, 20). The receptor activity regulates fast excitatory neurotransmission,

62

contribute to the regulation of neuronal gene expression and play key roles in the

63

formation of neural networks and the formation of long-term potentiation (LTP) and

64

memory (18). NMDAR is composed of proteins derived from the GluR1 (NR1) family

65

and the GluN2 (NR2) family of genes. The GluN1 family has a variety of splice variants,

66

and GluN2 family is comprised of four subunit classes, GluN2A, GluN2B, GluN2C and

67

GluN2D. The gene expression of both families shows temporal and spatial distributions

68

in the brain (13, 15, 22). NMDAR may also contain an inhibitory subunit, GluN3A

69

(NR3A, NMDAR-L, or chi-1) and GluN3B (NR3B) (2, 6, 19, 26). Incorporation of

70

GluN3A with GluN1/GluN2 subunits suppresses NMDAR activity. For example, co-

71

expression of GluN3A results in decreased NMDAR single channel conductance, Ca2+

72

permeability and the receptor Mg2+ sensitivity, which may limit synaptic strengthening

73

and stabilization (3, 6, 8, 25, 26). On the other hand, genetic knockout (KO) of GluN3A

74

in mice results in enhanced NMDA currents and increased dendritic spines in early

75

postnatal cerebrocortical neurons compared to wild type (WT) mice (8). In addition,

76

increases of intracellular Ca2+ concentration in the neuronal cells from GluN3A KO mice

77

contribute to cell death (3, 5, 14).

78

It is well known that GluN3A is a developmentally regulated NMDAR subunit.

79

GluN3A mRNA and protein levels peak at the end of the first postnatal week but

80

decreases to a low level by adulthood (6, 26). Based on these findings, studies on

4

81

GluN3A have been almost exclusively focused on the developing brain of neonates (26,

82

30). In a hypoxic-ischemic stroke model of neonatal mice, deletion of GluN3A caused

83

enlarged infarct formation while over-expression of GluN3A decreased the brain damage

84

(24). On the other hand, the significance of endogenous GluN3A in the adult brain has

85

been largely ignored, most likely due to the fact that the GluN3A level is down-regulated

86

in the adult brain and due to the assumption that the low level GluN3A has no or little

87

functional role. The pathological role of GluN3A in the adult brain, ironically, has never

88

been directly tested and no quantified experimental data from adult animals has been

89

reported before.

90

We recently demonstrated that even though the GluN3A expression level declines

91

to lower levels from the neonatal period to the adulthood, significant protein levels of

92

GluN3A are still detectable in the adult brain (21). An important role of GluN3A in

93

locomotion, pain perception, and cognitive functions was observed in adult animals (21).

94

The results indicate that even if at relatively lower expression level, GluN3A still can act

95

as an imperative player in the adult brain. In the present investigation, we extended the

96

examination into the pathological condition of ischemic stroke and tested the hypothesis

97

that a role of GluN3A in the adult stroke brain might have been overlooked before. We

98

also hypothesized that a neuroprotective role of GluN3A in the adult brain could indicate

99

a receptor subunit target for the development of therapeutic treatment of stroke.

100 101

Methods

102

Animals

103

NMDA receptor GluN3A (NR3A) constitutive knockout (KO) mice (kindly

5

104

provided by Dr. Stuart A. Lipton and Dr. Nobuki Nakanishi at Stanford University) and

105

littermate wild type (WT) mice at different postnatal days were used in the experiments.

106

The animals were maintained at a constant temperature of 21°C with a 12 hrs light/dark

107

cycle in the Laboratory Animal Center for Research, Medicine School of South Carolina.

108

All experimental and surgical procedures were approved by the Institutional Animal Care

109

and Use Committee (IACUC) at Emory University (Animal protocol number: 208-2008).

110

No human tissue or sample was tested in this study.

111 112 113

Genotyping The Genotyping of GluN3A KO and WT animals was performed as described

114

previously (21). DNA for genotyping was extracted from tail snips (approximately 2-4

115

mm). The separately primers were 5’-CCA CGG TGA GCT TGG GGA AG (NN494),

116

5’-GCC TGA AGA ACG AGA TCA GC (NN506) and 5’-TTG GGG AGC GCC CTG

117

CAT GG (NN507) (Sigma-Aldrich, St. Louis, MO, USA). The 251 bp product (NN494

118

and NN506) confutes GluN3A knockout mice and 200 bp product (NN494 and NN507)

119

confutes WT mice. DNA (2 µl) was amplified on a thermocycler (MJ minim, Personal

120

Thermal Cycler, Bio-Rad, CA, USA) for 40 cycles (95°C for 60 sec, 58°C for 30 sec,

121

72°C for 60 sec). After additional incubation at 72°C for 10 min and transferred to 4°C,

122

PCR products were subjected to electrophoresis in 1.5% agarose gel with ethidium

123

bromide. Relative intensity of PCR bands was analyzed using the InGenius3 manual gel

124

documentation system (Syngene, Frederick, MD, USA).

125 126

Neuronal cell cultures

6

127

The preparation of nearly pure cortical neurons was previously described (31).

128

Briefly, neurons were harvested from the cortex of gestational 14–16 day mouse embryos.

129

The pregnant mouse was sacrificed using overdosed isoflurane and brain dissection was

130

performed soon after heartbeat stopped and no response to pawn stimuli. The cortex was

131

dissociated in trypsin and plated on poly-D-lysine and laminin coated tissue culture

132

dishes. Cells were maintained in neurobasal media (Invitrogen, Carlsbad, CA) with B27

133

supplement and L-glutamine until the time of the experiment. Cytarabine (Ara-C) was

134

added on the third day to prevent the proliferation of glial cells. Mature neuronal cultures

135

(12-14 days in vitro (DIV)) were used for cell death assays.

136 137 138

NMDA toxicity and assessment of neuronal cell death Excitotoxicity was induced by 10 min exposure to NMDA (300 µM) alone or in

139

the presence of z-VAD-fmk (100 µM), carried out at room temperature in a HEPES

140

buffer solution (HBSS) containing (in mM): NaCl, 120; KCl, 5.4; MgCl2, 0.8; CaCl2,

141

1.8; HEPES, 20; glucose, 15; glycine, 0.01 (pH 7.4). The exposure solution was then

142

washed away and replaced by media stock (MS) supplemented with 10 µM glycine

143

before returning cultures to the incubator for 20-24 hrs.

144

Neuronal cell death was estimated by examination of the cultures under phase-

145

contrast microscopy, and quantitated by measurement of lactate dehydrogenase (LDH)

146

release by damaged cells into the bathing medium 24 hrs following the experimentation

147

(14). The LDH level corresponding to near-complete neuron death (without glial death)

148

was determined by assaying sister cultures exposed to 300 µM NMDA for 24 hrs in MS

149

supplemented with glycine. The percent neuronal death was normalized to the mean LDH

7

150

value released 24 hrs after a sham wash (defined as 0%) and continuous exposure to 300

151

µM NMDA (defined as 100%). Background LDH levels were determined in sister

152

cultures subjected to sham washing and subtracted from experimental values to yield the

153

signal specific to experimentally induced injury.

154 155 156

Focal cerebral ischemia in mice Focal cerebral ischemia, confined to the sensorimotor cortex in the right middle

157

cerebral artery (MCA) territory of WT and GluN3A KO male mice were induced by

158

permanent occlusions of distal branches of the right MCA and temporary (10 min)

159

ligations of both common carotid arteries (CCAs) (29). In brief, 8–10 week old male

160

mice (25-28 g) were anesthetized by 4% chloral hydrate (100 mg/kg, i.p.). Chloral

161

hydrate has been widely used as a sedation agent in human and animal

162

surgeries/procedures for a number of years. It was chosen in this investigation due to its

163

stable and well controlled anesthetic action and negligible effect on NMDA receptors (4).

164

The right MCA branches were permanently ligated using a 10-0 suture (Surgical

165

Specialties Co., Reading, PA) accompanied by a bilateral 10-min ligation of CCAs. Body

166

temperature was monitored during surgery and maintained at 37.0  0.3°C using a

167

temperature control unit and heating pads. The mortality rate due to ischemic surgery

168

and/or anesthesia failure was around 7% in these experiments. Free access to food and

169

water was allowed after recovery from anesthesia. All mice were kept in air-ventilated

170

cages with room temperature maintained at 24  0.5°C. Animals were euthanized and

171

decapitated at different time points after ischemic stroke. Brains were immediately

172

removed, mounted in optimal cutting temperature compound (Sakura Finetek USA, Inc.,

8

173

Torrance, CA) and stored at −80°C for further processing.

174 175 176

TTC staining of infarct volume measurement Twenty-four hrs after onset of MCAO, animals in different groups were sacrificed

177

for assessment of brain infarct formation. 2,3,5-triphenyltetrazolium chloride (TTC;

178

Sigma-Aldrich) staining was used to reveal damaged/dead brain tissue as previously

179

described (29). Brains were removed and placed in a brain matrix then sliced into 1-mm

180

coronal sections. Slices were incubated in 2% TTC solution at 37°C for 5 min, then

181

stored in 10% buffered formalin for 24 hrs. Digital images of the caudal aspect of each

182

slice were obtained by a flatbed scanner. Infarct, ipsilateral hemisphere, and contralateral

183

hemisphere areas were measured using ImageJ software (NIH, Bethasda, MD, USA). The

184

indirect method (subtraction of residual right hemisphere cortical volume from cortical

185

volume of the intact left hemisphere) will be used for infarct volumes calculation. Infarct

186

measurements were performed under double-blind conditions.

187 188 189

Western blot analysis Western blot analysis was used to detect the expression of NMDAR subunits.

190

Brain cortical tissue was lysed in a lysis buffer containing 0.02 M Na4P2O7, 10 mM Tris-

191

HCl (pH 7.4), 100 mM NaCl, 1 mM EDTA (pH 8.0), 1% Triton, 1 mM EGTA, 2 mM

192

Na3VO4, and a protease inhibitor cocktail (Sigma-Aldrich). The supernatant was

193

collected after centrifugation at 15,000 g for 10 min at 4°C. Protein concentration was

194

determined with a bicinchoninic acid assay (Pierce Biotechnology, Rockford, IL, USA).

195

Equivalent amounts of total protein were separated by molecular weight on an SDS-

9

196

polyacrylamide gradient gel, and then transferred to a PVDF membrane. The blot was

197

incubated in 5% bovine serum albumin for at least 1 hr and then reacted with primary

198

antibodies at 4°C for overnight. The primary antibodies used in this investigation

199

included: and the dilutions for each were rabbit anti-GluN2A antibody (Cell Signaling,

200

Danvers, MA, USA) at 1:2000, rabbit anti-GluN2B antibody (Cell Signaling) at 1:2000,

201

rabbit anti-GluN3A antibody (Abcam, Cambridge, MA, USA) at 1:1000, and mouse anti-

202

-actin (Sigma-Aldrich) at 1:5000 (Supporting Table 1). After washing with Tris-

203

buffered saline with Tween (TBST), membranes were incubated with AP-conjugated or

204

HRP-conjugated secondary antibodies (GE Healthcare, Piscataway, NJ, USA) for 1-2 hrs

205

at room temperature. After final washing with TBST, the signals were detected with

206

bromochloroidolylphosphate/nitroblue tetrazolium (BCIP/NBP) solution (Sigma-Aldrich)

207

or film. Signal intensity was measured by ImageJ and normalized to the actin signal

208

intensity.

209 210 211

Immunohistochemical assessment Frozen brain tissues were sliced into 10 µm-thick coronal sections using a

212

cryostat vibratome (Leica CM 1950; Leica Microsystems, Buffalo Grove, IL, USA).

213

Sections were dried on the slide warmer for 30 min, fixed with 10% formalin buffer,

214

washed with -20°C precooled ethanol: acetic acid (2:1) solution for 10 min, and finally

215

permeabilized with 0.2% Triton-X 100 solution for 5 min. All slides were washed 3 times

216

with PBS (5 min each) after each step. Then, tissue sections were blocked with 1% fish

217

gelatin (Sigma-Aldrich) in PBS for 1 hr at room temperature, and subsequently incubated

218

with the primary antibody: mouse anti-GluN1 antibody (1:200; Life Technologies, Grand

10

219

Island, NY, USA), rabbit anti-GluN2A antibody (1:200; Abcam), rabbit anti-GluN2B

220

(1:200; Abcam), rabbit anti-GluN3A (1:200; Abcam), and mouse anti-NeuN (1:400;

221

Millipore, Billerica, MA, USA) overnight at 4°C (Supporting Table 1). Next day, the

222

slides were washed 3 times with PBS for 5 min, then reacted with the secondary

223

antibodies Alexa Fluor®488 goat anti-mouse or rabbit (1:300; Life Technologies) and

224

Cy3-conjugated donkey anti-rabbit (1:300; Jackson ImmunoResearch Laboratories, West

225

Grove, PA, USA) or Cy5-conjugated donkey anti-mouse or rabbit (1:400; Jackson

226

ImmunoResearch Laboratories) for 80 min at room temperature. After 3 washes with

227

PBS, nuclei were stained with Hoechst 33342 (1:20,000; Life Technologies) for 5 min as

228

a counterstain. And then the brain sections were mounted, coverslipped, imaged, and

229

photographed under a fluorescent microscope (BX51, Olympus, Japan) and laser

230

scanning confocal microscope (Carl Zeiss Microimaging, Inc., Thornwood, NY, USA).

231 232

TUNEL staining and positive cell quantification

233

A terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay

234

kit was used to examine cell death by detecting fragmented DNA in 10-µm-thick coronal

235

fresh frozen sections as described previously (n=8 per group) (16). After fixation (10%

236

buffered formalin for 10 min and then ethanol:acetic acid (2:1) solution for 5 min) and

237

permeabilization (0.2% Triton X-100 solution), brain sections were incubated in the

238

equilibration buffer for 10 min. Recombinant terminal deoxynucleotidyl transferase

239

(rTdT) and nucleotide mixture were then added on the slide at 37°C for 60 min in the

240

dark. Reactions were terminated by 2x SSC solution for 15 min. Nuclei were

241

counterstained with Hoechst 33342 (1:20,000; Molecular Probes) for 5 min.

11

242 243 244

Cell counting Cell counting was performed following the principles of design based stereology

245

(16). Systematic random sampling was employed to ensure accurate and non-redundant

246

cell count. Every section under analysis was at least 90 µm apart from the next. Three 10

247

µm thick sections spanning the entire region of interest were selected for cell

248

quantification. Counting was performed on 5 to 8 non-overlapping randomly selected 40x

249

fields per section in the specified region of the cortex. ImageJ (NIH) was used to analyze

250

each picture. All analysis was performed in a double-blinded fashion.

251 252 253

Adhesive removal test The adhesive removal test measures sensorimotor function as previously

254

described (1, 10). Briefly, a small adhesive dot was placed on each forepaw, and the

255

amount of time (seconds) needed to contact and remove the tape from each forepaw was

256

recorded. Mice were trained three times for three days (1–2 trials per day) before

257

ischemia induction to ensure that they were able to remove the tape within 12 seconds to

258

get a basal level of performance. Mice that show no response or prolonged time to

259

remove the tape (>20 sec) during training period were excluded from future experiments.

260

Animals were tested before ischemia and 3 and 7 days after ischemia by an investigator

261

who was blind to the experimental groups. The test was performed four times per mouse,

262

and the average time was used in the analysis. In this behavioral study, 16 animals were

263

randomly assigned to WT and GluN3A KO groups before ischemia and 3 days after

12

264

ischemia. At 7 days after stroke, 6 animals were used in each group. All testing trials

265

were conducted during the day time.

266 267 268

Locomotor activity test Behavioral changes of experimental rats were monitored and analyzed using the

269

TopScan System (Clever Sys Inc., Reston, VA, USA). Mice were allowed to freely

270

move in an open field container (50 cm × 50 cm × 50 cm). For locomotor activity,

271

travelled distance and velocity were recorded for 1 hr before surgery and at 3 and 7 days

272

after focal cerebral ischemia. After the recording, the videos were analyzed by the

273

TopScan Realtime Option Version 3.0 (Clever Sys Inc.).

274 275 276

Cylinder test A unilateral injury to the motor cortex results in an asymmetry in the forelimb

277

used for support the body during rearing, which can be measured using the cylinder test.

278

The mice were placed in a glass cylinder (9.5 cm diameter and 11 cm height), and the

279

number of times each forelimb or both forelimbs were used to support the body on the

280

wall of the cylinder was counted for 5 min. The animals were evaluated before surgery

281

and at 3 and 7 days after focal cerebral ischemia. Two mirrors were placed behind the

282

cylinder to view all directions. The numbers of impaired and non-impaired forelimb

283

contacts were calculated as a percentage of total contacts.

284 285

Statistical Analysis

13

286

All results are expressed as mean ± S.E.M. Statistical comparisons, using Graph

287

Pad Prism 6 (Graph Pad Software, Inc., San Diego, CA), were analyzed by unpaired

288

Student’s t-test and Fisher’s test with least significant difference to identify significant

289

differences. P < 0.05 was considered significant for all comparisons.

290 291 292

Results

293

GluN3A expression patterns and cellular distribution in the mouse brain

294

To characterize the expression of GluN3A, we measured the expression level of

295

GluN3A at postnatal day 5, 15, 30, and 70 in WT mice using Western blot analysis. As

296

reported before, the highest level of GluN3A was observed in the P5 brain and the

297

GluN3A expression was then declined to lower levels in the following days (Fig. 1A and

298

1B). However, even if the GluN3A level was relatively lower in the adult brain (P30-

299

P70) than that in the neonatal brain (P5-P15), the GluN3A expression was readily

300

detectable. In WT and GluN3A KO brains, Western blotting confirmed that this NMDAR

301

subunit was largely eliminated or undetectable in the KO brain tissues (Fig. 1D).

302

Immunohistochemical analysis revealed that GluN3A was observed in the neuron of

303

cortex including sensorimotor cortex from WT adult brain (Fig. 1C). We and others

304

showed before that deletion of GluN3A does not affect the expression levels of GluN1

305

and GluN2 (21, 24). Since GluN2 subunits especially GluN2B are specifically linked to

306

excitotoxicity (27), we measured the GluN2A and GluN2B protein levels in the adult WT

307

and GluN3A KO brains and the expression in cortical neurons. There was no change in

308

GluN2A and GluN2B levels in the absence of GluN3A (Fig. 1D and 1E). Also, the

14

309

number of GluN2B-postive cells didn’t show any difference between WT and GluN3A

310

KO mice (Fig. 1F).

311

Using immunocytochemical staining, we specifically examined the cellular

312

distribution of GluN3A in cultured cortical neurons. It was clear that GluN3A localized

313

to the cell membrane and dendrites (Fig. 2A-2C). Moreover, they were colocalized with

314

the GluN1 subunit, suggesting formation of functional NMDARs in these locations (Fig.

315

2D).

316 317

Neuroprotective effects of GluN3A expression against in vitro ischemic insult in

318

cortical cultures

319

We next examined NMDA-induced neuronal cell death in cortical neuronal

320

cultures of 12-14 DIV. The neuronal culture was exposed to 300 µM NMDA for 10 min

321

and washed out with normal culture media. This short-term NMDA insult caused marked

322

cell death 24 hrs later. The neuronal cell death showed apoptotic features such as cell

323

body shrinkage and sensitivity to the block by the caspase inhibitor z-VAD-fmk (Fig. 3A

324

and 3B). Compared to WT neurons, GluN3A KO neurons showed enhanced vulnerability

325

to the NMDA insult, there was significant more neuronal death in GluN3A KO cultures

326

(Fig. 3B).

327 328

Neuroprotective effects of GluN3A expression against ischemic damage in the adult

329

brain

330 331

The focal cerebral ischemia in our experiments selectively damaged the somatosensory cortex of adult mice. Twenty four hrs after the ischemic insult, brain

15

332

sections were stained with TTC solution to identify the damaged/died brain tissue (Fig.

333

4A). The infarction volume and ratio of the cerebral cortex in GluN3A KO mice were

334

significantly larger than that in WT mice (Fig. 4B and 4C). Using Western blotting, we

335

compared the levels of GluN3A before and after ischemia in WT mice and did not

336

observe significant change (data not shown).

337

To confirm the difference in the ischemia-induced injury between the two groups

338

at cellular levels, TUNEL staining was applied to evaluate the cell death in the penumbra

339

region 24 hrs after focal cerebral ischemia. The number of dead/injured cells measured as

340

TUNEL-positive cells was noticeable higher in GluN3A brain than that in WT stroke

341

mice (Fig. 4D and 4E).

342 343 344

Effect of GluN3A on functional recovery after focal cerebral ischemia Several behavior tests measured the functional outcomes after focal cerebral

345

ischemia. In the adhesive dot removal test, the latency times to contact and remove the

346

sticky dot from the right and left forelimbs were recorded at 3 and 7 days after focal

347

cerebral ischemia (Fig. 5A and 5B). Before ischemic insult, the contact and removal

348

times were similar between WT and GluN3A KO mice. After focal cerebral ischemia,

349

animals showed prolonged times in response to the sticky dot attached to their left paws

350

due to impaired sensorimotor function on the right side of the sensorimotor cortical area.

351

GluN3A KO mice showed significantly hindered performance at 3 days after ischemia on

352

both time to contact and time to remove sticky dot activities (Fig. 5A and 5B). TopScan

353

system analysis was used to measure the locomotor activity after focal cerebral ischemia.

354

GluN3A KO mice showed significant less travelled distance and lower velocity than WT

16

355

mice at 3 days after focal cerebral ischemia (Fig. 5C and 5D). Consistently, GluN3A KO

356

mice showed significant worse performance in cylinder test at 3 days after focal cerebral

357

ischemia (Fig. 5E).

358 359 360 361

Discussion This investigation reveals that, event at a relatively lower level than in the

362

neonatal brain, the NMDAR inhibitory subunit GluN3A in the adult brain exhibits a

363

neuroprotective effect in ischemic stroke. We demonstrate that deletion of this subunit

364

results in enlarged infarct volume and increased cell death in the ischemic cortex of the

365

adult mouse brain. Also, GluN3A KO mice exhibited hindered functional recovery

366

compared to WT mice. These observations, on one hand, are surprising based on the

367

common belief that GluN3A has little, if any, functional role in the adult brain due to its

368

low expression level (26, 30). On the other hand, the result is consistent with our previous

369

investigation showing that GluN3A plays an imperative role in pain perception,

370

locomotion, and cognitive function in adult mice (21). Taking together, it is suggested

371

that although the expression level of GluN3A in the adult brain is down-regulated from

372

its peak level at the neonatal stage, this subunit in the adult brain still have significant

373

impacts on multiple brain activities under physiological and pathological conditions such

374

as in ischemic stroke. We noticed that the difference in the infarct volume between WT

375

and GluN3A KO mice is relative moderate, which is consistent with the low level of

376

GluN3A in the adult brain. The present investigation, however, is not an investigation of

377

stroke treatment, which is largely judged by how big the protective effect is. Instead, we

17

378

aimed to reveal that whether GluN3A could play a functional role in cerebral ischemia of

379

the adult brain. The observation provides important information for a previously

380

unidentified function of the GluN3A subunit after ischemic stroke in adults. It is expected

381

that genetic manipulations and other strategies of increasing the GluN3A level in the

382

adult brain can amplify the endogenous defense mechanism against ischemic stroke.

383

A neuroprotective role of the GluN3A subunit was demonstrated in an earlier

384

investigation in cortical cultures subjected to excitotoxic insults and in a neonatal

385

hypoxia-ischemia model of mice (24). It was mentioned in the abstract of the report that a

386

neuroprotective effect against ischemic stroke was only observed in neonatal but not in

387

adult GluN3A KO mice (24). Unfortunately, no experimental data on the adult stroke

388

experiment was presented in the report (24). On the other hand, the authors did show a

389

neuroprotective effect of the GluN3A subunit in adult animals subjected to NMDA-

390

induced retina injury and in the GluN3A-overexpressing transgenic mice after transient

391

ischemic stroke (24). The protective effect of overexpressing GluN3A was also shown in

392

a recent study on cultured hippocampal neuronal cell death induced by oxygen glucose

393

deprivation (OGD) (28). We show in adult animals that the basal level of this subunit

394

already shows a moderate neuroprotection, supporting that increasing the GluN3A level

395

can be an effective therapeutic target for the treatment of stroke.

396

Recent studies raise an intriguing possibility that NMDAR hypofunction in

397

schizophrenia and bipolar disorder may arise from an excess increase or decrease of

398

GluN3A expression. GluN3A mRNA levels are significantly elevated in the dorsolateral

399

prefrontal cortex of schizophrenic patients and decreased in that of bipolar disorder

400

patients (23). Altered GluN3A expression levels regulate the development of spine

18

401

densities in schizophrenic patients (11, 12). Taken together, these findings suggest that

402

GluN3A expression can be an important regulator on the development of schizophrenia

403

and bipolar disorder and a key target gene for treating the patients with psychotic

404

disorders.

405

In conclusion, the present study shows a NMDAR subunit-mediated endogenous

406

neuroprotective action after focal cerebral ischemia in the adult brain. Together with the

407

previous reports of the GluN3A regulation on pain sensation, cognitive activities,

408

locomotion and retina excitotoxicity in adult animals (21, 24), the expression of GluN3A

409

thus shows significant physiological and pathophysiological roles in adults. In this study,

410

we used conventional GluN3A KO mice. It is possible that the loss of GluN3A activity

411

during the development stage may mask and change the role of the gene in the adult stage.

412

Therefore, further studies using conditional KO approach may be needed to provide more

413

inside information on the developmentally regulated neuroprotective mechanism.

414

19

415 416

Acknowledgement This work was supported by the NIH grants NS062097, NS073378, NS075338, a VA

417

Merit grant, an American Heart Association Grant-in-Aid grant 12GRNT12060222 and an AHA

418

Established Investigator Award.

419 420 421

Disclosures The authors have no disclosure of conflicts of interest related to this investigation.

422

20

423

References

424

1. Bouet V, Boulouard M, Toutain J, Divoux D, Bernaudin M, Schumann-Bard P,

425

and Freret T. The adhesive removal test: a sensitive method to assess sensorimotor

426

deficits in mice. Nat Protoc 4: 1560-1564, 2009.

427

2. Chatterton JE, Awobuluyi M, Premkumar LS, Takahashi H, Talantova M, Shin

428

Y, Cui J, Tu S, Sevarino KA, Nakanishi N, Tong G, Lipton SA, and Zhang D.

429

Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits.

430

Nature 415: 793-798, 2002.

431 432 433

3. Chen ST, Hsu CY, Hogan EL, Juan HY, Banik NL, and Balentine JD. Brain calcium content in ischemic infarction. Neurology 37: 1227-1229, 1987. 4. Chizh BA. Low dose ketamine: a therapeutic and research tool to explore N-methyl-

434

D-aspartate (NMDA) receptor-mediated plasticity in pain pathways. Journal of

435

psychopharmacology 21: 259-271, 2007.

436 437 438

5. Choi DW, and Rothman SM. The role of glutamate neurotoxicity in hypoxicischemic neuronal death. Annu Rev Neurosci 13: 171-182, 1990. 6. Ciabarra AM, Sullivan JM, Gahn LG, Pecht G, Heinemann S, and Sevarino KA.

439

Cloning and characterization of chi-1: a developmentally regulated member of a

440

novel class of the ionotropic glutamate receptor family. J Neurosci 15: 6498-6508,

441

1995.

442 443

7. Cull-Candy S, Brickley S, and Farrant M. NMDA receptor subunits: diversity, development and disease. Curr Opin Neurobiol 11: 327-335, 2001.

444

8. Das S, Sasaki YF, Rothe T, Premkumar LS, Takasu M, Crandall JE, Dikkes P,

445

Conner DA, Rayudu PV, Cheung W, Chen HS, Lipton SA, and Nakanishi N.

21

446

Increased NMDA current and spine density in mice lacking the NMDA receptor

447

subunit NR3A. Nature 393: 377-381, 1998.

448 449 450

9. Dingledine R, Borges K, Bowie D, and Traynelis SF. The glutamate receptor ion channels. Pharmacol Rev 51: 7-61, 1999. 10. Freret T, Bouet V, Leconte C, Roussel S, Chazalviel L, Divoux D, Schumann-

451

Bard P, and Boulouard M. Behavioral deficits after distal focal cerebral ischemia in

452

mice: Usefulness of adhesive removal test. Behav Neurosci 123: 224-230, 2009.

453

11. Glantz LA, and Lewis DA. Decreased dendritic spine density on prefrontal cortical

454 455 456 457

pyramidal neurons in schizophrenia. Arch Gen Psychiatry 57: 65-73, 2000. 12. Glantz LA, and Lewis DA. Dendritic spine density in schizophrenia and depression. Arch Gen Psychiatry 58: 203, 2001. 13. Goebel DJ, and Poosch MS. NMDA receptor subunit gene expression in the rat

458

brain: a quantitative analysis of endogenous mRNA levels of NR1Com, NR2A,

459

NR2B, NR2C, NR2D and NR3A. Brain Res Mol Brain Res 69: 164-170, 1999.

460

14. Koh JY, and Choi DW. Vulnerability of cultured cortical neurons to damage by

461

excitotoxins: differential susceptibility of neurons containing NADPH-diaphorase. J

462

Neurosci 8: 2153-2163, 1988.

463 464 465

15. Laurie DJ, and Seeburg PH. Regional and developmental heterogeneity in splicing of the rat brain NMDAR1 mRNA. J Neurosci 14: 3180-3194, 1994. 16. Lee JH, Wei L, Gu X, Wei Z, Dix TA, and Yu SP. Therapeutic effects of

466

pharmacologically induced hypothermia against traumatic brain injury in mice. J

467

Neurotrauma 31:1417-1430, 2014.

22

468 469 470 471

17. Lipton SA, and Rosenberg PA. Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med 330: 613-622, 1994. 18. Ma OK, and Sucher NJ. Molecular interaction of NMDA receptor subunit NR3A with protein phosphatase 2A. Neuroreport 15: 1447-1450, 2004.

472

19. Matsuda K, Kamiya Y, Matsuda S, and Yuzaki M. Cloning and characterization of

473

a novel NMDA receptor subunit NR3B: a dominant subunit that reduces calcium

474

permeability. Brain Res Mol Brain Res 100: 43-52, 2002.

475 476

20. McBain CJ, and Mayer ML. N-methyl-D-aspartic acid receptor structure and function. Physiol Rev 74: 723-760, 1994.

477

21. Mohamad O, Song M, Wei L, and Yu SP. Regulatory roles of the NMDA receptor

478

GluN3A subunit in locomotion, pain perception and cognitive functions in adult

479

mice. The Journal of physiology 591: 149-168, 2013.

480

22. Monyer H, Burnashev N, Laurie DJ, Sakmann B, and Seeburg PH.

481

Developmental and regional expression in the rat brain and functional properties of

482

four NMDA receptors. Neuron 12: 529-540, 1994.

483

23. Mueller HT, and Meador-Woodruff JH. NR3A NMDA receptor subunit mRNA

484

expression in schizophrenia, depression and bipolar disorder. Schizophr Res 71: 361-

485

370, 2004.

486

24. Nakanishi N, Tu S, Shin Y, Cui J, Kurokawa T, Zhang D, Chen HS, Tong G,

487

and Lipton SA. Neuroprotection by the NR3A subunit of the NMDA receptor. J

488

Neurosci 29: 5260-5265, 2009.

489 490

25. Roberts AC, Diez-Garcia J, Rodriguiz RM, Lopez IP, Lujan R, MartinezTurrillas R, Pico E, Henson MA, Bernardo DR, Jarrett TM, Clendeninn DJ,

23

491

Lopez-Mascaraque L, Feng G, Lo DC, Wesseling JF, Wetsel WC, Philpot BD,

492

and Perez-Otano I. Downregulation of NR3A-containing NMDARs is required for

493

synapse maturation and memory consolidation. Neuron 63: 342-356, 2009.

494

26. Sucher NJ, Akbarian S, Chi CL, Leclerc CL, Awobuluyi M, Deitcher DL, Wu

495

MK, Yuan JP, Jones EG, and Lipton SA. Developmental and regional expression

496

pattern of a novel NMDA receptor-like subunit (NMDAR-L) in the rodent brain. J

497

Neurosci 15: 6509-6520, 1995.

498

27. von Engelhardt J, Coserea I, Pawlak V, Fuchs EC, Kohr G, Seeburg PH, and

499

Monyer H. Excitotoxicity in vitro by NR2A- and NR2B-containing NMDA

500

receptors. Neuropharmacology 53: 10-17, 2007.

501

28. Wang H, Yan H, Zhang S, Wei X, Zheng J, and Li J. The GluN3A subunit exerts a

502

neuroprotective effect in brain ischemia and the hypoxia process. ASN Neuro 5: 231-

503

242, 2013.

504

29. Wang LL, Chen D, Lee J, Gu X, Alaaeddine G, Li J, Wei L, and Yu SP.

505

Mobilization of endogenous bone marrow derived endothelial progenitor cells and

506

therapeutic potential of parathyroid hormone after ischemic stroke in mice. PloS one

507

9: e87284, 2014.

508

30. Wong HK, Liu XB, Matos MF, Chan SF, Perez-Otano I, Boysen M, Cui J,

509

Nakanishi N, Trimmer JS, Jones EG, Lipton SA, and Sucher NJ. Temporal and

510

regional expression of NMDA receptor subunit NR3A in the mammalian brain. J

511

Comp Neurol 450: 303-317, 2002.

24

512

31. Xiao AY, Wei L, Xia S, Rothman S, and Yu SP. Ionic mechanism of ouabain-

513

induced concurrent apoptosis and necrosis in individual cultured cortical neurons. J

514

Neurosci 22: 1350-1362, 2002.

515 516 517

25

518

Figure Legends

519

Figure 1. Expression patterns of GluN3A in the mouse brain

520

Expression patterns of GluN3A and its effect on other NMDAR subunits were

521

examined using Western blot analysis and immunohistochemical method in the mouse

522

brain. A and B. The peak level of GluN3A was detected in the P5 brain and its expression

523

declines to lower levels in the following days. However, GluN3A expression was still

524

detectable in even P30 and P70 brain. N = 4-5 animals in each group; * P

A neuroprotective role of the NMDA receptor subunit GluN3A (NR3A) in ischemic stroke of the adult mouse.

GluN3A or NR3A is a developmentally regulated N-methyl-d-aspartate receptor (NMDAR) subunit, showing a unique inhibitory role that decreases NMDAR cur...
1MB Sizes 1 Downloads 6 Views