• Nem Talált Eredményt

INTRODUCTION CEREBRALARTERYOCCLUSION EXACERBATESNEURONALDAMAGEAFTERTRANSIENTMIDDLE POST-ISCHEMICTREATMENTWITHL-KYNURENINESULFATE

N/A
N/A
Protected

Academic year: 2022

Ossza meg "INTRODUCTION CEREBRALARTERYOCCLUSION EXACERBATESNEURONALDAMAGEAFTERTRANSIENTMIDDLE POST-ISCHEMICTREATMENTWITHL-KYNURENINESULFATE"

Copied!
7
0
0

Teljes szövegt

(1)

1

2

POST-ISCHEMIC TREATMENT WITH L-KYNURENINE SULFATE

3

EXACERBATES NEURONAL DAMAGE AFTER TRANSIENT MIDDLE

4

CEREBRAL ARTERY OCCLUSION

5 L.GELLE´RT,aL.KNAPP,aK.NE´METH,bJ.HERE´DI,a

6 D.VARGA,aG.OLA´H,aK.KOCSIS,aA´.MENYHA´RT,a

7 Z.KIS,aT.FARKAS,aL.VE´CSEIc,dANDJ.TOLDIa*

8 aDepartment of Physiology, Anatomy and Neuroscience, 9 University of Szeged, Ko¨ze´p fasor 52, H-6726 Szeged, Hungary 10 bDepartment of Cognitive Science, University of Technology and 11 Economics, Egry Jo´zsef utca 1, T building V. 506, H-1111 Budapest, 12 Hungary

13 cDepartment of Neurology, University of Szeged, Semmelweis u.

14 6, H-6725 Szeged, Hungary

15 dNeurology Research Group of the Hungarian Academy of 16 Science and University ofSzeged, Hungary

17 Abstract—Since brain ischemia is one of the leading causes of adult disability and death, neuroprotection of the ische- mic brain is of particular importance. Acute neuroprotective strategies usually have the aim of suppressing glutamate excitotoxicity and an excessive N-methyl-D-aspartate (NMDA)receptor function. Clinically tolerated antagonists should antagonize an excessive NMDA receptor function without compromising the normal synaptic function.

Kynurenic acid (KYNA) an endogenous metabolite of the tryptophan metabolism, may be an attractive neuroprotec- tant in this regard. The manipulation of brain KYNA levels was earlier found to effectively enhance thehistopatholo- gical outcome of experimental ischemic/hypoxic states.

The present investigation of the neuroprotective capacity of L-kynurenine sulfate (L-KYNs) administered systemically after reperfusion in a novel distal middle cerebral artery occlusion (dMCAO) model of focal ischemia/reperfusion revealed that in contrast with earlier results, treatment with L-KYNs worsened the histopathological outcome of dMCAO. This contradictory result indicates that post-ische- mic treatment with L-KYNs may be harmful.Ó2013 Published by Elsevier Ltd. on behalf of IBRO.

Key words: focalcerebral ischemia,neuroprotection,glycine co-agonist site, NMDAR, MCAO model,kynurenines.

18

INTRODUCTION 19

As a result of the high energy demands of the central 20

nervous system (CNS), a deprivation of oxygen and 21

glucose leads in a short time to abnormal glutamatergic 22

transmission. Malfunctioning of the ATP-dependent 23

transporters results in a disturbance of ionic 24

homeostasis, depolarization and the excessive release 25

of glutamate from neural and glial stores in the 26

extrasynaptic space. Acute or prolonged over-activation 27

ofN-methyl-D-aspartate receptors (NMDARs) allows the 28

excessive entry of Ca2+, initiating glutamate 29

excitotoxicity, the common core feature of many 30

neuropsychiatric disorders, including stroke, epilepsy, 31

Alzheimer’s disease and Huntington’s disease (Endres 32

and Dirnagl, 2002; Moskowitz et al., 2010). 33

Neuroprotective strategies usually have the aim of 34

suppressing an excessive NMDAR function. Indeed, a 35

number of NMDA antagonists have proven to be robust 36

neuroprotectants in animal models of an ischemic/ 37

hypoxic (I/H) state, but many failed in clinical trials in 38

consequence of their adverse side-effects (Ikonomidou 39

and Turski, 2002; Muir, 2006). 40

The destructive effect of NMDAR over-activity is in 41

contrast with the phenomenon that synaptic NMDAR 42

activity mediates the survival of several types of 43

neurons (Hetman and Kharebava, 2006; Hardingham, 44

2009). It has been reported that neurodegeneration in 45

the basal ganglia is exacerbated by NMDAR antagonists 46

(Ikonomidou et al., 2000), that an NMDAR antagonist 47

enhanced apoptotic cell loss in a head trauma model 48

(Pohl et al., 1999), and that synaptic NMDAR activity 49

boosts intrinsic antioxidant defenses (Papadia et al., 50

2008). Furthermore, the targeting of ischemic brain 51

areas by global NMDAR antagonism can confuse the 52

functioning of brain areas unaffected by ischemic 53

damage (Gunduz-Bruce, 2009). In this regard, a 54

clinically tolerated neuroprotectant should antagonize 55

the NMDAR function when it is excessive, but not later, 56

without compromising the normal synaptic function. 57

NMDAR activation requires the definite depolarization 58

of the cell and the presence of both glutamate and the full 59

co-agonists glycine or D-serine (Kussius and Popescu, 60

2009; Papouin et al., 2012). Furthermore, the glycine 61

co-agonist site is not saturated under physiological 62

conditions, but is in a hyperactive state (Li et al., 2009; 63

Fuchs et al., 2012). Glycine-site antagonists may be 64

attractive neuroprotectants in this respect. 65

0306-4522/13 $36.00Ó2013 Published by Elsevier Ltd. on behalf of IBRO.

http://dx.doi.org/10.1016/j.neuroscience.2013.04.063

*Corresponding author. Tel: +36-62-544153; fax: +36-63-544291.

E-mail address:toldi@bio.u-szeged.hu(J. Toldi).

Abbreviations: 3-HK, 3-hydroxykynurenine; CNS, central nervous system; dMCA, distal middle cerebral artery; dMCAO, distal middle cerebral artery occlusion; EEG, electroencephalography; I/H, ischemic/

hypoxic; KYNA, kynurenic acid; L-KYNs, L-kynurenine sulfate; NDS, normal donkey serum; NMDA, N-methyl-D-aspartate; NMDAR, N-methyl-D-aspartate receptors; PB, phosphate buffer; QUIN, quinolinic acid.

Q2 Q1

Neurosciencexxx (2013) xxx–xxx

1

(2)

66 Kynurenic acid (KYNA) is an endogenous metabolite

67 of the tryptophan metabolism. It is produced from its

68 precursor L-kynurenine (KYN) by the enzyme

69 kynurenine-aminotransferase II (KATII), and discharged

70 from the astrocytes in the CNS (Swartz et al., 1990).

71 KYNA is a competitive antagonist at theglycine/D-serine

72 co-agonist site of the NMDAR. Furthermore, it plays a

73 versatile role in pathological states, including

74 inflammatory (Moroni et al., 2012), vascular (Sas et al.,

75 2003) and antioxidant (Lugo-Huitron et al.) processes.

76 Acting on thea7 nicotinic acetylcholine receptor, KYNA

77 also influences the excitability of neurons (Banerjee et

78 al.). A huge body of evidence indicates that

79 manipulation of the brain KYNA levels can effectively

80 ameliorate the histopathological outcome of

81 experimental I/H state (Stone, 2000; Wu et al., 2000;

82 Schwarcz and Pellicciari, 2002; Stone and Addae, 2002;

83 Vamos et al., 2009; Zadori et al., 2009). The

84 neuromodulatory properties of KYNA are now well-

85 established (Vecsei et al., 2012).

86 In the present study, we investigated whether

87 L-kynurenine sulfate (L-KYNs) administered after

88 reperfusion (in a dose, formerly proved to be

89 neuroprotective) diminishes the neuronal damage

90 triggered by short-term occlusion of the distal middle

91 cerebral artery (dMCA) in the rat cerebral

92 somatosensory cortex. This novel dMCA occlusion

93 (dMCAO) model was recently developed and

94 characterized from histological and electrophysiological

95 aspects in our research group (L. Knapp, manuscript

96 under review).

97 EXPERIMENTAL PROCEDURES

98 Animals

99 Male Wistar rats (n= 23) weighing 200–250g were

100 used. The animals were kept under controlled laboratory

101 conditions with free access to food and water. The

102 experiments were carried out in accordance with the

103 protocol for animal care approved by both the

104 Hungarian Health Committee (1998) and the European

105 Communities Council Directive (86/609/EEC).

106 Surgical procedure

107 Experiments were carried out under Nembutal

108 anesthesia. The body temperature was maintained at

109 37 ± 0.5°C with a self-regulating heating pad and rectal

110 probe (Supertech TMP-5a). The animals were fixed in a

111 stereotaxic headholder (David Kopf Instr.) and the left

112 masticatory muscle was removed. The surface of the

113 temporal skull was cleaned and the brain was exposed

114 with a high-speed microdrill. The exposed cortical

115 surface involved the trunk and main branches of the

116 MCA. To induce ischemia, the MCA was carefully lifted

117 through 1200lm with a Fisher microsurgery hook with

118 the aid of a micromanipulator, and occluded for 30 min.

119 To terminate the occlusion, the hook was carefully

120 removed, and restoration of the blood flow was

121 confirmed under an operating microscope. Finally, the

122 dura and the temporal muscle were replaced, the skin

was closed with a silk suture and the wound was 123

cleaned with iodine solution. All interventions were 124

strictly synchronized in time, to make the effect of 125

Nembutal on the experiment uniform. 126

Electrophysiology 127

60sofelectroencephalography (EEG) was recorded on 128

the surface of the skull with a silver electrode (2 mm 129

lateral to the sutura sagittalis and 3 mm behind the 130

bregma), promptly before and in the29th–30thmin after 131

dMCAO (sampling rate: 1024 Hz; gain: 1000) with 132

Experimetria NeuroSys software (Experimetria Ltd., 133

Hungary). 134

EEG power analysis was performed with the EEGLab 135

toolbox (Delorme and Makeig, 2004) and custom-written 136

MATLAB 7.1 (Mathworks, Natick, Massachusetts, USA) 137

software. 138

The range of frequency of interest was assigned to 139

2–20Hz and further analysis was performed within this 140

range. 141

Histology 142

Tissue processing. For the histological study, 5 days 143

after dMCAO, animals were anesthetized with an 144

overdose of urethane and perfused transcardially with 145

ice-cold phosphate buffer (PB, 0.1 M, pH 7.4) and 4% 146

paraformaldehyde (dissolved in 0.1 M PB, pH 7.4). The 147

brains were removed and postfixed overnight in 148

paraformaldehyde. On the next day, 20-lm coronal 149

sections were obtained with a vibratome (Leica VT1000 150

S) between0.5 and 4 mm behind the bregma (Paxinos 151

et al., 1980). Two adjacent slices were collected in 152

500-lm steps, one for double immunostaining and the 153

other for Fluoro Jade-C staining. Fluorescent 154

photomicrographs were obtained with an Olympus BX51 155

microscope fitted with a DP70 digital imaging system. 156

Fluoro Jade-C staining. Fluoro Jade-C (FJ-C) staining 157

was performed with the literature protocol (Schmued 158

et al., 2005) with some modification. The slices were 159

mounted on gelatine-coated slides, then coverslipped 160

with Fluoromount. FJ-C-positive (FJ-C+) cells were 161

counted in the ispilateral cortex at 40 magnification. 162

Automated counting of FJ-C+ cells was performed with 163

custom-written software in MATLAB 7.1 (Mathworks, 164

Natick, Massachusetts, USA). After automated threshold 165

adjustment and noise reduction, 25–400-lm2 166

fluorescent objects wereacceptedas cells and counted 167

in binary images. 168

Immunohistochemistry. Glial reaction was detected 169

with an indirect immunohistochemical method. 20-lm- 170

thick free-floating sections were washed in PB, and then 171

incubated in 10% normal donkey serum (NDS). For the 172

detection of activated microglia (mouse anti-CD11b, 173

clone OX42, 1:1000, Millipore) and reactive astrocytes 174

(rabbit anti-S100, 1:2000, DAKO), sections were 175

exposed to the primary antibodies overnight at 4°C, and 176

to the appropriate secondary antibodies for 2 h at room 177

(3)

178 temperature. Primary and secondary antibodies were

179 diluted in 0.1 M PB containing 0.4% Triton-X100, 2%

180 NDS and 0.01 % sodium azide. The sections were

181 coverslipped with an aqueous mounting medium.

182 Drug administration

183 The rats were divided into two groups: L-KYNs-treated

184 animals(n= 11)received 300 mg/kg L-KYNs (dissolved

185 in 5% NaOH, pH 7.4) intraperitoneally, immediately after

186 reperfusion, while the control animals (n= 12) were

187 treated with the vehicle.

188 All chemicals were purchased from VWR Ltd.,

189 Hungary, and Sigma, St. Louis,MO, USA.

190 Statistical analysis

191 Electrophysiology. EEG power spectra filtered at

192 2–20Hz were decomposed at 1-Hz intervals. The EEG

193 power of a given frequency was considered as an

194 individual case. Analysis was performed with General

195 LinearModel/Repeated measures (IBM SPSS Statistics

196 version 20).

197 Histology. Numbers of FJ-C+ cells were compared

198 with the General Linear Model. The effects of the

199 different rats were used as random effects and the

200 different treatments were used as fixed effects in the

201 mixed effect linear model (IBM SPSS Statistics version

202 20).

203 RESULTS

204 Electrophysiology

205 The EEG registered for 60 s filtered for2–20Hz revealed

206 a marked and characteristic change in EEG during

207 dMCAO (Fig. 1). The power values in each frequency

208 bin were submitted to separate repeated-measures

209 analysis of variance, with period and frequency as

210 within-subject factors. All effects with two or more

211 degrees of freedom were adjusted for violations of

212 sphericity according to the Greenhouse–Geisser

213 correction.

The ischemic period significantly reduced the power of 214

the signal as compared with the power of the EEG 215

registered before ischemia (main effect of period: 216

F(1,21) = 32.989, p< 0.0001, g2= 0.61; Fig. 2, panel 217

A, B). It was earlier observed that somatosensory- 218

evoked responses disappearcompletely during dMCAO 219

(L. Knapp, manuscript under review). Together, these 220

data indicate, that the dMCAO in our model resulted in 221

a clean-cut decay of activity in the somatosensory 222

cortices, i.e. the animals underwent a 30-min I/H period. 223

Histology 224

After a 5-day survival period, definite FJ-C staining and 225

astrocyte/microglial activation throughout the 226

somatosensory cortices emerged in approximately half 227

of the animals, ipsilateral to the dMCAO (6/12 of the 228

saline-treated animals; 5/11 of the L-KYNs-treated 229

animals). In the remaining animals, no FJ-C staining 230

and no glial reaction were observed, i.e. complete 231

staining negativity. Ipsilateral to the dMCAO, astrocyte 232

activation was characterized by hypertrophic astrocytes 233

with prominent, thick processes and small vacuoles in 234

the cell bodies as compared with the contralateral cortex 235

(Fig. 3, panel A and insert). The microglia also revealed 236

the activated phenotype ipsilateral to the dMCAO. 237

Enlarged somata and the loss of secondary and tertiary 238

branching were characteristic (Fig 3, panel B and 239

insert). The glial reaction was more prominent in the 240

L-KYNs-treated group (visual observation). The FJ-C 241

staining distribution was similar to that in the activated 242

microglia (compare Fig. 3, panels B, C). The groups 243

were compared quantitatively for FJ-C staining. The 244

number of FJ-C+ neurons was significantly higher in 245

the L-KYNs-treated group (Fig. 4, General Linear Model; 246

p= 0.023). 247

DISCUSSION 248

Physiological glutamatergic transmission through 249

NMDARs is essential in the brain, playing a key role in 250

development and synaptic plasticity. Due to its high 251

permeability for Ca2+, the NMDAR is linked to several 252

cell-signaling pathways, and to learning and memory 253

(Nakazawa et al., 2004; Zhang et al., 2007). In certain 254

acute and chronic neuropsychiatric disorders, however, 255

Ca2+ entry is the key mediator of glutamate 256

excitotoxicity and the NMDAR is the primary source of a 257

toxic Ca2+ influx (Stanika et al., 2012). NMDAR 258

antagonism is therefore an obvious neuroprotective 259

approach. 260

The failure of numerous antagonists in clinical trials is 261

due in part to the different roles of synaptic and 262

extrasynaptic NMDARs during excitotoxic processes. 263

The hypothesis that extrasynaptic NMDARs mediate cell 264

death, while synaptic NMDARs may promote survival 265

was recently discussed (Hardingham and Bading, 2010; 266

Li and Ju, 2012). From this respect, the selective 267

targeting of extrasynaptic receptors without interfering 268

with the normal synaptic function will involve a great 269

advance (Chen and Lipton, 2006). 270 Fig. 1.EEG recordings from a rat somatosensory cortex ipsilateral to

the dMCAO. 60-s EEG recordings during control (black) and ischemic (red) periods are superimposed. The EEG filtered for 2–20 Hz revealed a marked change during dMCAO. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

(4)

271 It has been argued that systemically administered

272 L-KYNs is neuroprotective in different I/H states (Gigler

273 et al., 2007; Sas et al., 2008). In such experiments, the

I/H model triggered massive excitotoxicity and a high- 274

level, long-lasting glutamate spillover. On the other 275

hand, pre-ischemic treatment was effective, since the 276 Fig. 2.(Panel A) EEG power decomposed at 1-Hz intervals. Lines demonstrate the EEG power of given frequencies during the control (line) and ischemic (dashed line) period (mean ± S.E.M.). (Panel B) The EEG power decreased significantly during the ischemic period (Repeated measures:

F(1,21) = 32.989,p< 0.0001,g2= 0.61; mean ± S.E.M.).

Fig. 3.Representative photomicrograph of the rat somatosensory cortex ipsilateral to the dMCAO after L-KYNs treatment. Double immunostaining of reactive astrocytes and microglia from the same slice; FJ-C staining from the adjacent slice (100magnification, scale bars = 500lm).

Astrocyte activation was characterized by hypertrophic astrocytes with prominent, thick processes and small vacuoles in the cell bodies (panel A, and insert). The microglia also revealed an activated phenotype, enlarged somata and the loss of secondary and tertiary branching (panel B, and insert). A high number of FJ-C+ neurons were seen throughout the cortex (panel C). The FJ-C staining pattern closely followed the microglia distribution (compare panels B and C).

Fig. 4.FJ-C+ cells counted in the rat somatosensory cortex ipsilateral to the dMCAO. The numbers of FJ-C+ cells were compared with the General Linear Model, and plotted in a bar chart. The cell number was significantly higher in the L-KYNs-treated group (General Linear Model;

p= 0.023; mean ± S.E.M.).

(5)

277 KYN/KYNA transition in the astrocytes is time-consuming

278 (Swartz et al., 1990).

279 However, in a recent study we showed that a KYNA

280 derivative significantly diminished hippocampal

281 neurodegeneration, even if administered at the time of

282 reperfusion (Gellert et al., 2011).

283 A relatively brief MCAO evokes clean-cut

284 neurodegeneration in only a fraction of the animals

285 (Memezawa et al., 1992; Aspey et al., 2000; Popp

286 et al., 2009). Similarly, in our experiment only half of the

287 animals exhibited neurodegeneration, irrespectively of

288 whether they received L-KYNs or saline treatment.

289 However, the amplitude of the evoked responses (L.

290 Knapp, manuscript under review) and the EEG power

291 decreased markedly during dMCAO, and it may be

292 therefore postulated, that the somatosensory cortices

293 were subjected to an I/H state. This indicates that

294 endogenous protective processes are able to withstand

295 a short I/H state in this cortical area.

296 Systemic treatment with L-KYNs in our experiment did

297 not alter the probability of occurrence of

298 neurodegeneration, but extended the damaged area,

299 the glial activation and the number of FJ-C+ cells in the

300 animals, which ignited cell-death pathways.

301 Around one-quarter of the extrasynaptic NMDARs in

302 adult hippocampal slices are perisynaptic (within 100 nm

303 of the postsynaptic density). Of the dendritically

304 localized extrasynaptic NMDARs, around one-third is

305 adjacent to glia-like processes (Petralia et al., 2010).

306 KYNA produced in the glia may therefore, antagonize

307 both synaptic and extrasynaptic NMDARs, influencing

308 pro-death or survival mechanisms, respectively.

309 The emergence of KYNA produced de novo from

310 systemically administered L-KYNs takes time that is

311 considerable from the aspect of an excitotoxic process

312 (Swartz et al., 1990). However, KYNA or KYNA analogs

313 can act quickly after administration. Furthermore, during

314 a brief I/H state the presence of excessive glutamate

315 and concomitant extrasynaptic NMDAR activation can

316 last for minutes (Benveniste et al., 1984; Ikonomidou

317 and Turski, 2002). The phenomenon that the KYNA

318 analog, but not L-KYNs, is neuroprotective when

319 administered after reperfusion may depend on the

320 intensity and duration of the I/H state, the concomitant

321 glutamate spillover, and the duration of the KYN-KYNA

322 turnover.

323 Another possible explanation would be that L-KYNs

324 administration led to the increased concentrations of

325 quinolinic acid (QUIN) and 3-hydroxykynurenine (3-HK),

326 neurotoxic components of the kynureninepathway.

327 Several studies observed that increased brain KYNA

328 levels follow systemic administration of L-KYNs. Swartz

329 et al. found that striatal KYNA level increased gradually

330 as a result of L-KYNs administered systemically in

331 gradually increased doses. The main conclusion of this

332 study was that extracellular levels of KYNA can be

333 dramatically increased by pharmacologic manipulation

334 of precursor levels (Swartz et al., 1990).

335 In another study concerning the effect of systemically

336 administered L-KYNs on cortical spreading depression,

337 intraperitoneal injections of L-KYNs were found to

increase cortical KYNA level about 40-fold in rats 338

(Chauvel et al., 2012). 339

Investigating the effect of systemically administered L- 340

KYNs on sensory gating, Shepard and associates found 341

that systemic administration of L-KYNs was not followed 342

by an increase of the harmful L-KYN metabolite, QUIN 343

(Shepard et al., 2003). 344

Astrocytes do not contain kynurenine 3-hydroxylase 345

and therefore cannot produce 3-HK, but are able to 346

produce large amounts of KYN and KYNA, whereas 347

microglial cells preferentially produce intermediates of 348

the quinolinic branch of the KYN pathway. It has also 349

been demonstrated that the other main source of QUIN 350

is the macrophage, infiltrated during inflammatory 351

processes (Guillemin et al., 2001; Wonodi and 352

Schwarcz, 2010). 353

In the main, the activation of the microglia increases 354

extracellular levels of QUIN or other kynurenines that 355

exacerbate neuronal damage (Schwarcz and Pellicciari, 356

2002). 357

In gerbils subjected to a period of cerebral ischemia, 358

50-fold QUIN level increases were observed 7days 359

after the onset of ischemia (Heyes and Nowak, 1990). 360

Finally, increased L-kynurenine influx from the blood 361

exceeds the catabolic capacity of kynurenine 3- 362

hydroxylase in microglia, promoting KYNA production in 363

the astrocytes (Wonodi and Schwarcz, 2010). 364

Microglia activation and the infiltration of the 365

macrophages follow the ischemic insult with a certain 366

delay. So we might reasonably conclude that the 367

extension of the damaged area in our experiments is 368

not the result of high 3-HK or QUIN levels originated 369

from L-KYNs administered promptly after reperfusion. 370

Extension of the neural damage is attributable to the 371

disturbedNMDAR-mediatedsurvival mechanisms. 372

These data indicate that kynurenergic manipulation 373

remains a potent strategy against excitotoxic cell death, 374

but the excitotoxic state and treatment pattern should be 375

well-tuned. 376

CONCLUSION 377

Suppression of excessive NMDA function has long been 378

the focus of research aimed at neuroprotection after 379

brain ischemia. However, robust NMDA antagonism is 380

not acceptable from the clinical point of view, since 381

normal synaptic NMDA function should not be inhibited, 382

even in the ischemic brain. The endogenous KYNA 383

acting at the glycine/D-serine co-agonist site of the 384

NMDA receptors is a pharmacon that might potentially 385

absolve this contradiction. Indeed, a huge body of 386

evidence indicates that manipulation of the brain KYNA 387

levels can effectively enhance the histopathological 388

outcome of the experimental I/H state. However, the 389

neuroprotective potential of L-KYNs administered after 390

brief focal ischemia has not yet been tested; 391

surprisingly, treatment with L-KYNs worsened the 392

histopathological outcome in our experiments. This 393

contradictory result indicates that post-ischemic 394

treatment with L-KYNs may be harmful. 395

(6)

396 Acknowledgement—This work was supported by OTKA grant 397 K105077.The publication is supported by the European Union 398 and co-funded by the European SocialFund.Project title:

399 ‘‘Broadening the knowledge base and supporting the long term 400 professional sustainability of the Research University Centre of 401 Excellence at the University of Szeged by ensuring the rising 402 generation of excellentscientists.’’

403 Project number: TA´MOP4.2.2-A-11/KONV-2012-0052.

404 Research project TA´MOP 4.2.4.A/2-11-1-2012-0001 is assisted 405 by the National Excellence Programa convergence program 406 to support domestic students and researchers, funded by the 407 EU and co-financed by the European SocialFund.

408 T.F. was a Bolyai Fellow of the Hungarian Academy ofSciences.

409 This research was realized in the frames of TA´MOP 4.2.4. A/1- 410 11-1-2012-0001 ‘‘National Excellence Program – Elaborating 411 and operating an inland student and researcher personal support 412 system’’.The project was subsidized by the European Union and 413 co-financed by the European Social Fund.’’

414 REFERENCES

415 Aspey BS, Taylor FL, Terruli M, Harrison MJ (2000) Temporary 416 middle cerebral artery occlusion in the rat: consistent protocol for 417 a model of stroke and reperfusion. Neuropathol Appl Neurobiol 418 26:232–242.

419 Banerjee J, Alkondon M, Pereira EF, Albuquerque EX Regulation of 420 GABAergic inputs to CA1 pyramidal neurons by nicotinic 421 receptors and kynurenic acid. J Pharmacol Exp Ther 341:500–

422 509.

423 Benveniste H, Drejer J, Schousboe A, Diemer NH (1984) Elevation of 424 the extracellular concentrations of glutamate and aspartate in rat 425 hippocampus during transient cerebral ischemia monitored by 426 intracerebral microdialysis. J Neurochem 43:1369–1374.

427 Chauvel V, Vamos E, Pardutz A, Vecsei L, Schoenen J, Multon S 428 (2012) Effect of systemic kynurenine on cortical spreading 429 depression and its modulation by sex hormones in rat. Exp 430 Neurol 236:207–214.

431 Chen HS, Lipton SA (2006) The chemical biology of clinically 432 tolerated NMDA receptor antagonists. J Neurochem 433 97:1611–1626.

434 Delorme A, Makeig S (2004) EEGLAB: an open source toolbox for 435 analysis of single-trial EEG dynamics including independent 436 component analysis. J Neurosci Methods 134:9–21.

437 Endres M, Dirnagl U (2002) Ischemia and stroke. Adv Exp Med Biol 438 513:455–473.

439 Fuchs SA, Peeters-Scholte CM, de Barse MM, Roeleveld MW, Klomp 440 LW, Berger R, de Koning TJ (2012) Increased concentrations of 441 both NMDA receptor co-agonists D-serine and glycine in global 442 ischemia: a potential novel treatment target for perinatal asphyxia.

443 Amino Acids 43:355–363.

444 Gellert L, Fuzik J, Goblos A, Sarkozi K, Marosi M, Kis Z, Farkas T, 445 Szatmari I, Fulop F, Vecsei L, Toldi J (2011) Neuroprotection with 446 a new kynurenic acid analog in the four-vessel occlusion model of 447 ischemia. Eur J Pharmacol 667:182–187.

448 Gigler G, Szenasi G, Simo A, Levay G, Harsing Jr LG, Sas K, Vecsei 449 L, Toldi J (2007) Neuroprotective effect of L-kynurenine sulfate 450 administered before focal cerebral ischemia in mice and global 451 cerebral ischemia in gerbils. Eur J Pharmacol 564:116–122.

452 Guillemin GJ, Kerr SJ, Smythe GA, Smith DG, Kapoor V, Armati PJ, 453 Croitoru J, Brew BJ (2001) Kynurenine pathway metabolism in 454 human astrocytes: a paradox for neuronal protection. J 455 Neurochem 78:842–853.

456 Gunduz-Bruce H (2009) The acute effects of NMDA antagonism:

457 from the rodent to the human brain. Brain Res Rev 60:279–286.

458 Hardingham GE (2009) Coupling of the NMDA receptor to 459 neuroprotective and neurodestructive events. Biochem Soc 460 Trans 37:1147–1160.

Hardingham GE, Bading H (2010) Synaptic versus extrasynaptic 461 NMDA receptor signalling: implications for neurodegenerative 462 disorders. Nat Rev Neurosci 11:682–696. 463

Hetman M, Kharebava G (2006) Survival signaling pathways 464 activated by NMDA receptors. Curr Top Med Chem 6:787–799. 465 Heyes MP, Nowak Jr TS (1990) Delayed increases in regional brain 466

quinolinic acid follow transient ischemia in the gerbil. J. Cerebral 467 Blood Flow Metab. 10:660–667. 468

Ikonomidou C, Stefovska V, Turski L (2000) Neuronal death 469 enhanced by N-methyl-D-aspartate antagonists. Proc Natl Acad 470 Sci U S A 97:12885–12890. 471

Ikonomidou C, Turski L (2002) Why did NMDA receptor antagonists 472 fail clinical trials for stroke and traumatic brain injury? Lancet 473 Neurol 1:383–386. 474

Kussius CL, Popescu GK (2009) Kinetic basis of partial agonism at 475 NMDA receptors. Nat Neurosci 12:1114–1120. 476

Li ST, Ju JG (2012) Functional roles of synaptic and extrasynaptic 477 NMDA receptors in physiological and pathological neuronal 478 activities. Curr Drug Targets 13:207–221. 479

Li Y, Krupa B, Kang JS, Bolshakov VY, Liu G (2009) Glycine site of 480 NMDA receptor serves as a spatiotemporal detector of synaptic 481 activity patterns. J Neurophysiol 102:578–589. 482

Lugo-Huitron R, Blanco-Ayala T, Ugalde-Muniz P, Carrillo-Mora P, 483 Pedraza-Chaverri J, Silva-Adaya D, Maldonado PD, Torres I, 484 Pinzon E, Ortiz-Islas E, Lopez T, Garcia E, Pineda B, Torres- 485 Ramos M, Santamaria A, La Cruz VP On the antioxidant 486 properties of kynurenic acid: free radical scavenging activity and 487 inhibition of oxidative stress. Neurotoxicol Teratol 33:538–547. 488 Memezawa H, Smith ML, Siesjo BK (1992) Penumbral tissues 489

salvaged by reperfusion following middle cerebral artery occlusion 490 in rats. Stroke 23:552–559. 491

Moroni F, Cozzi A, Sili M, Mannaioni G (2012) Kynurenic acid: a 492 metabolite with multiple actions and multiple targets in brain and 493 periphery. J Neural Transm 119:133–139. 494

Moskowitz MA, Lo EH, Iadecola C (2010) The science of stroke: 495 mechanisms in search of treatments. Neuron 67:181–198. 496 Muir KW (2006) Glutamate-based therapeutic approaches: clinical 497

trials with NMDA antagonists. Curr Opin Pharmacol 6:53–60. 498 Nakazawa K, McHugh TJ, Wilson MA, Tonegawa S (2004) NMDA 499

receptors, place cells and hippocampal spatial memory. Nat Rev 500 Neurosci 5:361–372. 501

Papadia S, Soriano FX, Leveille F, Martel MA, Dakin KA, Hansen HH, 502 Kaindl A, Sifringer M, Fowler J, Stefovska V, McKenzie G, 503 Craigon M, Corriveau R, Ghazal P, Horsburgh K, Yankner BA, 504 Wyllie DJ, Ikonomidou C, Hardingham GE (2008) Synaptic NMDA 505 receptor activity boosts intrinsic antioxidant defenses. Nat 506 Neurosci 11:476–487. 507

Papouin T, Ladepeche L, Ruel J, Sacchi S, Labasque M, Hanini M, 508 Groc L, Pollegioni L, Mothet JP, Oliet SH (2012) Synaptic and 509 extrasynaptic NMDA receptors are gated by different endogenous 510 coagonists. Cell 150:633–646. 511

Paxinos G, Watson CR, Emson PC (1980) AChE-stained horizontal 512 sections of the rat brain in stereotaxic coordinates. J Neurosci 513 Methods 3:129–149. 514

Petralia RS, Wang YX, Hua F, Yi Z, Zhou A, Ge L, Stephenson FA, 515 Wenthold RJ (2010) Organization of NMDA receptors at 516 extrasynaptic locations. Neuroscience 167:68–87. 517

Pohl D, Bittigau P, Ishimaru MJ, Stadthaus D, Hubner C, Olney JW, 518 Turski L, Ikonomidou C (1999) N-Methyl-D-aspartate antagonists 519 and apoptotic cell death triggered by head trauma in developing 520 rat brain. Proc Natl Acad Sci U S A 96:2508–2513. 521

Popp A, Jaenisch N, Witte OW, Frahm C (2009) Identification of 522 ischemic regions in a rat model of stroke. PLoS ONE 4:e4764. 523 Sas K, Csete K, Vecsei L, Papp JG (2003) Effect of systemic 524

administration of L-kynurenine on corticocerebral blood flow 525 under normal and ischemic conditions of the brain in conscious 526 rabbits. J Cardiovasc Pharmacol 42:403–409. 527

Sas K, Robotka H, Rozsa E, Agoston M, Szenasi G, Gigler G, Marosi 528 M, Kis Z, Farkas T, Vecsei L, Toldi J (2008) Kynurenine diminishes 529 the ischemia-induced histological and electrophysiological deficits 530 in the rat hippocampus. Neurobiol Dis 32:302–308. 531

Q3

(7)

532 Schmued LC, Stowers CC, Scallet AC, Xu L (2005) Fluoro-Jade C 533 results in ultra high resolution and contrast labeling of 534 degenerating neurons. Brain Res 1035:24–31.

535 Schwarcz R, Pellicciari R (2002) Manipulation of brain kynurenines:

536 glial targets, neuronal effects, and clinical opportunities. J 537 Pharmacol Exp Ther 303:1–10.

538 Shepard PD, Joy B, Clerkin L, Schwarcz R (2003) Micromolar brain 539 levels of kynurenic acid are associated with a disruption of 540 auditory sensory gating in the rat. Neuropsychopharmacology 541 28:1454–1462.

542 Stanika RI, Villanueva I, Kazanina G, Andrews SB, Pivovarova NB 543 (2012) Comparative impact of voltage-gated calcium channels 544 and NMDA receptors on mitochondria-mediated neuronal injury. J 545 Neurosci 32:6642–6650.

546 Stone TW (2000) Development and therapeutic potential of kynurenic 547 acid and kynurenine derivatives for neuroprotection. Trends 548 Pharmacol Sci 21:149–154.

549 Stone TW, Addae JI (2002) The pharmacological manipulation of 550 glutamate receptors and neuroprotection. Eur J Pharmacol 551 447:285–296.

552 Swartz KJ, During MJ, Freese A, Beal MF (1990) Cerebral synthesis 553 and release of kynurenic acid: an endogenous antagonist of 554 excitatory amino acid receptors. J Neurosci 10:2965–2973.

Vamos E, Pardutz A, Klivenyi P, Toldi J, Vecsei L (2009) The role of 555 kynurenines in disorders of the central nervous system: 556 possibilities for neuroprotection. J Neurol Sci 283:21–27. 557 Vecsei L, Szalardy L, Fulop F, Toldi J (2012) Kynurenines in the CNS: 558

recent advances and new questions. Nat Rev Drug Discov 559 12:64–82. 560

Wonodi I, Schwarcz R (2010) Cortical kynurenine pathway 561 metabolism: a novel target for cognitive enhancement in 562 Schizophrenia. Schizophr Bull 36:211–218. 563

Wu HQ, Guidetti P, Goodman JH, Varasi M, Ceresoli-Borroni G, 564 Speciale C, Scharfman HE, Schwarcz R (2000) Kynurenergic 565 manipulations influence excitatory synaptic function and 566 excitotoxic vulnerability in the rat hippocampus in vivo. 567 Neuroscience 97:243–251. 568

Zadori D, Klivenyi P, Vamos E, Fulop F, Toldi J, Vecsei L (2009) 569 Kynurenines in chronic neurodegenerative disorders: future 570 therapeutic strategies. J Neural Transm 116:1403–1409. 571 Zhang SJ, Steijaert MN, Lau D, Schutz G, Delucinge-Vivier C, 572

Descombes P, Bading H (2007) Decoding NMDA receptor 573 signaling: identification of genomic programs specifying 574 neuronal survival and death. Neuron 53:549–562. 575

576

577 (Accepted 30 April 2013)

578 (Available online xxxx)

Hivatkozások

KAPCSOLÓDÓ DOKUMENTUMOK

Angiography was performed from radial artery access, revealed left external iliac artery (EIA) occlusion with severe superficial femoral artery stenosis and good distal

A, Brachial artery angiography of a patient with acute hand ischemia shows a total occlusion of the axillary artery (white arrow).. B, Control angiography after 24 hr local

Pivotal role of mast cell carboxypeptidase A in mediating protection against small intestinal ischemia-reperfusion injury in rats after ischemic preconditioning. Jansson L, Carlsson

Ischemic preconditioning (IPC) was performed by three repetitive 5-min cycles of ischemia (blue color of distal coronary artery) /reperfusion (red color of

ACoA: anterior communicating artery, LACA A1: left anterior cerebral artery A1 segment, CoW: Circle of Willis, LACA A2: left anterior cerebral artery A2 segment, LMCA: left

The main goal of the present work was to reveal how the presence of L-alanine (L-Ala), L- phenylalanine (L-Phe) and L-tryptophan (L-Trp) amino acids modified the micellization

Infection with the lower dose (1 × 10 7 ) similarly showed the attenuated virulence of the cell wall mutant strain with the significantly decreased colony counts of the och1 1 / 1

In summary, our data demonstrate that systemic administration of high-dose L-KYNs induces cerebral hypoperfusion transients while it does not alter systemic blood pressure...