NSC 15371

No. of Pages 8, Model 5G

5 May 2014 Please cite this article in press as: Zweckberger K et al. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience (2014), http://dx.doi.org/10.1016/j.neuroscience.2014.04.040 1

Neuroscience xxx (2014) xxx–xxx

2 3

GLIBENCLAMIDE REDUCES SECONDARY BRAIN DAMAGE AFTER EXPERIMENTAL TRAUMATIC BRAIN INJURY

4 5 6

Q1 K. ZWECKBERGER, * K. HACKENBERG C. S. JUNG,

7

Department of Neurosurgery, University Heidelberg, Germany

8

Abstract—Following traumatic brain injury (TBI) SUR1-regulated NCCa-ATP (SUR1/TRPM4) channels are transcriptionally up-regulated in ischemic astrocytes, neurons, and capillaries. ATP depletion results in depolarization and opening of the channel leading to cytotoxic edema. Glibenclamide is an inhibitor of SUR-1 and, thus, might prevent cytotoxic edema and secondary brain damage following TBI. Anesthetized adult Sprague–Dawley rats underwent parietal craniotomy and were subjected to controlled cortical impact injury (CCI). Glibenclamide was administered as a bolus injection 15 min after CCI injury and continuously via osmotic pumps throughout 7 days. In an acute trial (180 min) mean arterial blood pressure, heart rate, intracranial pressure, encephalographic activity, and cerebral metabolism were monitored. Brain water content was assessed gravimetrically 24 h after CCI injury and contusion volumes were measured by MRI scanning technique at 8 h, 24 h, 72 h, and 7 d post injury. Throughout the entire time of observation neurological function was quantified using the ‘‘beam-walking’’ test. Glibenclamide-treated animals showed a significant reduction in the development of brain tissue water content(80.47% ± 0.37% (glibenclamide) vs. 80.83% ± 0.44% (control); p < 0.05; n = 14). Contusion sizes increased continuously within 72 h following CCI injury, but glibenclamide-treated animals had significantly smaller volumes at any time-points, like 172.53 ± 38.74 mm3 (glibenclamide) vs. 299.20 ± 64.02 mm3 (control) (p < 0.01; n = 10; 24 h) or 211.10 ± 41.03 mm3 (glibenclamide) vs. 309.76 ± 19.45 mm3 (control) (p < 0.05; n = 10; 72 h), respectively. An effect on acute parameters, however, could not be detected, most likely because of the up-regulation of the channel within 3–6 h after injury. Furthermore, there was no significant effect on motor function assessed by the beam-walking test throughout 7 days. In accordance to these results and the available literature, glibenclamide seems to have promising potency in the treatment of TBI. Ó 2014 Published by Elsevier Ltd. on behalf of IBRO.

Key words: traumatic brain injury, glibenclamide, secondary brain damage, brain edema, cerebral metabolism, epileptic seizures.

D. N. HERTLE, K. L. KIENING, A. W. UNTERBERG AND O. W. SAKOWITZ

9

INTRODUCTION Despite significant improvements in intensive care Q4 management, severe traumatic brain injury (TBI) still remains the leading cause of death and long-term disability and dependency in young patients. Brain edema, leading to uncontrollable increased intracranial pressure (ICP), is predominantly responsible for the development of secondary brain damage characterized by enlarged contusion volumes, and hence, deteriorated neurological outcome. Treatment guidelines, thus currently focus on the control and the reduction of brain swelling (Brain Trauma Foundation, 2007). Osmotherapy has been the mainstay of pharmacological therapy and is typically administered as an escalating treatment scheme (Brain Trauma Foundation, 2007; Walcott et al., 2012). Among others, a novel treatment target for cerebral edema might be the SUR1-regulated NCCa-ATP (SUR1/TRPM4) channel (Simard et al., 2006; Walcott et al., 2012). Recently, this non-specification channel has been described in ischemic astrocytes, neurons, and capillaries regulated by sulfonylurea receptor 1 (SUR1). Depletion of ATP, as it can frequently be observed in the peri-contusional area following trauma, causes depolarization and opening of this channel. This finally leads to an uncontrolled influx of cations and, according to the osmotic pressure, of water resulting in cell ‘‘blebbing’’ that is characteristic of cytotoxic edema (Simard et al., 2006). In situations of mechanical stress, inflammation, and hypoxia an up-regulation of the ABcc8 gene that encodes for SUR1 receptors could be observed (Simard et al., 2012a). SUR-1 receptors can specifically be blocked by the administration of glibenclamide, explaining the fact, that patients with diabetes type II receiving glibenclamide seem to be in favor to reach a better neurological outcome after stroke quantified by the Modified Ranking Scale and the National Institute of Health Stroke Scale in comparison to controls (Kunte et al., 2007). Furthermore, in a rodent model of cerebral stroke, blocking of SUR-1 receptors with low-dose glibenclamide has significantly reduced cerebral edema, infarct volume, and mortality rate (Simard et al., 2006, 2009a; Zhou et al., 2009). In experimental models of subarachnoid hemorrhage glibenclamide seems to have positive effects on inflamma-

*Corresponding author. Address: Department of Neurosurgery, University of Heidelberg, ImNeuenheimer Feld 400, 69120 Q2 Heidelberg, Germany. Tel.: +49 62215636314. E-mail address: [email protected] (K. Zweckberger). Q3 Abbreviations: CCI, controlled cortical impact injury; EEG, electroencephalography; HF, heart rate; ICP, intracranial pressure; MANOVA, multivariate analysis of variance; MAP, mean arterial blood pressure; MD, microdialysis; TBI, traumatic brain injury. http://dx.doi.org/10.1016/j.neuroscience.2014.04.040 0306-4522/Ó 2014 Published by Elsevier Ltd. on behalf of IBRO. 1

10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53

NSC 15371

No. of Pages 8, Model 5G

5 May 2014

2

K. Zweckberger et al. / Neuroscience xxx (2014) xxx–xxx

67

tion and edema formation (Simard et al., 2009b). In experimental spinal cord injury, glibenclamide diminishes the development of hemorrhagic necrosis, leading to improved neurological outcome (Simard et al., 2007a, 2012b,c; Popovich et al., 2012). The inhibition of the upregulated SUR1 receptor by low-dose glibenclamide displays furthermore a key role in the prevention of developing a progressive secondary hemorrhage following TBI (Simard et al., 2009c). In this current study the effect of glibenclamide on the development of secondary brain damage after experimental TBI is investigated emphasizing the influence on brain edema, contusion volume, cortical activity, and cerebral metabolism.

68

EXPERIMENTAL PROCEDURES

54 55 56 57 58 59 60 61 62 63 64 65 66

69

Animals

70

Q5 A total of 68 Sprague–Dawley rats (Charles River,

71

Germany, 350–380 g) were subjected to this study. Guidelines for laboratory animal care were strictly followed. Animals had free access to food and water throughout the study and all experiments were performed under deep anesthesia with isoflurane.

72 73 74 75

76

Experimental groups

77

93

A total of three experimental sections were investigated: In sections 1 (n = 20), acute (patho-) physiological changes, encompassing mean arterial blood pressure (MAP), heart rate (HF), body temperature, ICP, encephalographic activity, and cerebral metabolism (microdialysis (MD)) were monitored continuously over 180 min following trauma. In section 2 (n = 28), brain water content was assessed gravimetrically 24 h after controlled cortical impact injury (CCI), and, in section 3 (n = 20), contusion volumes by MRI scanning and neurological assessment were quantified 8 h, 24 h, 72 h, and 7 d following trauma, respectively. In all sections animals were randomized either to a treatment-, receiving glibenclamide, or to a control group that received placebo. In section 1, furthermore a shamoperated group was added. In this group animals were not traumatized, but only received a craniotomy.

94

Anesthesia and trauma application

95

All experiments were performed under deep anesthesia using 1.8–2.2% isoflurane, 68% N2O and 30% O2. During the entire time of anesthesia a catheter in the tail artery digitally monitored systemic MAP and HF. Furthermore, blood gases were controlled in regular intervals. In order to control and maintain body temperature at 37.0 ± 0.5 °C, rats were positioned on a feedback-controlled heating pad. As previously described, experimental TBI was performed using a ‘‘Controlled Cortical Impact’’ injury (CCI) device (Zweckberger et al., 2003, 2006). Briefly, after induction of anesthesia, the head was fixed in a stereotactic frame and, using a micro-drill under permanent cooling with saline, a temporo-parietal craniotomy was performed.

78 79 80 81 82 83 84 85 86 87 88 89 90 91 92

96 97 98 99 100 101 102 103 104 105 106 107 108

Thereby special attention was paid to leave the dura intact. The trauma was performed perpendicular to the surface of the brain (90 degrees) by an impactor that was adjusted at an angle of 35 degrees. The diameter of the flat impactor (with rounded edges) was 5 mm, the velocity 7.5 m/s, the impact depth 1.5 mm, and the impact duration 300 ms. In order to avoid any decompressive effects created by the craniotomy itself, the bone-flap was replaced immediately after CCI and fixed with dental cement.

109

Drug administration

119

15 min after CCI, a loading dose of glibenclamide (10 lg/kg) was administered subcutaneously, and additionally, osmotic pumps (AlzetÒ, Model 2ML1; ALZET Osmotic Pumps, DURECT Corporation, USA) Q6 were implanted subcutaneously in the left flank region for continuous delivery of the drug throughout 7 days (10 ll/h). All rats, randomized either to the treatment or Q7 the control group, have received the same total volume. According to Simard et al. [13], a stock solution of 25 mg of glibenclamide (Sigma, St. Louis, MO, USA) in 10 ml dimethylsulfoxide (DMSO) was prepared. The infusion/injection solution was made by mixing 200 ll stock in 25 ml un-buffered normal saline (0.9%NaCl), thus, administering a total concentration of 200 ng/h.

120

ICP and electro-encephalography (EEG)

135

ICP was measured using an intraparenchymal micro-ICP probe (CodmanÒ MicroSensor Basic Kit; Raynham, MA 02767) that was inserted in the cortex via a borehole 2 mm rostral from the anterior edge of the craniotomy. Monitoring started 10 min prior to CCI receiving a physiological baseline, and was continued throughout the entire time of observation following CCI injury (180 min). In order to monitor encephalographic activity (EEG) two stainless screws were inserted in two boreholes fronto-lateral and parieto-occipital referred to the craniotomy. Bipolar recording was amplified using a Bio Potential Amplifier Module type 675 (Hugo Sachs Electronic-Harvard Apparatus GmbH, March-Hugstetten, Germany). Filter limits were set to 0.003–100 Hz. To avoid electrical interference at 50 Hz a notch filter was applied. These records were screened for epileptiform activity by one of the investigators who was blinded to the entire treatment procedure. Quantitative measurements of the EEG signals, such as root mean square values, delta (1–3 Hz), theta- (4–7 Hz), alpha(8–15 Hz), and beta (15–30 Hz) power, as well as the mean frequency, were derived from the artifact-free recordings 10 min before and up to 180 min after CCI (Sakowitz et al., 2002).

136

MD

161

Following CCI, the MD catheter (CMA12: length 14 mm; length of the permeable membrane 2 mm; diameter

162

Please cite this article in press as: Zweckberger K et al. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience (2014), http://dx.doi.org/10.1016/j.neuroscience.2014.04.040

110 111 112 113 114 115 116 117 118

121 122 123 124 125 126 127 128 129 130 131 132 133 134

137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160

163

NSC 15371

No. of Pages 8, Model 5G

5 May 2014

3

K. Zweckberger et al. / Neuroscience xxx (2014) xxx–xxx 164 165 166 167 168 169 170 171 172 173

0.5 mm; molecular cut-off 100,000 Dalton) was inserted 2 mm rostral from the contusion, in the peri-contusional area representing the most active metabolic region of the traumatized brain. The perfusion rate was 1.2 ll/min. Using the MD technique extracellular concentrations of glutamate, lactate, pyruvate, and glucose were assessed in the acute stage up to 180 min following CCI injury. Dialysates were collected at regular 30-min intervals and stored immediately at 70 °C for further spectral photometric analyses.

174

Brain water content

175

In order to assess brain water content, animals were sacrificed in deep isoflurane anesthesia 24 h after CCI. After careful removal of the brains, hemispheres were divided subsequently. Finally, brain water content was assessed gravimetrically, as already described (Zweckberger et al., 2003, 2006).

176 177 178 179 180

181

Contusion volume

182

To quantify posttraumatic contusion volumes, repeated MRI scans were performed with a 2.35-T animal Ò scanner (Biospect 24/40, BRUKER Medizintechnik, Ettlingen, Germany) 8 h, 24 h, 72 h, and 7 days after trauma, offering both the advantage to re-investigate the same animals at different time-points and therefore, to reduce the total number of needed animals. Contusion volumes were estimated using 2-mm slices of T2-weighted spin echo (TSE) sequences and an Image J software package (ImageJ v.1.36, US National Institutes of Health, Bethesda, MD).

183 184 185 186 187 188 189 190 191 192

193

Neurological outcome

194

In order to assess neurological function and impairment the ‘‘beam-walking test’’ representing motor-function and -coordination was performed. Tests started 3 days prior surgery receiving reliable baseline scores, were repeated daily, and performed throughout the 7 days of observation.

195 196 197 198 199

200

Data analysis

201

Statistical analyses were performed using a standard statistic software package (IBM SPSS Statistics 18; SPSS Inc., an IBM Company, Chicago, IL, SAD). All averaged data are given as mean ± standard deviation of the mean (SD) if not indicated otherwise. A threshold value for significance (p-value) of less than 0.05 was presumed. Student’s t-test was used for comparison of two independent normal distributed samples, repeatedmeasures analysis of variance (ANOVA) with Bonferroni comparison for analysis within a series of measurements and multivariate ANOVA (MANOVA) for measurement series between two groups. If significance was proven in MANOVA, individual group comparisons were conducted using post hoc tests.

202 203 204 205 206 207 208 209 210 211 212 213 214

RESULTS

215

Physiological parameters

216

In the present study physiological parameters were neither impaired by anesthesia nor by the administration of glibenclamide. MAP remained constant throughout the entire time of acute monitoring (104.62 ± 12.31 mmHg, 30 min after CCI; 102.00 ± 15.23 mmHg, 180 min after CCI, respectively). None of the animals showed any hypotension characterized by a MAP less than 80 mmHg. Furthermore, there was no influence on HF that ranged within physiological levels at any time (in average from 393.26 ± 37.04/min, 120 min after CCI to 403.32 ± 33.89/min, 120 min after CCI, respectively, with a maximal range from 326/min to 476/min). Body temperature was kept constant by using a feedback control heating pad and ranged in average at 37.47 ± 0.07 °C excluding hypo- or hyperthermia. Blood gas analyses that were assessed both in the acute and the chronic trial ruled out glibenclamide-induced hypoglycemia (serum) and showed physiological concentrations of serum electrolytes. Continuous administration of low-dose glibenclamide for 7 days led not to significant differences between the treatment and control groups that ranged within physiological levels (glibenclamide: 167.72 ± 30.24 mg/dl vs. control: 156.22 ± 56.28 mg/dl; p = 0.57; n = 10).

217

ICP

241

Ten minutes prior to CCI injury baseline ICP averaged about 8 mmHg (glibenclamide: 8.30 ± 1.10 mmHg vs. control: 8.08 ± 1.79 mmHg; p > 0.05; n = 10) and did not show any significant difference between the treatment and the control groups. Following CCI injury, ICP increased significantly (p < 0.05) and reached 14.80 ± 2.90 mmHg in the treatment group vs. 13.80 ± 2.15 mmHg in controls (p > 0.05; n = 10) 120 min after trauma. A significant difference between the treatment and control groups, however, could not be observed at this early stage after trauma. (Fig. 1).

242

Cerebral MD

253

In parallel to ICP measurements, concentrations of extracellular metabolic parameters were assessed in the peri-contusional region in the acute trial up to 180 min after CCI injury. Instantaneously after trauma, glucose concentrations dropped to a ‘‘close to zero’’ level (0.036 ± 0.022 mmol/l (glibenclamide) vs. 0.023 ± 0.019 mmol/l (control); p > 0.05) and recovered within 120 min (0.143 ± 0.110 mmol/l (glibenclamide) vs. 0.115 ± 0.080 mmol/l (control); p > 0.05). As a sign of anaerobic metabolism and cell death the concentrations of lactate and glutamate increased between 30 and 60 min following trauma in both groups. (glutamate 30 min. after CCI: 1.487 ± 1.387 lmol/l (glibenclamide) vs. 0.904 ± 0.326 lmol/l (control), p > 0.05, and glutamate 60 min. after CCI: 12.857 ± 8.742 lmol/l (glibenclamide) vs. 11.879 ± 10.151 lmol/l (control), p > 0.05, respectively). At later time-points of observation both, glutamate and lactate concentrations

254

Please cite this article in press as: Zweckberger K et al. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience (2014), http://dx.doi.org/10.1016/j.neuroscience.2014.04.040

218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240

243 244 245 246 247 248 249 250 251 252

255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271

NSC 15371

No. of Pages 8, Model 5G

5 May 2014

4

K. Zweckberger et al. / Neuroscience xxx (2014) xxx–xxx

Fig. 1. Continuous measurement of the intracranial pressure (ICP) from 10 min before to 180 min after controlled cortical impact injury (CCI): Following stable baseline conditions, ICP has increased significantly within 30 min after trauma and has stabilized at a moderate level without showing any significant difference between the treatment (glibenclamide) and the control groups.

274

declined continuously. Following a non-significant peak of the lactate/pyruvate ratio 60 min after CCI, its level was kept constant in both groups (Fig. 2).

275

EEG

272 273

276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304

Both, in the control and treatment groups average spectral frequency was reduced after CCI injury and showed a significant difference in comparison to shamoperated animals throughout all time-points. In Q8 comparison to baseline levels, in the glibenclamide group a-, c-, and d-power kept significantly increasing 90 min after CCI injury, but there was no significant difference between the treatment and control groups. Glibenclamide-treated animals, however, showed, in comparison to controls, a trend for increased EEGc-, and d-power 90 min after CCI injury (c-power: 508.77 ± 274.34 lV2 (Glibenclamide) vs. 346.36 ± 338.20 lV2 (Control); d-power: 107.53 ± 60.65 lV2 (Glibenclamide) vs. 81.11 ± 80.53 lV2 (Control)). Neither during baseline monitoring, nor in sham-operated animals any epileptic seizures could be detected. In summary, in nine traumatized animals seizures occurred (four in the control-, and five in the treatment group). Animals, treated with glibenclamide showed on the average 2.8 ± 6.2 seizures with a total duration of 35.7 ± 94.7 s and an average duration of 4.3 ± 5.7s. In control animals, both, number (4.1 ± 7.2) and duration were increased (total duration: 64.1 ± 103.4 s; average duration: 6.8 ± 8.9 s), but have not reached significance. In comparison between EEG and MD data, furthermore, there was a positive correlation between e.c. glutamate concentrations and the number (r = 0.54; p < 0.01) and duration of epileptic seizures (r = 0.46; p < 0.01) at 90 and 120 min after trauma.

Brain water content

305

24 h following CCI injury brain water content of the traumatized and non-traumatized hemispheres were separately assessed gravimetrically. Brain water content Q9 of the non-injured hemisphere was 78.52 ± 0.19% in the Glibenclamide group, and 78.48 ± 0.29% in the control group and thus, represented physiological levels. In the injured hemisphere, however, a significant increase of brain water content could be observed. In comparison to the control group, rats having been randomized to the treatment group showed a significant reduction in brain water content (80.47 ± 0.37% vs. 80.83 ± 0,44%; p < 0.05; n = 14). Treatment with Glibenclamide, thus, could reduce the post-traumatic increase of brain water content by 15.3% (Fig. 3).

306

Contusion volume

320

Treatment with glibenclamide had a significant beneficial effect on the development of contusion volumes. Using MRI scanning technique, a reliable and non-invasive method quantifying contusion sizes, significant effects of glibenclamide on contusion volume could be detected 8 h, 24 h, 72 h, and 7 days following trauma(Fig. 4). Within the first 72 h a continuous increase of contusion size from158.00 ± 36.82 mm3 (glibenclamide) vs. 264.11 ± 44.37 mm3 (control) (n = 10; p < 0.05) 8 h, to 172.53 ± 38.74 mm3 (glibenclamide) vs. 299.20 ± 64.02 mm3 (control) (p < 0.01; n = 10) 24 h and, finally, to 211.10 ± 41.03 mm3 (glibenclamide) vs. 309.76 ± 64.38 mm3 (control) (p < 0.01; n = 10) 72 h after CCI injury could be measured. Contusion volumes were reduced 7 days after trauma, but still, a significant difference between the treatment and the control groups

321

Please cite this article in press as: Zweckberger K et al. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience (2014), http://dx.doi.org/10.1016/j.neuroscience.2014.04.040

307 308 309 310 311 312 313 314 315 316 317 318 319

322 323 324 325 326 327 328 329 330 331 332 333 334 335 336

NSC 15371

No. of Pages 8, Model 5G

5 May 2014

K. Zweckberger et al. / Neuroscience xxx (2014) xxx–xxx

5

Q12 Fig. 2. Concentration of extracellular metabolic parameters, such as glutamate, lactate, pyruvate and the lactate/pyruvate ratio between 30 and 180 min after CCI injury: Not showing any differences between the treatment and control groups, the post-traumatic increase of e.c. lactate and glutamate concentrations represent an anaerobic metabolism and cell death. (Very low concentrations, e.g. lactate and pyruvate at 30 min are not detectable). 337 338

was evident: 107.13 ± 21.33 mm3 (glibenclamide) vs. 163.96 ± 28.99 mm3 (control) (p < 0.01; n = 10).

339

Neurological function

340

Motor function and coordination were assessed by performing the ‘‘beam-walking’’ test. Prior CCI injury animals showed 0.15 ± 0.36 misplacements referring to the corresponding contralateral hind paw. After trauma the number of misplacements increased significantly and reached 10.89 ± 2.83 (glibenclamide) and 10.24 ± 4.20 (control) (p < 0.01; n = 10) followed by continuous recovery in both groups. A significant difference between the treatment and control groups, however, could not be observed within 7 days of observation.

341 342 343 344 345 346 347 348 349

350

DISCUSSION

351

In this study it was found that treatment with glibenclamide can reduce brain edema development and contusion volume following experimental TBI.

352 353

Despite several attempts to influence inflammation, excitotoxicity, and vascularization, clinical treatment of post-traumatic brain edema is still a challenge, and osmotherapy has been the mainstay for years. Both, mannitol and hyper-oncotic saline are well-established (Berger et al., 1995; Brain Trauma Foundation, 2007; Finke, 2012; Marks et al., 2012), although, there is no treatment available that already prevents the development of brain edema. Among others, the currently described SUR1regulated NCCa-ATP (SUR1/TRPM4) channel might offer a new treatment target (Simard et al., 2006; Walcott et al., 2012). This channel is not constitutively expressed, but is transcriptionally up-regulated de novo in all cells of the neurovascular unit in many forms of central nervous system injury, including cerebral ischemia, TBI, spinal cord injury, and subarachnoid hemorrhage (Simard et al., 2012a). In comparison and unlike the well-known KATP-channel, SUR-1, but not Kir6.1 or Kir6.2 is transcriptionally up-regulated in ischemic astrocytes, neurons, and capillaries representing the regulatory subunit of this

Please cite this article in press as: Zweckberger K et al. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience (2014), http://dx.doi.org/10.1016/j.neuroscience.2014.04.040

354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374

NSC 15371

No. of Pages 8, Model 5G

5 May 2014

6

K. Zweckberger et al. / Neuroscience xxx (2014) xxx–xxx

Fig. 3. Gravimetrical assessment of brain water content 24 h following controlled cortical impact injury (CCI): Animals treated with glibenclamide showed significant reduced brain edema (white). In reference to physiological levels of the contralateral hemisphere, the post-traumatic increase of brain edema could be reduced by 15.3%.

Fig. 4. Post-traumatic contusion volume quantified by MRI scanning technique at 8 h, 24 h, 72 h, and 7 d post injury. Animals treated with glibenclamide showed significant reduced contusion sizes at any time-points (white).

375 376 377 378 379 380 381 382 383

channel (Simard et al., 2006). On the other hand, Trpm4 is considered to be the pore-forming unit (Simard et al., 2007b; Woo et al., 2013). Ischemia-induced increase in SUR-1 was corroborated by measurements of Abcc8 mRNA, encoding SUR-1, which increased two- to threefold in the core center of the infarct in a MCA stroke model 3 h after the insult (Deeley et al., 2006; Simard et al., 2006). In the peri-infarct region the ascent of SUR-1 was delayed and a peak could sharply be demarcated

8–16 h post stroke (Simard et al., 2006, 2012a). The NCCa-ATP channel is highly dependent on ATP. Depletion of ATP, as it can be observed frequently in the peri-contusional area following trauma or in the so-called penumbra region after stroke causes depolarization and, thus, opening of the pore-unit. As a consequence cations, accompanied by water following the osmotic gradient, are floating into cells resulting in ‘‘cell blebbing’’ that is characteristic of cytotoxic edema (Simard et al., 2006).

Please cite this article in press as: Zweckberger K et al. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience (2014), http://dx.doi.org/10.1016/j.neuroscience.2014.04.040

384 385 386 387 388 389 390 391 392

NSC 15371

No. of Pages 8, Model 5G

5 May 2014

K. Zweckberger et al. / Neuroscience xxx (2014) xxx–xxx

Administration of low-dose glibenclamidecan inhibits the regulatory sub-unit SUR-1 of the NCCa-ATP channel. 395Q10 As a rare fluke patients with stroke that were on 396 glibenclamide treatment having diabetes mellitus seem 397 to be in favor to reach a better neurological 398 outcome(Kunte et al., 2007). The group of Simard et al. 399 could further convincingly show in a rodent model of cere400 bral MCA stroke that administration of glibenclamide can 401 significantly reduce cerebral edema, infarct volume, and 402 mortality rate (Simard et al., 2006, 2009a; Zhou et al., 403 2009). 404 Following experimental TBI using the Feeney device, 405 Simard et al. (2009c) could also demonstrate that SUR406 1 is up-regulated, especially in capillaries, within 3 h in 407 the tissue beneath the impact. Furthermore a prominent 408 peak could be shown in deeper structures, like the hippo409 campus, 24 h after trauma. By treating rats with glibencla410 mide fragmentation of capillaries and, thus, hemorrhagic 411 necrosis measured by Drabkin’s reagent and contusion 412 size (Nissl sections) could be reduced resulting in a better 413 neurological outcome quantified by ‘‘spontaneous vertical 414 exploration’’ (Simard et al., 2009c). 415 Despite these promising results, experimental data 416 addressing post-traumatic pathologies, like ICP, 417 metabolic changes, and post-traumatic brain edema 418 have been lacking, so far. In the current study using a 419 CCI model treatment with low-dose glibenclamide has 420 resulted in a highly significant reduction of brain edema 421 assessed gravimetrically 24 h post CCI injury. In MRI 422 scans a continuous increase of contusion sizes could be 423 depicted at 8, 24, and 72 h following CCI injury. In 424 comparison to controls, animals that were treated with 425 glibenclamide furthermore showed significant smaller 426 contusion sizes at any time. Despite those results, ICP 427 and extracellular metabolism were not significantly 428 influenced by glibenclamide in the acute stage within 429 180 min following trauma. This could be due to the fact 430 that SUR-1 is transcriptionally up-regulated de novo 431 within 3–6 h following trauma, as mentioned above. 432 Thus, treatment in the very acute stage seems to be 433 less effective, although, in accordance with the results 434 of Simard et al. (2006, 2012a), we could show that contu435 sion sizes were already reduced 8 h after injury. Espe436 cially with reference to the MD data, the intensity of the 437 trauma can be regarded as moderate. After an acute drop 438 of the e.c. glucose concentration (‘‘close to zero level’’) 439 that is typical following trauma and representing an acute 440 energy failure (Hutchinson et al., 1991, 2000; Persson 441 et al., 1996; Goodman et al., 1999; Kett-White et al., 442 2002), e.c. glutamate and lactate concentrations have 443 significantly increased within 60 min, but continuously 444 recovered throughout the further time of observation. 445 The incline of glutamate and lactate in the peri446 contusional region implies a shift toward anaerobic 447 metabolism leading to cell death that only occurred in 448 the very acute stage (60 min), but did not continue or 449 aggravate throughout the remaining time of observation 450 as it would be expected in severe TBI (Obrenovitch, 451 1999; Alessandri et al., 2003; Hillered et al., 2005; 452 Chamoun et al., 2010). This could potentially explain the 453 lack of difference between the treatment and the control 393 394

7

454 groups in the ‘‘beam-walking’’ test assessing misplace455 ments of the contralateral hind paw. Post CCI injury rats 456 of the treatment and the control groups showed signifi457 cantly more misplacements, but recovered very fast 458 within a few days and did not show long-lasting impair459 ments. More specifically, glibenclamide seems to improve 460 ‘‘behavioral abnormalities’’ quantified by ‘‘spontaneous 461 vertical exploration’’ (Simard et al., 2009c) and to reduce 462 hippocampal injury resulting in improved spatial learning 463 (Patel et al., 2010). 464 Impairment of neurotransmitter metabolism leads to 465 an imbalance in neurotransmitter receptor expression 466 (Cremer et al., 2009). Changes in the membrane distribu467 tion of N-methyl-D-aspartate (NMDA) glutamate recep468 tors, e.g., seem to produce sweeping modifications in 469 neuronal excitability (Va`zquez-Lo`pez et al., 2005). In vitro 470 experiments could show that induced astrocyte glutamate 471 release triggers transient depolarization and epileptic 472 form discharges (Kang et al., 2005). Buckingham et al. (2011) could further demonstrate that the release of gluta- Q11473 474 mate after the transplantation of glioma cells into mice 475 have led to epileptiform hyper-excitability that spreads 476 over the adjacent brain tissue, explaining the observation 477 that seizures are a common comorbidity in primary brain 478 tumors. Therefore, the correlation between e.c. glutamate 479 concentrations and the number and duration of seizures, 480 as mentioned above, are well explained and might be an 481 interesting point for pursuing investigations. 482 In summary and in accordance with the available 483 literature, we could demonstrate that treatment with low484 dose glibenclamide reduces post-traumatic brain edema 485 and contusion volume following CCI injury in rats. 486 Possibly owing to the delayed transcriptional up487 regulation of the regulatory sub-unit of the NCCa-ATP 488 channel, no effect on ICP and cerebral metabolism in 489 the very acute stage following CCI injury could be 490 detected. Although glibenclamide seems to have high 491 therapeutic potency in the treatment of traumatic brain 492 and spinal cord injury, as well as stroke, further 493 experimental and clinical studies are necessary to 494 further delineate the role of glibenclamide in the 495 treatment of TBI.

REFERENCES

496

Alessandri B, Heimann A, Filippi R, Kopacz L, Kempski O (2003) Moderate controlled cortical contusion in pigs: effects on multiparametric neuromonitoring and clinical relevance. J Neurotrauma 20(12):1293–1305. Berger S, Schu¨rer L, Ha¨rtl R, Messmer K, Baethmann A (1995) Reduction of post- traumatic intracranial hypertension by hypertonic /hyperoncotic saline/dextran and hypertonic mannitol. Neurosurgery 37(1):98–107. Brain Trauma Foundation, (BTF), AANS Joint Section on Neurotrauma and Critical Care (2007) Guidelines for the management of severe head injury: 3rd Edition. J Neurotrauma (Suppl)24(1):1–106. Buckingham SC, Campbell SL, Haas BR, Montana V, Robel S, Ogunrinu T, Sontheimer H (2011) Glutamate release by primary brain tumors induces epileptic activity. Nat Med 17(10):1269–1274. Chamoun R, Suki D, Gopinath SP, Goodman JC, Robertson C (2010) Role of extracellular glutamate measured by cerebral

497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514

Please cite this article in press as: Zweckberger K et al. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience (2014), http://dx.doi.org/10.1016/j.neuroscience.2014.04.040

NSC 15371

No. of Pages 8, Model 5G

5 May 2014

8 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 635 636 637

K. Zweckberger et al. / Neuroscience xxx (2014) xxx–xxx

microdialysis in severe traumatic brain injury. J Neurosurg 113(3):564–570. Cremer CM, Palomero-Gallagher N, Bidmon HJ, Schleicher A, Speckmann EJ, Zilles K (2009) Pentylenetetrazole-induced seizures affect binding site densities for GABA, glutamate and adenosine receptors in the rat brain. Neuroscience 163(1): 490–499. Deeley RG, Westlake C, Cole S (2006) Transmembrane transport of endo- and xenobiotics by mammalian ATP-binding cassette multidrug resistance proteins. Physiol Rev 86:849–899. Finke ME (2012), Osmotherapy for intracranial hypertension: mannitol versus hypertonic saline. Continuum (MinneapMinn). 2012, 18(3), pp. 640–654. Goodman JC, Valadka AB, Gopinath SP, Uzura M, Robertson CS (1999) Extracellular lactate and glucose alterations in the brain after head injury measured by microdialysis. Crit Care Med 27(9):1965–1973. Hillered L, Vespa PM, Hovda DA (2005) Translational neurochemical research in acute human brain injury: the current status and potential future for cerebral microdialysis. J Neurotrauma 22(1):3–41. Hutchinson PJ, Al-Rawi PG, O’Connell MT, Gupta AK, Maskell LB, Hutchinson DB, Pickard JD, Kirkpatrick PJ (1991) Monitoring of brain metabolism during aneurysm surgery using microdialysis and brain multiparameter sensors. Neurol Res 21(4):352–358. Hutchinson PJ, Al-Rawi PG, O’Connell MT, Gupta AK, Pickard JD, Kirkpatrick PJ (2000) Biochemical changes related to hypoxia during cerebral aneurysm surgery: combined microdialysis and tissue oxygen monitoring: case report. Neurosurgery 46(1):201–206. Kang N, Xu J, Xu Q, Nedergaard M, Kang J (2005) Astrocytic glutamate release-induced transient depolarization and epileptiform discharges in hippocampal CA1 pyramidal neurons. J Neurophysiol 94(6):4121–4230. Kett-White R, Hutchinson PJ, Al-Rawi PG, Gupta AK, O’Connell MT, Pickard JD, Kirkpatrick PJ (2002) Extracellular lactate/pyruvate and glutamate changes in patients during per-operative episodes of cerebral Ischaemia. Acta Neurochir Suppl 81:363–365. Kunte H, Schmidt S, Eliasziw M, del Zoppo GJ, Simard JM, Masuhr F, Weih M, Dirnagl U (2007) Sulfonylureas improves outcome in patients with type 2 diabetes and acute ischemic stroke. Stroke 38(9):2526–2530. Marks JA, Li S, Gong W, Sanati P, Eisenstadt R, Sims C, Smith DH, Reilly PM, Pascual JL (2012) Similar effects of hypertonic saline and mannitol on the inflammation of the blood-brain barrier microcirculation after brain injury in a mouse model. J Trauma Acute Care Surg 73(2):351–357. Obrenovitch TP (1999) High extracellular glutamate and neuronal death in neurological disorders. Cause, contribution or consequence? Ann N Y Acad Sci 890:273–286. Patel AD, Gerzanich V, Geng Z, Simard JM (2010) Glibenclamide reduces hippocampal injury and preserves rapid spatial learning in a model of traumatic brain injury. J Neuropathol Exp Neurol 69(12):1177–1190. Persson L, Valtysson J, Enblad P, Warme PE, Cesarini K, Lewen A, Hillered L (1996) Neurochemical monitoring using intracerebral microdialysis in patients with subarachnoid hemorrhage. J Neurosurg 84(4):606–616. Popovich PG, Lemeshow S, Gensel JC, Tovar CA (2012) Independent evaluation of the effects of glibenclamide on

reducing progressive hemorrhagic necrosis after cervical spinal cord injury. Exp Neurol 233(2):615–622. Sakowitz OW, Unterberg AW, Stover JF (2002) Neuronal activity determined by quantitative EEG and cortical microdialysis is increased following controlled cortical impact injury in rats. Acta Neurochir Suppl 81:221–223. Simard JM, Chen M, Tarasov KV, Bhatta S, Ivanova S, Melnitchenko L, Tsymbalyuk N, West GA, Gerzanich V (2006) Newly expressed SUR-1 regulated NC Ca-ATP channel mediates cerebral edema after ischemic stroke. Nat Med 12(4):433–440. Simard JM, Tsymalyuk O, Ivanov A, Ivanova S, Bhatta S, Geng Z, Woo SK, Gerzanich V (2007a) Endothelial sulfonylurea receptor 1 regulates NCCa–ATP channels mediated progressive hemorrhagic necrosis following spinal cord injury. J Clin Invest 117(8):2105–2113. Simard JM, Tarasov KV, Gerzanich V (2007b) Non-selective cation channels, transient receptor potential channels and ischemic stroke. Biochim Biophys Acta 1772:947–957. Simard JM, Yurovsky V, Tsymbalyuk N, Melnichenko L, Ivanova S, Gerzanich V (2009a) Protective effects of delayed treatment with low-dose glibenclamide in three models of ischemic stroke. Stroke 40(2):604–609. Simard JM, Geng Z, Woo SK, Ivanova S, Tosun C, Melnichenko L, Gerzanich V (2009b) Glibenclamide reduces inflammation, vasogeneticedema, and caspase-3 activation after subarachnoid hemorrhage. J Cereb Blood Flow Metab 29(2):317–330. Simard JM, Kibourne M, Tsymalyuk O, Tosun C, Caridi J, Ivanova S, Keledjian K, Bochicchio G, Gerzanich V (2009c) Key role of sulfonylurea receptor 1 in progressive secondary hemorrhage after brain contusion. J Neurotrauma 26(12):2257–2267. Simard JM, Woo SW, Schwartzbauer GT, Gerzanich V (2012a) Sulfonylurea receptor 1 in central nervous system injury: a focused review. J Cereb Blood Flow Metab 32(9):1699–1717. Simard JM, Tsymalyuk O, Keledjian K, Ivanov A, Ivanova S, Gerzanich V (2012b) Comparative effects of glibenclamide and riluzole in a rat model of severe cervical spinal cord injury. Exp Neurol 233(1):566–574. Simard JM, Popovich PG, Tsymalyuk O, Gerzanich V (2012c) Spinal cord injury with unilateral versus bilateral primary hemorrhage – effects of glibenclamide. Exp Neurol 233(2):829–835. Va`zquez-Lo`pez A, Sierra-Paredes G, Sierra-Maruno G (2005) Seizures induced by microperfusion of glutamate and glycine in the hippocampus of rats pretreated with latrunculin A. Neurosci Lett 388(2):81–85. Walcott BP, Kahle KT, Simard JM (2012) Novel treatment targets for cerebral edema. Neurotherapeutics 9(1):65–72. Woo SK, Kwon MS, Ivanov A, Gerzanich V, Simard JM (2013) The sulfonylurea receptor 1 (Sur1)-transient receptor potential melastatin 4 (Trpm4) channel. J Biol Chem 288(5):3655–3667. Zhou Y, Fathali N, Lekic T, Tang J, Zhang JH (2009) Glibenclamide improves neurological function in neonatal hypoxia-ischemia in rats. Brain Res 1270:131–139. ZweckbergerK Stoffel M, Baethmann A, Plesnila N (2003) Effect of decompression craniotomy on increase of contusion volume and functional outcome after controlled cortical impact in mice. J Neurotrauma 20(12):1307–1314. Zweckberger K, Ero¨s C, Zimmermann R, Kim SW, Engel D, Plesnila N (2006) Effect of early and delayed decompressive craniectomy on secondary brain damage after controlled cortical impact in mice. J Neurotrauma 23(7):1083–1093.

(Accepted 17 April 2014) (Available online xxxx)

Please cite this article in press as: Zweckberger K et al. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience (2014), http://dx.doi.org/10.1016/j.neuroscience.2014.04.040

575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634

Glibenclamide reduces secondary brain damage after experimental traumatic brain injury.

Following traumatic brain injury (TBI) SUR1-regulated NCCa-ATP (SUR1/TRPM4) channels are transcriptionally up-regulated in ischemic astrocytes, neuron...
959KB Sizes 0 Downloads 6 Views