BRES : 44296

pp:  1210ðcol:fig: : NILÞ

Model7 brain research ] (]]]]) ]]]–]]]

121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 Q1 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180

Available online at www.sciencedirect.com

www.elsevier.com/locate/brainres

Research Report

Paclitaxel improves outcome from traumatic brain injury Donna J. Crossa,n, Gregory G. Garwina, Marcella M. Clinea, Todd L. Richardsa, Vasily Yarnykha, Pierre D. Mouradc, Rodney J.Y. Hob, Satoshi Minoshimad a

Departments of Radiology, University of Washington, 1959 N.E. Pacific Street, Seattle, WA, 98195-7115, USA Departments of Pharmaceutics, University of Washington, 1959 N.E. Pacific Street, Seattle, WA 98195-7115, USA c Departments of Neurological Surgery, University of Washington, 1959 N.E. Pacific Street, Seattle, WA 98195-7115, USA. d Department of Radiology, University of Utah, 30 North 1900 East, Salt Lake City, UT 84132, USA b

art i cle i nfo

ab st rac t

Article history:

Pharmacologic interventions for traumatic brain injury (TBI) hold promise to improve

Accepted 6 June 2015

outcome. The purpose of this study was to determine if the microtubule stabilizing therapeutic paclitaxel used for more than 20 years in chemotherapy would improve outcome after TBI. We assessed neurological outcome in mice that received direct

Keywords:

application of paclitaxel to brain injury from controlled cortical impact (CCI). Magnetic

Traumatic brain injury

resonance imaging was used to assess injury-related morphological changes. Catwalk Gait

MR imaging Microtubule stabilization Neurological function

Analysis showed significant improvement in the paclitaxel group on a variety of parameters compared to the saline group. MRI analysis revealed that paclitaxel treatment resulted in significantly reduced edema volume at site-of-injury (11.9273.0 and 8.8672.2 mm3 for saline vs. paclitaxel respectively, as determined by T2-weighted analysis; p r0.05), and significantly increased myelin tissue preservation (9.4570.4 vs. 8.9570.3, p r0.05). Our findings indicate that paclitaxel treatment resulted in improvement of neurological outcome and MR imaging biomarkers of injury. These results could have a significant impact on therapeutic developments to treat traumatic brain injury. & 2015 Published by Elsevier B.V.

1.

Introduction

Conservative estimates indicate that approximately 1.7 million traumatic brain injuries (TBIs) occur per year in the United States and this injury is the leading cause of death for Abbreviations: TBI, AD,

traumatic brain injury; IED,

ages 1–45 years. In addition, widespread use of improvised explosive devices (IEDs) against US military has resulted in approximately 17% of veterans reporting persistent cognitive deficits and post-concussive symptoms years after blast-TBI (Sources: Centers for Disease Control and Prevention and The

improvised explosive device; CTE,

Alzheimer's disease; mTBI, mild traumatic brain injury; CCI,

chronic traumatic encephalopathy;

controlled cortical impact; DTI,

diffusion tensor imaging;

FA, fractional anisotropy; MPF, macromolecular proton fraction imaging; ROI, region-of-interest; CNS, n Corresponding author. Tel.: þ1 206 598 3702; fax: þ1 206 598 6406. E-mail address: [email protected] (D.J. Cross).

central nervous system

http://dx.doi.org/10.1016/j.brainres.2015.06.006 0006-8993/& 2015 Published by Elsevier B.V.

Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

181 182 183 184 185 186 187 188 189 190 191 192 193 194 195

BRES : 44296

2

196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255

brain research ] (]]]]) ]]]–]]]

Defense and Veterans Brain Injury Center). In addition to an increasing concern for the acute effects of traumatic brain injury as a significant healthcare issue, TBI is a known environmental risk factor for the development of neurodegenerative diseases such as chronic traumatic encephalopathy (CTE) and Alzheimer's disease (AD) (Fleminger et al., 2003; Gavett et al., 2011). Repetitive mild TBI (mTBI) in boxers can lead to a dementia-like syndrome that includes motor impairment and cognitive symptoms such as bradyphrenia (slowed thinking), confusion, and memory deficits (Corsellis et al., 1973; Critchley, 1957; Martland, 1928; McKee et al., 2009b; Roberts, 1969). It is becoming increasingly evident that chronic mTBI experienced by football players is associated with CTE in mid-life and is evidenced by diffuse neurofibrillary tangles and occasionally, amyloid plaques (Erlanger et al., 1999; McKee et al., 2009b; Omalu et al., 2005). In addition, both moderate and severe head injuries significantly increase the risk of developing AD (Fleminger et al., 2003; Plassman et al., 2000). Pharmacologic interventions for traumatic brain injury hold promise to improve patient outcome, however no therapeutic has proven clinically effective to date. In this preliminary study, we tested a hypothesis if stabilizing microtubules might provide therapeutic benefits. In TBI, shear and rotational forces generated by impact cause microtubule destabilization and axonal transport disruption, which may progress to axonal swelling and disconnection (Giza and Hovda, 2001; Johnson et al., 2013; Mac Donald et al., 2007; Smith et al., 2003; Tang-Schomer et al., 2010; Tang-Schomer et al., 2012). Neuronal microtubules are part of the cytoskeletal structure and provide a framework for axonal transport processes. Tau protein, which helps stabilize microtubule structure under normal conditions (Brunden et al., 2011), accumulates in the brains of patients with CTE and AD (McKee et al., 2009a; Petrie et al., 2009). These findings led to our hypothesis that preventing microtubule destabilization may improve recovery from TBI. Paclitaxel has been administered for more than 20 years as a common treatment for ovarian, breast, lung, bladder, esophageal, and other types of solid tumor cancers (Yusuf et al., 2003), and is well characterized for its role in binding of microtubules and providing Q3 structural stabilization (Amos and Lowe, 1999; Arnal and Wade, 1995). The drug inhibits cellular mitosis by stabilizing the GDP-bound tubulin in microtubules thereby preventing depolymerization, and consequently, tumor cell division (Alberts et al., 1994; Amos and Lowe, 1999; Díaz et al., 2003). Although higher doses can be neurotoxic (Gornstein and Schwarz, 2014), limited research has suggested specific neuroprotective and neurotherapeutic effects of low dose paclitaxel (Adlard et al., 2000; Hellal et al., 2011). Microtubule stabilizers have been suggested as potential therapeutics for neurodegenerative disease based on effects on the cytoskeleton and, in particular, tau protein, which is the main component of the pathology of CTE (Brunden et al., 2011; Michaelis et al., 2002). An recent investigation into the effects of dynamic stretch injury on micropatterned neuronal cell cultures revealed that paclitaxel application prior to injury greatly reduced axonal degeneration, and resulted in greater axon preservation compared to nontreated cultures (TangSchomer et al., 2010). However, the application of paclitaxel to

brain injury in vivo has not been fully explored. This investigation is to provide evidence of feasibility for this therapeutic strategy through a direct, topical application of the drug to the injury site. As a Pgp substrate, paclitaxel does not readily cross the blood–brain barrier (BBB) and we wished to assess therapeutic efficacy without out confounding results by issues related to delivery. Future investigations to overcome this limitation for clinical translation are presented in the discussion. Here, we present findings that topical application of paclitaxel after controlled cortical impact (CCI) improve functional outcome in mice and we assessed the basis for this improvement with in vivo MR imaging. Our primary outcome measure was functional improvement in gait, which was associated with improvements in MR imaging biomarkers of injury volume, injury-associated edema, and relative preservation of myelin surrounding the injury.

2.

Results

2.1. Functional improvement after application of microtubule-stabilizing drug At 7 days post-CCI surgery, C57BL/6J mice (n ¼15, male, 10 wks) were tested in for gait abnormalities using the CatWalk automated gait analysis system (Noldus Information Technology, Wageningen, The Netherlands). Paclitaxel treated mice (n¼ 6) showed significant gait improvement over saline group (n¼ 6) in several indices that have been shown to be impaired with CCI in a previous study (Neumann et al., 2009). Spatial parameters related to individual limbs were improved as follows (Fig. 1A–F). Mean intensity, which is a measure of paw pressure on the floor was improved for all paws (22%: 78.20714.5 vs. 64.1574.1, 19%: 81.58714.5 vs. 68.4275.1, 19%: 74.17712.5 vs. 62.2373.5, and 19%: 83.86713.5 vs. 70.7374.7 for right front (RF), right hind (RH), left front (LF) and left hind (LH) respectively, arbitrary units). Maximum area is the total floor area of the paw contact in cm2 and was also improved for all paws (RF, 22%: 0.3870.1 vs. 0.3170.1, RH, 52%: 0.4070.1 vs. 0.2670.1, LF, 27%: 0.3670.1 vs. 0.2870.1 and LH, 33%: 0.3970.1 vs. 0.2970.1). Print area, assessing the complete print that comprises the stance in cm2 was also improved significantly for all limbs (RF, 21%: 0.4470.1 vs. 0.3670.1, RH, 45%: 0.4770.1 vs. 0.3370.1, LF, 20%: 0.4270.1 vs. 0.3570.1 and LH, 25%: 0.4570.1 vs. 0.3670.1). The parameter of print width, expressed in cm was significantly improved for RF, RH and LF only (RF, 5%: 0.8970.1 vs. 0.8470.1, RH, 17%: 0.9370.1 vs. 0.7970.1, and LF, 5%: 0.8170.03 vs. 0.7770.04). Print length expressed in cm was improved by paclitaxel treatment for RH only (12%: 1.0270.1 vs. 0.9170.1). The final parameter that showed significant improvement with paclitaxel was swing, which is expressed in seconds and measures the time interval between two consecutive paw placements of the same paw. Improvement in this parameter is a decrease in the time interval, which was seen in the RF and LF (RF,  12%: 0.1270.01 vs. 0.1370.02). A threshold of pr0.05 was used to determine significant improvement. No significant changes

Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315

BRES : 44296 brain research ] (]]]]) ]]]–]]]

316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375

were found in the other parameters of stance, cadence, and walk speed.

2.2.

MR imaging biomarkers indicate improvement

At 4 days postinjury, mice were anesthetized with isoflurane and scanned over the entire brain using an ultra hi-res 14 T MRI (Avance III, vertical bore, Bruker BioSpin Corp, Billerica, MA). Several MR acquisitions were used to discriminate different physiological features of the brain injury response to paclitaxel treatment. The CCI injury resulted in a reproducible and definable injury on T1-weighted images that we could use to evaluate differences in injury volume in our paclitaxel treated mice compared to those treated with saline. Analysis of injury on T1-weighted images indicated a 20% reduction in volume (9.9672.3 and 7.9471.5 mm3 for saline (n¼ 4) vs. paclitaxel (n ¼6) treatment, respectively pr 0.05). Two saline subjects had artifacts that obscured the injury on T1-weighted images and were excluded from the analysis. Sham subjects (n¼ 3) did not have an injury to quantitate, so were not used for this analysis. Quantitative T2 mapping can be used to assess the size and extent of injury associated edema and hemorrhage in TBI (Kharatishvili et al., 2009). Using thresholding to delineate the voxels with edema, we found that paclitaxel treatment reduced the volume of injury-associated edema by 26% (11.9273.0 and 8.8672.2 mm3 for saline (n¼ 4) vs. paclitaxel (n¼ 6) treatment, respectively p r0.05). The two saline subjects that were excluded from the previous T1-weighted analysis did not have a detectable artifact on the T2 maps, so were therefore included in this analysis. As might be expected, the edema volume assessed on the T2 maps was larger than the volume of actual injured tissue measured on T1-weighted images (11.9273.0 edema volume vs. 9.9672.3

3

lesion volume for saline and 8.8672.2 vs. 7.9471.5 for paclitaxel treated animals (Fig. 2A–C). We also examined the effect of paclitaxel treatment on non-neuronal cells associated with the brain injury. Macromolecular proton fraction imaging (MPF) of myelin content/ degradation utilizes a recently published fast single-point method to quantitatively measure MR modality the magnetization transfer between protons bound to water and protons bound to macromolecules (Underhill et al., 2009; Underhill et al., 2011; Yarnykh, 2012). We used MPF imaging to investigate if paclitaxel treatment had a beneficial effect on the preservation of non-neuronal brain cells following traumatic brain injury. MPF bound % was significantly increased by 6.6% in paclitaxel treated subjects (9.4570.4 vs. 8.9570.3, pr 0.05), Fig. 3A and B). Diffusion tensor imaging (DTI) evaluates the directional diffusion of water molecules in the brain. In white matter tracts, water molecules diffuse more readily along the direction of axonal fibers and movement is more restricted in the radial directions. Fractional anisotropy (FA) derived from DTI is a measure of the relative diffusion along fiber tracts, where decreased values reflect a loss of tract integrity including disruption of axonal structure and/or myelin loss, and have been used to valuate injury from CCI in rodents (Mac Donald et al., 2007). In this study we hypothesized that paclitaxel treated subjects would have increased FA values in the white matter tract directly underlying the injury site (external capsule), due to improved microtubule stabilization after injury. However, contrary to our hypothesis no significant improvement was found in the underlying white matter integrity following CCI with administration of paclitaxel. CCI surgery decreased FA in the external capsule, however saline treated subjects were slightly above the statistical threshold due to a higher standard deviation (saline: 0.3370.02,

Fig. 1 – Gait improvement in paclitaxel treated subjects. (A) Individual paw pressure on the walkway, (B–E) spatial parameters related to paw print and, (F) the temporal parameter of swing were significantly improved in mice receiving paclitaxel after TBI, indicating improved sensorimotor coordination. (*pr 0.05) RF: right front, RH: right hind, LF: left front, LH left hind. Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435

BRES : 44296

4

436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495

brain research ] (]]]]) ]]]–]]]

p¼ 0.057, paclitaxel: 0.3370.01, p¼ 0.01 vs. sham: 0.3570.01, Fig. 4A and B).

3.

Discussion

In this study, we showed that direct application of paclitaxel to the site of controlled cortical impact injury in the brain resulted in neurological improvement at 7 days post injury and that injury-related parameters assessed by MR imaging supported this improved outcome. The findings from imaging biomarkers suggested that the neurological improvement could be mediated by reduced overall injury volume, reduced injury-associated edema, and preservation of myelin in tissue surrounding the injury. Improvement of functional outcome by paclitaxel administration after traumatic brain injury has not been reported previously. We assessed outcome following CCI using a Noldus Catwalk automated gait analysis system, which permits simultaneous, observer-independent analysis of both temporal and spatial aspects of interlimb coordination. This system has been used in previous studies to assess the degree of functional impairment after brain injury in rodents (Mountney et al., 2013; Neumann et al., 2009; Wang et al.,

2011). One of these previous investigations used the Catwalk system to assess injury in a mouse model with similar CCI parameters to those used in our study and found deficits related to the injury that overlapped with most of the parameters that were improved in by paclitaxel treatment in our study, including intensity, maximum area, print area, print width, and swing (Neumann et al., 2009). The unilateral injury model to the fronto-parietal cortex that was employed both by our study and this previous investigation caused deficits in spatial parameters involved with interlimb coordination and also affected sensorimotor function, as evidenced by alterations in paw print measurements. These neurological functions were significantly improved with paclitaxel administration. Most of the literature regarding the effect of paclitaxel on neuronal populations is devoted to assessing the mechanisms by which the drug causes peripheral neuropathy during chemotherapy (Gornstein and Schwarz, 2014). In fact, these potential mechanisms are still being debated and symptoms are avoided primarily by lowering acute doses and decreasing the dose frequency should symptoms appear during treatment (Carlson and Ocean, 2011). However, prior to our study there were two reports indicating a neurotherapeutic effect of lower dose paclitaxel on central nervous system (CNS) injury.

Fig. 2 – Volume of injury and Injury-associated edema reduced with paclitaxel. (A) Representative coronal T2-quantitative maps from saline vs. paclitaxel subjects that have been thresholded to delineate pixels with edema. Green (saline) and blue (paclitaxel) arrows indicate injury edema thru several sections. Total number of injury-associated edema pixels were quantified and converted to mm3. (B) Difference in injury volume from T1-weighted images indicated significant reduction with paclitaxel. (C) Area of edema was larger for both saline and paclitaxel treated subjects than the volume of injury and also reduced significantly by treatment. (pr0.05). Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555

BRES : 44296 brain research ] (]]]]) ]]]–]]]

556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615

Hellal et al. (2011), administered paclitaxel directly to the site of spinal cord injury in a rat model . The findings from that study indicated that paclitaxel reduced fibrotic scarring by altering the dynamics of microtubule-based cargo transport. This study also found that paclitaxel promoted axonal regeneration in the spinal cord injury and improved functional recovery as assessed by the grid test. Another study by Adlard et al. (2000) applied paclitaxel to the brain directly following a needle stick injury. Although this study did not assess functional or neurological outcomes, the results from immunohistochemistry indicated a significant reduction in the density of abnormal neurites and a relative preservation of MAP2 labeling of dendrites at the injury site.

5

Our study used MR imaging to evaluate the effect of paclitaxel on the morphological features related to the injury. Use of imaging as a surrogate biomarker for traumatic brain injuries is still under development and the association of imaging findings with clinical symptoms, both acute and chronic, is the goal of numerous research investigations. Imaging modalities such as T2-mapping and DTI as used in this study, as well as positron emission tomography with 18Ffluorodeoxyglucose (FDG-PET) have been proposed to evaluate TBI in clinical settings (Hunter et al., 2012). The imaging modalities selected for this study, in particular the use of macromolecular proton fraction imaging, were in part based on findings from our study of blast-induced mild TBI in

Fig. 3 – Paclitaxel preserves myelin density around injury after CCI. Macromolecular proton fraction imaging measures the magnetization transfer between protons bound to water and protons bound to macromolecules. Yarnykh et al. showed that the bound pool fraction was strongly associated with myelin density (Underhill et al., 2011). (A) an example of a raw MPF map of a coronal section from a mouse brain after CCI, which is then thresholded to exclude injury pixels (hypointense regions). Mean myelin density was calculated for a ROI (yellow circle) that was larger than the actual injury (1.5 mm). (B) shows the bound pool fraction (associated strongly with myelin density) is more preserved with paclitaxel treatment (*pr0.05). Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675

BRES : 44296

6

676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735

brain research ] (]]]]) ]]]–]]]

veterans (Peskind et al., 2011; Petrie et al., 2014). The caveat being that blast-induced repetitive TBI causes a diffusely distributed injury and our controlled cortical impact injury model produces a focally well-defined lesion-type injury. Therefore, we preferentially chose imaging (T2-mapping and T1-weighted structural imaging) to assess the morphological changes that might be detected by these modalities in response to paclitaxel treatment. The reduction of injury volume from T1-weighted images indicates that paclitaxel may have affected apoptotic processes in a manner that reduced cell death triggered by the CCI. On T2-maps, the volume of injury edema was decreased by paclitaxel application. This finding may indicate that the drug affected the neuroinflammatory response to the injury, possibly thru a modification of glial proliferation and motility. Our study in blast-exposed veterans indicated that MPF imaging specifically was a sensitive indicator of blast injury, and other studies have indicated myelin damage as a consequence of TBI (Goldstein et al., 2012; Johnson et al., 2013). Using MPF imaging, our results indicated that paclitaxel helped to preserve the myelin density on the injury margins. This finding was not predicted by our hypothesis that paclitaxel would stabilize microtubules in neurons. However, we speculate that oligodendrocytes with their highly ordered microtubule

arrays, might also benefit from stabilization as do neuronal populations (Lunn et al., 1997). The cellular substrates for the improved imaging outcomes in this study require further investigation. Other neurotherapeutic applications of microtubule stabilizing drugs have been proposed, which include treatment of AD and other tauopathies (Brunden et al., 2011; Lou et al., 2014). The rationale for this therapeutic strategy is that hyperphosphorylated tau in AD disengages from MTs and this loss of normal stabilization leads to a perturbation of neuronal functions including decreased axonal transport and overall loss of cytoskeletal integrity. One such drug, Epothilone D, which has good blood-brain barrier (BBB) penetration, was shown to improve cognitive performance and reduce associated tau pathology in transgenic mouse models (Barten et al., 2012; Brunden et al., 2011). The findings from these studies further support the use of paclitaxel and other microtubule stabilizers for neurotherapeutic applications and suggest that this therapeutic strategy may benefit the underlying association between TBI and future development of neurodegenerative disorders. One obstacle that must be overcome for the development of paclitaxel as a therapeutic option for CNS disorders is that, as a P-glycoprotein (P-gp) substrate, the drug has limited BBB

Fig. 4 – Fractional Anisotropy in the white matter underlying injury not improved by paclitaxel. (A) indicates representative coronal brain images from saline, paclitaxel and sham subjects with ROI indicated (yellow outline). (B) indicates that subjects undergoing CCI procedure had decreased FA values as compared to sham controls. (*p r0.01, however #p ¼0.057, due to high standard deviation in saline group). However, no significant difference was seen between treatment groups. Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795

BRES : 44296 brain research ] (]]]]) ]]]–]]]

796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855

permeability. Our study used direct application to the injury because our CCI injury model employs a craniotomy. However, this is not often the case for clinically presented TBI, and direct cortical drug administration may not be feasible in a clinical setting. One potential delivery option under development by our group involves findings that the blood-brainbarrier is transiently disrupted after TBI even in mild cases (Alves, 2014; Lucke-Wold et al., 2014; Si et al., 2014). Experiments to define the parameters of paclitaxel uptake to the brain after TBI in relation to BBB disruption are underway by our group. Future studies will focus on more conventional methods of drug delivery as well as an evaluation of drug pharmacokinetics to define brain uptake in relation to BBB disruption and therapeutic time window of drug delivery after injury. Contrary to our hypothesis, no improvement in the underlying cytoskeletal injury was observed on the fractional anisotropy maps from our DTI acquisitions. There may be several reasons that explain this finding. First, our DTI imaging protocol may not be sensitive enough and the signal-to-noise ratio may be too low to detect subtle improvements from the paclitaxel administration. We are working to improve the DTI acquisition protocol by modifying parameters, lengthening the acquisition time and employing stronger head stabilization measures as small movements from respiration and cardiac output can cause motion artifacts at 14 T. Another possibility for this finding is that with direct application of paclitaxel to the injury surface, the drug did not penetrate deep enough into the tissue to reach the underlying white matter tracts. It is also possible that the timing of imaging compared to hypothesized therapeutic effect may be not optimized for this biomarker. We performed DTI imaging at 4 days post-injury and its possible that the therapeutic effects of paclitaxel on the white matter tracts could take longer than 4 days to manifest. Alternatively, the acute neuroprotective/neurotherapeutic effects of paclitaxel for TBI could be mediated by processes other than the maintenance of cytoskeletal integrity. These different possibilities are under investigation by our group to assess the contributing factors to this unexpected result. Through preliminary, these results support our hypothesis that direct cortical application of paclitaxel may improve recovery from TBI. Future directions for this research will investigate more clinically relevant administration routes and will assess the effect of dose response for therapeutic efficacy. The current study applied the paclitaxel directly after CCI injury, however the therapeutic time window for efficacy of administration with respective time of injury is not known. This will also be a focus of further investigation.

indicate that paclitaxel treatment resulted in improvement of neurological outcome and MR imaging biomarkers of injury suggested that the improvement could be mediated by reduced overall injury volume, reduced injury-associated edema, and preservation of myelin in tissue surrounding the injury. These results could have a significant impact on therapeutic developments to treat traumatic brain injury.

5.

Experimental procedure

5.1.

Subjects

All procedures were conducted in accordance with the animal care guidelines issued by the National Institutes of Health and by the Institutional Animal Care and Use Committee. Adult male C57BL/6 J mice, 10 weeks of age, were purchased from the Jackson Laboratory, Bar Harbor, ME. Animals were kept on a 12-h light/12-h dark cycle with ad libitum access to water and food before and during experimental procedures, and were randomly assigned to three groups (paclitaxel, n¼ 6; saline control, n¼ 6; and sham surgery control, n¼ 3).

5.2.

Surgery and drug administration

Animals were anesthetized with isoflurane (1.5–2.5% in flowing O2) during surgery and their core temperature was maintained at 37 ˚C with a heating pad. After securing the animal in a stereotaxic frame, the scalp was shaved and the skin scrubbed with Betadine. Lidocaine, Bupivacaine (1 mg/ kg) and Buprenorphine (0.5 mg/kg) were injected subcutaneously into the scalp and an incision at the midline was made. Fascia and skin were retracted, and a 5 mm craniotomy was performed with a high-speed surgical drill (Dremel, Racine, WI) over the right frontoparietal cortex center point at 2.5 mm behind bregma and 2.5 mm off the midline. Through this opening, the animal was subjected to a controlled cortical impact using a pneumatic impactor (AmScien Instruments, Richmond, VA, USA) with a 3 mm convex tip. The impact injury was generated using the following parameters: 6 m/s strike velocity, 1 mm depth of penetration, and 150 ms contact time. Immediately following impact, 5 ml of sterile isotonic saline or 10 ml of 1.8 mg/ml of paclitaxel (Hospira, Inc, Lake Forest, IL) was applied to the cortex at the injury site at a flow rate of 2 ml/min. The scalp was then closed with sutures and the animal was given an interscapular subcutaneous injection of 1.0 ml isotonic saline to prevent dehydration. Sham controls received a craniotomy, but no impact injury.

5.3.

4.

7

Functional assessment

Conclusions

Pharmacologic interventions for traumatic brain injury hold promise to improve outcome, but no therapeutic has proven clinically effective to date. The goal of this study was to determine if the microtubule stabilizing therapeutic paclitaxel, a unique approach recently tested in neurological conditions such as AD and spinal cord injury (Brunden et al., 2011; Hellal et al., 2011) would improve outcome after TBI. Our findings

Catwalk gait analysis has been used previously to evaluate impairment following CCI in rodents (Mountney et al., 2013; Neumann et al., 2009; Wang et al., 2011). CCI injury is unilateral and with our mild impact, we required a sensitive test of motor/neurological function. The advantage of the gait analysis system is that it is automated and assessment of gait parameters is observer independent (results are checked only for non “paw” marks such as fecal droppings to be excluded

Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915

BRES : 44296

8

916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975

brain research ] (]]]]) ]]]–]]]

from analyses). Animals underwent gait assessment at 7 days after surgery using the CatWalk automated gait analysis system (Noldus Information Technology, Wageningen, The Netherlands). The testing device included a 1.3-m-long glass plate with dim fluorescent light beamed into the glass from the side. Under dim overhead lighting, the light was reflected downward and the images of the footprints were recorded by the camera under a walkway as the mouse's paws came in contact with the glass surface. Mice performed 3 consecutive runs through the apparatus. Images from each trial were converted into digital signals and processed with a threshold of 179 arbitrary units (au). Following the identification and labeling of each footprint, gait data were generated for spatial parameters related to individual paws (mean intensity, maximum contact area, print area, print width, and print length) and temporal parameters (swing, stance, cadence, and walk speed). Paclitaxel vs. saline controls were compared using a ttest and pr 0.05 threshold for significance. One sham control died prior to catwalk testing (did not recover from imaging anesthesia) and could not be used in the analysis and that left only 2 shams, which were not enough for statistical comparison to TBI groups.

5.4.

Imaging and image processing

At 4 days post injury, mice were anesthetized with isoflurane and scanned over the entire brain using an ultra hi-res 14 T MRI (Avance III, vertical bore, Bruker BioSpin Corp, Billerica, MA). Image acquisitions included T1-weighted structural imaging using a 3D Modified Driven Equilibrium Fourier Transform (MDEFT) sequence with a voxel size of 0.14  0.14  0.25 mm3, 64 slices, flip angle/repetition time/echo time (FA/TR/TE): 121/5000 ms/1.9 ms that was used to assess total injury volume and T2 quantitative mapping (T2) with a voxel size of 0.12  0.12  1.0 mm3, 15 slices, TR¼ 2000 ms,16 echoes, spacing: 6.7 ms, TE Effective 1: 6.7 ms, TE Effective 2: 13.4 ms, which was used to measure the volume of the edema directly associated with the injury site. To evaluate axonal integrity in the white matter, diffusion tensor imaging (DTI) was acquired over the entire brain using a 4-shot echo-planar imaging (EPI) sequence applied along 30 noncolinear diffusion-gradient directions with a voxel size of 0.195  0.195  0.5 mm3, 35 slices, FA/TR/TE: 901/8750 ms/ 17.8 ms. DTI processing included the application of a custom algorithm, which allows elimination of image frames contaminated by motion-derived artifacts, followed by eddy current and B0 corrections and confers several advantages. The resultant diffusivity maps exhibit little spatial distortion, artifacts of subject motion or inhomogeneities of magnetic (B0) or RF fields; and show a high degree of white/gray matter contrast. Fractional anisotropy (FA) maps were constructed from DTI by FSL software (Analysis Group, FMRIB, Oxford, UK). Macromolecular proton fraction imaging (MPF) of myelin content/degradation utilizes a recently published fast singlepoint method to quantitatively measure MR modality the magnetization transfer between protons bound to water and protons bound to macromolecules (Underhill et al., 2009; Underhill et al., 2011; Yarnykh, 2012). They showed that the bound pool fraction was strongly associated with myelin density in both white matter and gray matter. In brief, this modality uses a single magnetization transfer (MT) image with

off-resonance saturation, a reference image, and a T1 map to compute an MPF map based on the pulsed MT model with appropriate constraints for other model parameters. Source data included three spoiled gradient-echo (GRE) images (TR/TE¼ 20/ 3.2 ms, excitation flip angles [FA] α¼ 3, 10, and 201) for variable flip angle (VFA) T1 mapping, an MT GRE image (TR/TE¼ 40/ 3.2 ms, α¼ 101) with off-resonance Gaussian saturation pulse (offset Δ¼ 4 kHz, effective FA 9501, duration 19 ms), and a reference GRE image with the same parameters and without off-resonance saturation. All images were acquired in 3D mode with a 144  144  500 μm3 voxel size (FOV 23x23x16 mm3, matrix 160  160  32).

5.5.

Image analysis

After processing of MR images to generate FA and MPF quantitative maps, all images were thresholded to isolate the feature of interest or to exclude pixels with extremely high values due to image noise (examples of image thresholding can be seen in subsequent figures for each image result). Manual region-ofinterest (ROI) analysis was performed using ImageJ software. For T1-weighted images, the identifiable injury area (hypointensity compared to surrounding uninjured cortex) was manually traced on each slice and the total number of voxels was multiplied by voxel size to estimate injury volume in mm3. On the T2 quantitative maps, threshold bounds were set from 36 to 200, which included regions of edema and ventricles, but excluded normal uninjured cortex and white matter. Manually drawn ROIs on images displayed by Image J software included the volume of voxels within the threshold boundary and total area of injuryassociated edema was calculated by multiplying with voxel size in mm3. On MPF maps we wished to assess the preservation of the myelin density associated with the boundary of the injury site. For this purpose we again used image thresholding to exclude hypointense voxels representing the actual injury and hyperintense voxels that represent noise on the brain/skull boundary and drew a large circular ROI that exceeded the injury size on 5 slices that centered on the injury site. For this analysis, lower-bound thresholding was used to exclude hypointense voxels of the injury and include only the voxels that bordered the injury within an ROI of a specified diameter (1.5 mm). The mean intensity of the voxels within this thresholded region was calculated to assess preserved myelin density on the injury boundary. For DTI, the white matter integrity of the external capsule directly below the injury was assessed on FA maps using the mean intensity values over a manually drawn ROI. All ROI analyses were performed by an investigator blinded to treatment group. For all ROI values group-wise mean and standard deviation were compared using unpaired t-tests with pr0.05 statistical significance.

Conflicts of interest There are no conflicts of interest to report.

Uncited reference Westin et al., 2002.

Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 Q2 1033 1034 1035

BRES : 44296 brain research ] (]]]]) ]]]–]]]

1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095

Acknowledgments Support for this research was provided by the Institute for Translational Health Sciences pilot project grant opportunity (UL1TR000423) and the UW. Center on Human Development and Disability animal imaging and animal behavioral cores The authors would like to acknowledge Nathalie Martin and Abigail McClintic for their assistance with this project.

r e f e r e n c e s

Adlard, P.A., King, C.E., Vickers, J.C., 2000. The effects of taxol on the central nervous system response to physical injury. Acta Neuropathol. 100, 183–188. Alberts, B., et al., 1994. Molecular biology of the cell. Garland Publishing, Inc, New York. Alves, J.L., 2014. Blood-brain barrier and traumatic brain injury. J. Neurosci. Res. 92, 141–147. Amos, L., Lowe, J., 1999. How taxol (R) stabilises microtubule structure. Chem. Biol. 6, R65–R69. Barten, D.M., et al., 2012. Hyperdynamic microtubules, cognitive deficits, and pathology are improved in tau transgenic mice with low doses of the microtubule-stabilizing agent BMS241027. J. Neurosci.: off. J. Soc. Neurosci. 32, 7137–7145. Brunden, K.R., et al., 2011. The characterization of microtubulestabilizing drugs as possible therapeutic agents for Alzheimer’s disease and related tauopathies. Pharmacol. Res. 63, 341–351. Carlson, K., Ocean, A.J., 2011. Peripheral neuropathy with microtubule-targeting agents: occurrence and management approach. Clin. Breast Cancer 11, 73–81. Corsellis, J.A., Bruton, C.J., Freeman-Browne, D., 1973. The aftermath of boxing. Psychol. Med. 3, 270–303. Critchley, M., 1957. Medical aspects of boxing, particularly from a neurological standpoint. Br. Med. J. 1, 357–362. Dı´az, J.F., Barasoain, I., Andreu, J.M., 2003. Fast kinetics of Taxol binding to microtubules. Effects of solution variables and microtubule-associated proteins. J. Biol. Chem. 278, 8407–8419. Erlanger, D.M., et al., 1999. Neuropsychology of sports-related head injury: dementia Pugilistica to Post Concussion Syndrome. Clin. Neuropsychol. 13, 193–209. Fleminger, S., et al., 2003. Head injury as a risk factor for Alzheimer’s disease: the evidence 10 years on; a partial replication. J. Neurol. Neurosurg. Psychiatr. 74, 857–862. Gavett, B.E., Stern, R.A., McKee, A.C., 2011. Chronic traumatic encephalopathy: a potential late effect of sport-related concussive and subconcussive head trauma. Clin. Sports Med. 30, 179–188. Giza, C.C., Hovda, D.A., 2001. The neurometabolic cascade of concussion. J. Athl. Train. 36, 228–235. Goldstein, L.E., et al., 2012. Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Sci. Transl. Med. 4. Gornstein, E., Schwarz, T.L., 2014. The paradox of paclitaxel neurotoxicity: Mechanisms and unanswered questions. Neuropharmacology 76, 175–183Pt A. Hellal, F., et al., 2011. Microtubule stabilization reduces scarring and causes axon regeneration after spinal cord injury. Science 331, 928–931. Hunter, J.V., et al., 2012. Emerging imaging tools for use with traumatic brain injury research. J. Neurotrauma 29, 654–671. Johnson, V.E., Stewart, W., Smith, D.H., 2013. Axonal pathology in traumatic brain injury. Exp. Neurol. 246, 35–43. Kharatishvili, I., et al., 2009. Quantitative T2 mapping as a potential marker for the initial assessment of the severity of

9

damage after traumatic brain injury in rat. Exp. Neurol. 217, 154–164. Lou, K., et al., 2014. brain-penetrant, orally bioavailable microtubule-stabilizing small molecules are potential candidate therapeutics for Alzheimer’s disease and related tauopathies. J. Med. Chem. 57, 6116–6127. Lucke-Wold, B.P., et al., 2014. Bryostatin-1 restores blood brain Q4 barrier integrity following blast-induced traumatic brain injury. Mol Neurobiol. Lunn, K.F., Baas, P.W., Duncan, I.D., 1997. Microtubule organization and stability in the oligodendrocyte. J. Neurosci. 17, 4921-4932. Mac Donald, C.L., et al., 2007. Detection of traumatic axonal injury with diffusion tensor imaging in a mouse model of traumatic brain injury. Exp. Neurol. 205, 116–131. Martland, H., 1928. Punch drunk. J. Am. Med. Assoc. 91, 1103–1107. McKee, A.C., et al., 2009a. Chronic traumatic encephalopathy in athletes: progressive tauopathy after repetitive head injury. J. Neuropathol. Exp. Neurol. 68, 709–735. McKee, A.C., et al., 2009b. Chronic traumatic encephalopathy in athletes: progressive tauopathy after repetitive head injury. J. Neuropathol. Exp. Neurol. 68, 709–735. Michaelis, M.L., Dobrowsky, R.T., Li, G., 2002. Tau neurofibrillary pathology and microtubule stability. J. Mol. Neurosci. 19, 289–293. Mountney, A., et al., 2013. Longitudinal assessment of gait abnormalities following penetrating ballistic-like brain injury in rats. J. Neurosci. Methods. 212, 1–16. Neumann, M., et al., 2009. Assessing gait impairment following experimental traumatic brain injury in mice. J. Neurosci. Methods. 176, 34–44. Omalu, B.I., et al., 2005. Chronic traumatic encephalopathy in a National Football League player. Neurosurgery. 57, 128-134; discussion 128-34. Peskind, E.R., et al., 2011. Cerebrocerebellar hypometabolism associated with repetitive blast exposure mild traumatic brain injury in 12 Iraq war Veterans with persistent post-concussive symptoms. Neuroimage 54 (Suppl 1), S76–S82. Petrie, E.C., et al., 2009. Preclinical evidence of Alzheimer changes: convergent cerebrospinal fluid biomarker and fluorodeoxyglucose positron emission tomography findings. Arch. Neurol. 66, 632–637. Petrie, E.C., et al., 2014. Neuroimaging, behavioral, and psychological sequelae of repetitive combined blast/impact mild traumatic brain injury in Iraq and Afghanistan war veterans. J. Neurotrauma. 31, 425–436. Plassman, B.L., et al., 2000. Documented head injury in early adulthood and risk of Alzheimer’s disease and other dementias. Neurology 55, 1158–1166. Roberts, G., 1969. Brain damage in boxers. Pitman Medical and Scientific Publishing Co. Ltd., London. Si, D., et al., 2014. Progesterone protects blood-brain barrier function and improves neurological outcome following traumatic brain injury in rats. Exp Ther Med. 8, 1010–1014. Smith, D.H., Meaney, D.F., Shull, W.H., 2003. Diffuse axonal injury in head trauma. J. Head Trauma Rehabil. 18, 307–316. Tang-Schomer, M.D., et al., 2010. Mechanical breaking of microtubules in axons during dynamic stretch injury underlies delayed elasticity, microtubule disassembly, and axon degeneration. FASEB J. 24, 1401–1410. Tang-Schomer, M.D., et al., 2012. Partial interruption of axonal transport due to microtubule breakage accounts for the formation of periodic varicosities after traumatic axonal injury. Exp. Neurol. 233, 364–372. Underhill, H.R., Yuan, C., Yarnykh, V.L., 2009. Direct quantitative comparison between cross-relaxation imaging and diffusion

Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155

BRES : 44296

10

1156 1157 1158 1159 1160 1161 1162 1163 1164

brain research ] (]]]]) ]]]–]]]

tensor imaging of the human brain at 3.0 T. NeuroImage. 47, 1568–1578. Underhill, H.R., et al., 2011. Fast bound pool fraction imaging of the in vivo rat brain: association with myelin content and validation in the C6 glioma model. NeuroImage 54, 2052–2065. Wang, Y., et al., 2011. Fluoxetine increases hippocampal neurogenesis and induces epigenetic factors but does not improve functional recovery after traumatic brain injury. J. Neurotrauma 28, 259–268.

Westin, C.-F., et al., 2002. Processing and visualization for diffusion tensor MRI. Med. Image Anal. 6, 93–108. Yarnykh, V.L., 2012. Fast macromolecular proton fraction mapping from a single off-resonance magnetization transfer measurement. Magn. Reson. Med. 68, 166–178. Yusuf, R.Z., et al., 2003. Paclitaxel resistance: molecular mechanisms and pharmacologic manipulation. Curr. Cancer Drug Targets 3, 1–19.

Please cite this article as: Cross, D.J., et al., Paclitaxel improves outcome from traumatic brain injury. Brain Research (2015), http://dx.doi.org/10.1016/j.brainres.2015.06.006

1165 1166 1167 1168 1169 1170 1171 1172

Paclitaxel improves outcome from traumatic brain injury.

Pharmacologic interventions for traumatic brain injury (TBI) hold promise to improve outcome. The purpose of this study was to determine if the microt...
2MB Sizes 7 Downloads 16 Views