Glioma-Derived Platelet-Derived Growth Factor-BB Recruits Oligodendrocyte Progenitor Cells via Platelet-Derived Growth Factor Receptor-α and Remodels Cancer Stroma

Glioma-Derived Platelet-Derived Growth Factor-BB Recruits Oligodendrocyte Progenitor Cells via Platelet-Derived Growth Factor Receptor-α and Remodels Cancer Stroma

1 2 3 4 5 6 7 8 9 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 ...

6MB Sizes 0 Downloads 21 Views

1 2 3 4 5 6 7 8 9 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 54 55 56 57 58 59 60 61 62

The American Journal of Pathology, Vol. -, No. -, - 2016

ajp.amjpathol.org

SHORT COMMUNICATION Glioma-Derived Platelet-Derived Growth Factor-BB Recruits Oligodendrocyte Progenitor Cells via Platelet-Derived Growth Factor Receptor-a and Remodels Cancer Stroma Q15

Yang Zheng,* Seiji Yamamoto,* Yoko Ishii,* Yang Sang,* Takeru Hamashima,* Nguyen Van De,* Hirofumi Nishizono,y Ran Inoue,z Hisashi Mori,z and Masakiyo Sasahara* From the Departments of Pathology* and Molecular Neuroscience,z Graduate School of Medicine and Pharmaceutical Sciences, and the Division of Animal Experimental Laboratory,y Life Science Research Center, University of Toyama, Toyama, Japan Accepted for publication December 21, 2015. Address correspondence to Yoko Ishii, M.D., Ph.D., Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0152, Japan. E-mail: [email protected].

Q3

Glioma is an aggressive and incurable disease, and is frequently accompanied by augmented platelet-derived growth factor (PDGF) signaling. Overexpression of PDGF-B ligand characterizes a specific subclass of glioblastoma multiforme, but the significance of the ligand remains to be elucidated. For this end, we implanted a glioma cell line transfected with PDGF-BBeoverexpressing vector (GL261-PDGF-BB) or control vector (GL261-vector) into wild-type mouse brain, and examined the effect of glioma-derived PDGF on the tumor microenvironment. The volume of GL261-PDGFBB rapidly increased compared with GL261-vector. Recruitment of many PDGF receptor (PDGFR)-a and Olig2-positive oligodendrocyte precursor cells and frequent hemorrhages were observed in GL261-PDGF-BB but not in GL261-vector glioma. We then implanted GL261-PDGF-BB into the mouse brain with and without Pdgfra gene inactivation, corresponding to PDGFRa-knockout (KO) and Flox mice, respectively. The recruitment of oligodendrocyte precursor cells was largely suppressed in PDGFRa-KO than in Flox mice, whereas the volume of GL261-PDGF-BB was comparable between the two genotypes. Frequent hemorrhage and increased IgG leakage were associated with aberrant vascular structures within the area where many recruited oligodendrocyte precursor cells accumulated in Flox mice. In contrast, these vascular phenotypes were largely normalized in PDGFRa-KO mice. Increased matrix metalloproteinase-9 in recruited oligodendrocyte precursor cells and decreased claudin-5 in vasculature may underlie the vascular abnormality. Glioma-derived PDGF-B signal induces cancer stroma characteristically seen in high-grade glioma, and should be therapeutically targeted to improve cancer microenvironment. (Am J Pathol 2016, -: 1e11; http:// dx.doi.org/10.1016/j.ajpath.2015.12.020)

Glioblastoma multiforme (GBM), World Health Organization grade IV astrocytoma, is the most common malignant brain tumor of adults. GBM is characterized by highly invasive features and prominent vascular involvement.1 In addition to the aggressive growth of tumor cells, the associating secondary events, like intracranial hemorrhage, are critical prognostic factors as well.2 Recently, tumor cells per se as well as microenvironmentddefined by extracellular matrix and tumor stromal cells, including vascular endothelial cells and glial cellsdgained attention as a therapeutic target.3,4 Typically, a

Q2

treatment targeting tumor angiogenesis using an antibody against vascular endothelial growth factor, bevacizumab, is used at tumor recurrence, and achieves significant improvement of the disease.5 Furthermore, the current efforts are developing novel and potent agents that target multiple angiogenic pathways, including platelet-derived growth factor (PDGF) and Supported by Ministry of Education, Culture, Sports, Science, and Technology grants-in-aid for scientific research 25293093 (M.S.) and 15K08396 (Y.I.). Q1 Disclosures: None declared.

Copyright ª 2016 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ajpath.2015.12.020

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124

125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186

Zheng et al fibroblast growth factor, for the treatment of advanced solid tumors.6 PDGF family members include PDGF-A, PDGF-B, PDGF-C, and PDGF-D, which are assembled as disulfidelinked homodimers or heterodimers. The PDGF signal is mediated by two types of PDGF receptors, PDGF receptor a (PDGFRa) and PDGFRb.7 PDGF ligands and receptors were expressed in the brain; among them, PDGFRa is most abundantly expressed in oligodendrocyte progenitor cells (OPCs), and is implicated in the regulation of the proliferation, migration, and differentiation of OPCs in developing brain.8 Furthermore, in the recent studies, PDGFRa signaling is well documented in the disruption of the blood-brain barrier in different pathological conditions.9,10 PDGF autocrine mechanisms are assumed to be most important in glioma,11 and proteomic analyses demonstrated that the overexpression of PDGF-B with increased PDGFRb phosphorylation characterized a specific subclass of GBM that comprises nearly 30% of human GBM.12 Because PDGF-B activates PDGFR-a and PDGFR-b and is a chemoattractant of glial and mesenchymal cells,13 PDGF-B overexpressed in GBM may chemoattract a range of intermingling nonneoplastic parenchymal cells that were characteristically seen in high-grade brain tumors.14 Along this line, many PDGFRapositive OPC-like cells were recruited toward PDGFBeinduced rodent glioma.15 However, the mechanism of GBM-derived PDGF-B to induce cancer stroma, and the functional implication of tumor microenvironment defined by cancer stroma, remains to be elucidated for the establishment of a novel therapeutic target of GBM. In the present study, we hypothesized that GBM-derived PDGF-B induces tumor microenvironment and may contribute to the aggressive behavior of GBM. For this end, we developed a PDGF-BBeoverexpressing glioma cell line (GL261-PDGF-BB) and implanted them into the mouse brain. Furthermore, the same transplantation was conducted after the inactivation of host PDGFRa signaling by conditionally induced Pdgfra gene inactivation. Paracrine PDGF signal from glioma cells was shown to recruit many OPCs, and the recruited OPCs were implicated in the regulation of vascular leakage and hyperplasia with remodeling of extracellular matrix.

Materials and Methods All experimental animal procedures were approved by the Committee for Institutional Animal Care and Use at the University of Toyama (Toyama, Japan). All of our study protocols were approved by the Ethics Committee of the University of Toyama.

The Generation of Pdgfra Conditionally Inactive Mice Mutant mouse harboring a genetically mutated Pdgfra gene, in which exons 4 and 5 of Pdgfra were flanked by

2

two loxP sequences (Pdgfraflox/flox), was generated as follows. Briefly, a BAC genomic clone (RP24-148N4), Q4 originating from the DNA of a C57BL/6 mouse and containing Pdgfra, was obtained from the BACPAC Resource Center CHORI. The constructed targeting vector included a DNA fragment containing a loxP sequence and pgk-Neo cassette flanked by two Flp recognition target (frt) sites16 and pMC1DTABGHA with a bovine growth hormoneederived polyadenylation signal sequence.17 The embryonic stem cell line RENKA, derived from the C57BL/6N strain,18 was used and the resulting targeting vector was electroporated, as described previously.19 The obtained male chimeric mouse was crossed with a female CAG-FLPe deleter mouse20 to remove the pgk-Neo selection cassette and obtain the heterozygous Pdgfraflox/þ strain. Genotyping was performed by PCR using the following primers: 50 -ATGCCAAACTCTGCCT- Q5 GATTGA-30 and 50 -CTCACGGAACCCCCACAAC-30 . Flox mice were crossbred with chicken b-actin-promoter/CMVenhanceredriven Cre-transgenic mice (CAGG-CreERþ/; The Jackson Laboratory, Bar Harbor, ME; CreER mouse)21 that harbor the fusion gene Cre recombinase and estrogen receptor (CreER). The resulting offspring were mice harboring CAGGCreERþ//Pdgfraflox/flox (CreER-Flox mice) or Pdgfraflox/flox (Flox mice). Mice with a systemic Pdgfra inactivation [PDGFRa-knockout (KO) mice] were obtained by tamoxifen (Sigma-Aldrich, St. Louis, MO) administration to CreER-Flox mice. Flox mice were treated identically and used as controls.

Generation of PDGF-BB Overexpression Glioma Cell Line To monitor the cells expressing the mouse PDGF-BB, we introduced IRES-EGFP DNA fragment from pIRES2EGFP (Clontech, Mountain View, CA) after the coding region of mouse Pdgfb expression plasmid pBLAST49mPDGFB (full) v24 (InvivoGen, San Diego, CA). The 0.12-Kbp DNA fragment containing mouse Pdgfb was amplified with primers PIE-U1 (50 -GGGGAATTCGGTGACCATTCGGACGGTGAG-30 ) and PIE-L1 (50 -GGGGGATCCCTAGGCTCCGAGGGTCTCC-30 ) using pBLAST49-mPDGFB (full) v24 as a template. The amplified DNA fragment was subcloned into the EcoRV site of the pLITMUS28 (New England Biolabs, Herts, UK) to yield the plasmid pLITPIE. The 0.12-Kbp EcoRI-BglII DNA fragment from pLITPIE was cloned between EcoRI and BamHI sites of the pIRES2-EGFP to yield the plasmid pIRES-PIE. The 1.4-kb BstEII-NotI (blunt) fragment from pIRES-PIE was ligated with the 3.6-kb BstEII-NheI (blunt) DNA fragment from pBLAST49-mPDGFB (full) v24 to yield the plasmid pPIE. Plasmid pPIE and empty vector were transfected to the GL261 glioma cell line (kindly provided by Dr. Atsushi Natsume, Department of Neurosurgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan) by electroporation (NEPA21 Electroporator; Nepa Gene, Chiba, Japan), according to the manufacturer’s procedure. Transfected cells were single cloned, and passaged several times to obtain

ajp.amjpathol.org

-

The American Journal of Pathology

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248

249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310

Cancer Stroma by PDGF Signal in Glioma stable expression clones. Resulting PDGF-BB overexpression and vector cell lines were termed GL261-PDGF-BB and GL261-vector, respectively.

Immunoblotting

Q6

Sample preparations and all other procedures for Western blot analysis have been described elsewhere.22 Briefly, cultured cells were lysed using radioimmunoprecipitation assay buffer (Thermo Fisher, Waltham, MA) on ice. Whole cell lysates were separated by SDS-PAGE and then electrophoretically transferred to polyvinylidene difluoride membranes. The membranes were blocked for 1 hour at room temperature in a 5% milk containing 50 mmol/L Tris buffer with 150 mmol/L NaCl and 0.05% Tween 20. The membranes were then probed with rabbit anti-PDGFRa (1:1000; Santa Cruz Biotechnology, Santa Cruz, CA), rabbit anti-PDGFRb (1:500; Merck Millipore, Billerica, MA), and mouse antieb-actin (1:20,000; Sigma-Aldrich, St. Louis, MO) antibodies. The membranes were washed in a 50 mmol/L Tris buffer containing 150 mmol/L NaCl and 0.05% Tween 20, and were incubated with the appropriate horseradish peroxidaseeconjugated secondary antibodies. Immunoreactive bands were detected using enhanced chemiluminescence reagents (GE Healthcare Bio-Sciences AB, Uppsala, Sweden), according to the manufacturer’s instructions.

RNA Extraction, cDNA Synthesis, and Real-Time PCR

Q7

Sample preparations and all other procedures for real-time PCR have been described elsewhere.23 Briefly, total RNA was isolated from cultured cells using miRNA easy Mini Kit (Qiagen, Valencia, CA), according to the manufacturer’s instructions. Total RNA (150 mg) was reverse transcribed, as detailed in the PrimeScript RT reagent kit protocol (Takara Bio Inc., Shiga, Japan). For real-time PCR performed with a Takara Thermal Cycler Dice Real Time System TP800 (Takara Bio Inc.), cDNAs were diluted 1:25 in the reaction mixture consisting of SYBR Premix EX Taq II (Takara Bio Inc.). The real-time PCR program consisted of hot start enzyme activation at 95 C for 10 seconds and 45 cycles of amplification at 95 C for 10 seconds and 60 C for 40 seconds. Finally, to obtain the dissociation curve, a final cycle was performed at 95 C for 1 minute, 60 C for 30 seconds, and then 95 C for 10 seconds. Primers were as follows: Pdgfb, 50 -TGCTGAGCGACCACTCCATC-30 and 50 -CTCGGGTCATGTTCAAGTCCA-30 ; Pdgfra, 50 -AGCAAACATCTTGACTTGGGAACA-30 and 50 -ACTTGCATCATTCCCGGACAC-30 ; Pdgfrb, 50 -GAACGACCATGGCGATGAGA-30 and 50 -GCATCGGATAAGCCTCGAACA-30 ; and Actb, 50 -CATCCGTAAAGACCTCTATGCCAAC-30 and 50 -ATGGAGCCACCGATCCACA-30 . For data analysis, mouse b-actin (Actb) housekeeping gene was used as an internal control. Induction values were calculated using analysis software (Takara Bio Inc.).

The American Journal of Pathology

-

Colony Formation Assay GL261-PDGF-BB cells and GL261-vector cells were cultured in a soft agarebased three-dimensional culture system, CytoSelect 96-Well in Vitro Tumor Sensitivity Assay (Cell Biolabs, San Diego, CA). Tumor cells were seeded at 5  103 cells per well in the 96-well plate, and were photographed under the conventional microscopy on day 1 after starting of culture. Six days later, tumor cellederived colonies were microphotographed, and the growth rate was measured by OD 570 nm after MTT reagent reaction, according to the manufacturer’s procedure.

Tumor Implantation and Tamoxifen Administration Tamoxifen (Sigma-Aldrich) was orally administered once a day (225 mg/kg body weight) for 5 consecutive days, starting on 2 days before tumor implantation. Eight-monthold male mice were used for the experiments. All surgical procedures were conducted under anesthesia with sodium pentobarbital (i.p. injection, 100 mg$kg1). Using a small Q8 dental drill, a hole was made in the skull at the correct stereotaxic coordinates. After this, the dura matter was removed using small curved forceps, and exposure of the meninges was confirmed by observation of a small amount of cerebrospinal fluid at the exposed position. A needle (internal diameter, 0.13 mm) was set in the stereotaxic device (David Kopf Instruments, Tujunga, CA), connected to a Hamilton microsyringe via polyethylene tubes filled with phosphate-buffered saline, and set on a microsyringe pump (CMA 400; Harvard Apparatus, Holliston, MA). The needle was inserted to the targeted location in the striatum at 1.0, 2.0, and 3.0 mm at anteroposterior relative to bregma, mediolateral, and dorsoventral from surface of the brain, respectively. Tumor cell suspension (1 mL; 1.0  105 cells$mL1) was infused at a flow rate of 0.1 mL$minute1. After the infusion, the needle was retained at the same position for 10 minutes to ensure no backflow. Under deep anesthesia with sodium pentobarbital (i.p. injection, 50 mg/kg body weight; Dainippon Sumitomo, Osaka, Japan), mice were transcardially perfused with phosphate-buffered saline, followed by perfusion and immersion in 4% paraformaldehyde at day 20 after tumor implantation.

Immunohistochemistry and Immunofluorescence Staining of Paraffin-Embedded Sections For paraffin section preparation, brains were immediately fixed with 4% paraformaldehyde in 0.1 mol/L phosphate buffer (pH 7.4), dehydrated with a graded series of ethanol solutions, and embedded in paraffin. The sections (5 mm thick) were subjected to either hematoxylin and eosin staining or immunostaining. Immunohistochemistry and immunofluorescence were performed, as described previously.24,25 Briefly, the sections were incubated at 4 C overnight with the following primary

ajp.amjpathol.org

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

3

311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372

A

B

Pdgfb mRNA

8000 6000 4000 2000 0

Pdgfra mRNA 5

Fold Change (AU)

Fold Change (AU)

10,000

1

C

2 3 4 GL261-PDGF-BB

1

4 3 2 1 0

2 3 4 GL261-Vector

1

2 3 4 GL261-PDGF-BB

1

2 3 4 GL261-Vector

D

Pdgfrb mRNA 8 6

PDGFRα 4 PDGFRβ 2 β-Actin 0

E

1

2 3 4 GL261-PDGF-BB

2 3 4 GL261-Vector

GL261-Vector

1 2 3 4 GL261-PDGF-BB

Day6

OD 570 nm Day 6

2 3 4 GL261-Vector

0.20 0.15 0.10

Figure 1 In vitro characterization of the platelet-derived growth factor (PDGF)-BB overexpression glioma cell lines. AeC: Real-time PCR analyses for the Pdgfb and Pdgfrs in glioma cell lines transfected with PDGF-BBeoverexpressing vector (GL261-PDGF-BB) and with control vector (GL261-vector). Four clones were examined from GL261-PDGF-BB and Gl261-vector, respectively. A: Pdgfb mRNA expression is much higher in GL261-PDGF-BB clones than in GL261-vector clones. B: Pdgf receptor a (Pdgfra) mRNA expression is lower in Gl26L-PDGF-BB than in GL261vector. C: Pdgfrb mRNA expression is comparable in all clones, except for one GL261-vector clone. D: Western blot analysis shows that PDGFRa is abundant in GL261-vector than in GL261-PDGFBB. PDGFRb was undetected in all examined cell lines. The lysates from cultured mouse skin fibroblasts were used as positive controls. Equal sample loading was confirmed by b-actin. E: Representative photographs of GL261-vector and GL261PDGF-BB cell lines in colony formation assay. F: Colony formation assay of four clones of GL261PDGF-BB and Gl261-vector. n Z 6 from each clone (AeC). Scale bar Z 200 mm (E). AU, arbitrary unit.

Q13

0.05 0.00

1

2 3 4 GL261-PDGF-BB

antibodies: goat polyclonal anti-PDGFRa (1:1000; Neuromics, Edina, MN), goat polyclonal anti-PDGFRb antibody (1:100; R&D, Minneapolis, MN), rabbit polyclonal anti-Olig2 (1:500; IBL, Hamburg, Germany), goat polyclonal anti-Sox10 (1:100; Santa Cruz Biotechnology), rat monoclonal anti-CD31 (1:100; Dianova, Hamburg, Germany), rabbit polyclonal anti-collagen type IV (1:500; Millipore, Darmstadt, Germany), rabbit polyclonal antiematrix metalloproteinase (MMP)-9 (1:100; Abcam, Cambridge, UK), and rabbit polyclonal antieclaudin-5 (1:100; Invitrogen, Carlsbad, CA). Colorimetric immunostaining was conducted using the appropriate Histofine Simple Stain Mice System (Nichirei Biosciences, Tokyo, Japan) and 3,30 diaminobenzidine tetrahydrochloride (Dako, Glostrup, Denmark) reaction. The leakage of serum protein was detected by immunohistochemistry for IgG using an anti-IgG detection system from Histofine Simple Stain Mice System (Nichirei Biosciences), as per the supplier’s protocol. For the fluorometric immunostaining, the positive immunoreactions were visualized using the following fluorescent dyeeconjugated secondary antibodies that were applied at room temperature for 3 hours: donkey anti-goat IgG conjugated with Alexa Fluor 488, donkey anti-rabbit IgG conjugated with Alexa Fluor 488, 594, or 633,

4

1

F

Day 1

GL261-PDGF-BB

1

Fibroblast

Fold Change (AU)

373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434

Zheng et al

1

2 3 4 GL261-Vector

and donkey anti-rat IgG conjugated with Alexa Fluor 594. These secondary antibodies were used at 1:500 dilutions and were obtained from Life Technologies Corporation (Carlsbad, CA). Images were captured by a microscopy system (BX 51; Olympus, Tokyo, Japan) connected to a digital camera (DP70; Olympus) and TCS SP5 confocal system (Leica, Heidelberg, Germany), and were processed using Photoshop software version 7.0 (Adobe, San Jose, CA).

Zymography Taken out brains under deep anesthesia were immediately Q9 snap frozen into liquid nitrogen. Frozen brains were serially cut with 30 mm intervals in the coronal plane to obtain a representative cut surface of the tumor using a Leica CM3000 cryostat (Leica Microsystems, Wetzlar, Germany). Then, the surrounding nonneoplastic tissues were manually trimmed off the frozen brain; at this step, we saved OPC-rich area in tumor side. Then, six of the frozen sections (30 mm thick) were cut from each mouse, and lysed in the sample buffer of Gelatin-Zymography Kit (Cosmo Bio, Tokyo, Japan). Protein concentration was measured by OD 280 nm

ajp.amjpathol.org

-

The American Journal of Pathology

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496

A

B

Olig2

D

E

GL261-PDGF-BB

GL261-Vector

10

***

GL261-PDGF-BB

GL261-Vector

600 PDGFRα

20

Olig2+ Cell No./Field

400 200

0

***

0 GL261 PDGF-BB

GL261 Vector

F

GL261 PDGF-BB

GL261 Vector

G

H 80

25 RBC No./mm

20 15 10 5

60 Hemorrage

Tumor Volume (mm3)

Olig2

800

30

PDGFRα+ Area (%)

H&E

GL261-Vector

GL261-PDGF-BB

H&E

C

print & web 4C=FPO

497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558

Cancer Stroma by PDGF Signal in Glioma

40 20

*

***

0

0 GL261 PDGF-BB

GL261 Vector

GL261 PDGF-BB

GL261 Vector

Figure 2 Histological characterization of platelet-derived growth factor (PDGF)-BBeoverexpressing glioma and control glioma after implantation in the brain of wild-type mouse. A and B: Hematoxylin and eosin (H&E) staining (A) and immunohistochemistry (B) for Olig2 of wild-type mouse brain at day 20 after the implantation of GL261-PDGF-BB and GL261-vector. C: Tumor volume (mm3) of GL261-PDGF-BB and GL261-vector at day 20 after implantation. D: Numbers of Olig2-positive cells per field in the marginal region of tumor. E: Immunohistochemistry for PDGF receptor a (PDGFRa) in the marginal region of tumors of GL261-PDGF-BB and GL261-vector. The images are from adjacent sections of boxed area in A and B. F: Percentage of PDGFRa-positive area per field in the marginal region of tumor of GL261-PDGF-BB and GL261vector. G: Numbers of extravasated red blood cells (RBCs) along the entire circle of tumor of GL261-PDGF-BB and GL261-vector. Counted numbers of RBCs were divided by the circumferential distance of each tumor for the comparison. H: Higher magnification of boxed area of H&E staining in the marginal region of tumors of GL261-PDGF-BB and GL261-vector. Dotted lines in E and H indicate the edge of tumor. Error bars represent SEM (C, D, F, and G). n Z 16 from four clones (C, GL261-PDGF-BB); n Z 14 from four clones (C, GL261-vector); n Z 8, two fields from four mice (D and F); n Z 4 mice (G). *P < 0.05, ***P < 0.001. Scale bar: 500 mm (A); 100 mm (E and H).

for ensuring equal protein volume loading. Samples were sonicated to reduce viscosity, and electrophoresis and complete blood cell count stainings were performed according to the manufacturer’s procedure. Translucent MMP enzymatic

The American Journal of Pathology

-

reaction bans were measured by ImageJ software (NIH, Q10 Bethesda, MD; http://imagej.nih.gov/ij), and normalized to the major protein bands of complete blood cell count staining.

ajp.amjpathol.org

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

5

559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620

A

Olig2

H&E

Flox

D PDGFRα-KO

Flox

PDGFRα-KO

Flox

PDGFRα-KO

Flox

PDGFRα-KO

PDGFRα

Hemorrage

Flox

F

IgG

Boxed Area

E

IgG

30 20 10

60 40 20

***

0

0 Flox

α-KO

J 80

30

60

20 10

α-KO

50

40 20

30 20

* 0

α-KO

40

10

*** Flox

Olig2

K

40

0 Flox

IgG

IgG+ Area (%)

Olig2+ Cell No./Field (×10)

Tumor Volume (mm3)

40

I 80

Olig2

RBC No./mm

H

50

PDGFRα+ Area (%)

G

6

Olig2

PDGFRα-KO

H&E

B

C

print & web 4C/FPO

621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682

Zheng et al

***

0 Flox

ajp.amjpathol.org

-

α-KO

Flox

α-KO

The American Journal of Pathology

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744

745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806

Cancer Stroma by PDGF Signal in Glioma 807 808 809 Images were obtained by a microscopy system (BX 51; 810 Olympus) connected to a digital camera (DP70; Olympus). PDGF-BBeOverexpressing Glioma Induces Massive 811 Morphological and morphometric analyses were performed Recruitment of OPCs and Exacerbates Glioma-Related 812 using a BioRevo BZ-9000 microscope (Keyence, Osaka, Hemorrhage 813 Q11 Japan), BZ-II Analyzer software (Keyence), and ImageJ soft814 ware. The tumor volume (mm3) was estimated on the basis of First, we implanted four clones of glioma cells from lines 815 the tissue section prepared from largest cut surface of each with and without PDGF-BB overexpression into the 816 817 tumor using the following equation: volume Z 4/3  p  striatum of wild-type mouse. The tumor volumes after 818 long axis  short axis2.26 transplantation were significantly larger in GL261-PDGF-BB than in GL261-vector (Figure 2, AeC). Olig2 is a marker ½F2 819 820 of oligodendrocyte lineage, in which lineage, PDGFRa821 Statistical Analysis 8 positive cells correspond to OPCs. At higher magnifi822 cation, we found that many Olig2 and PDGFRa-positive 823 Statistical significance was determined using a t-test. P < 0.05 OPCs recruited and surrounded the tumor mass in all of 824 was considered statistically significant. Quantified data are GL261-PDGF-BB glioma, but none of GL261-vector 825 presented as means  SEM. Graphs were drawn using glioma (Figure 2, A, B, and DeF). Furthermore, we 826 GraphPad Prism 6 (GraphPad Software, Inc., La Jolla, CA). found frequent hemorrhages around the tumor mass of 827 GL261-PDGF-BB, but not GL261-vector (Figure 2H). 828 829 The hemorrhagic foci were closely distributed to the Results 830 recruited OPCs. The number of extravasated red blood 831 cells around tumor was substantially larger in GL261Characterization of PDGF-BBeOverexpressing Glioma 832 PDGF-BB than in GL261-vector (Figure 2G). Cell Line 833 834 Glioma cell lines transfected with either PDGF-BBe Pdgfra Inactivation Suppresses OPC Recruitment, 835 overexpressing vector (GL261-PDGF-BB) or control vector Hemorrhage, and Vascular Permeability 836 (GL261-vector) were characterized in vitro before using in 837 transplantation into mouse brain. In real-time PCR analyses, To understand the relevance of PDGFRa signaling in the 838 four of the GL261-PDGF-BB cell lines expressed higher levels induction of cancer stroma, we next implanted GL261839 of Pdgfb mRNA compared with four of the GL261-vector cell PDGF-BB and GL261-vector clones into the striatum of 840 PDGFRa-KO and Flox mice. The tumor volumes after im½F1 lines (Figure 1A). The levels of Pdgfra mRNA expression were 841 842 plantation of GL261-PDGF-BB were similar between two lower in GL261-PDGF-BB cell lines than in GL261-vector cell 843 genotypes, and Pdgfra inactivation in host tissue did not lines (Figure 1B). Pdgfrb expression was low at comparable 844 significantly affect the growth of implants (P Z 0.146) levels in all examined clones, except that one GL261-vector cell (Figure 3, A, B, and G). The tumor volumes of GL261-vector ½F3 845 line showed Pdgfrb mRNA at an exceptionally high level 846 (Figure 1C). GL261-PDGF-BB clones released significant were also equivalent between the two genotypes of mice 847 3 3 amounts of PDGF-BB into culture media in enzyme-linked (Flox, 5.06  1.60 mm ; PDGFRa-KO, 8.02  1.91 mm ; 848 immunosorbent assay (data not shown). In Western blot anaP Z 0.276, data not shown). After GL261-PDGF-BB im849 lyses, PDGFRa protein expression levels were lower in plantation, we detected many recruited OPCs and hemor850 GL261-PDGF-BB than in GL261-vector cell lines (Figure 1D). rhagic foci around the tumor of Flox mice (Figure 3, A, C, D, 851 The expression of PDGFRb was undetectable in all cell lines and HeJ), as previously observed after implantation in 852 (Figure 1D). The colony-forming activity in GL261-vector cell wild-type mice (Figure 2, A and DeH). In contrast, the 853 854 lines was significantly higher than that in GL261-PDGF-BB recruitment of either Olig2-positive cells of oligoden855 cell lines (P < 0.001, four to five wells from each of the four drocyte lineage or PDGFRa-positive OPCs was largely 856 857 858 Figure 3 Histological characterization of platelet-derived growth factor (PDGF)-BBeoverexpressing glioma (GL261-PDGF-BB) 20 days after implantation in the 859 brain of PDGF receptor a (PDGFRa) knockout mouse (PDGFRa-KO) and PDGFRa-preserving control (Flox) mice. A and B: Hematoxylin and eosin (H&E) staining and immunohistochemistry for Olig2 of GL261-PDGF-BB in PDGFRa-KO and Flox mice. C: Immunohistochemistry for PDGFRa in the marginal region of tumor in Flox and 860 PDGFRa-KO mice. The images are from adjacent sections of boxed area in A and B. D: H&E staining in the marginal region of tumors in Flox and PDGFRa-KO. The 861 images are enlarged boxed area in A and B. Inset: Higher magnification of hemorrhage. E: Immunohistochemistry for IgG in Flox and PDGFRa-KO mice. F: Higher- Q14 862 magnification images of immunohistochemistry for IgG and Olig2 in the marginal region of tumor in Flox and PDGFRa-KO mice. Images correspond to the boxed 863 3 areas in E, respectively. G: Tumor volume (mm ) of GL261-PDGF-BB in Flox and PDGFRa-KO (a-KO). H: Number of Olig2-positive cells per field in the marginal region 864 of tumor. I: Percentage of PDGFRa-positive area per field in the marginal region of tumor. J: Numbers of extravasated red blood cells (RBCs) along the entire circle of 865 each tumor. Counted numbers of RBCs were divided by the circumferential distance of each tumor for the comparison. K: Percentage of PDGFRa-positive area per field 866 in the magical region of tumor. Dotted lines in C, D, and F indicate the edge of tumor. Error bars represent SEM (GeK). n Z 4 mice (G and J); n Z 8, two fields 867 from four mice (H, I, and K). *P < 0.05, ***P < 0.001. Scale bar: 500 mm (A, B, and E); 100 mm (C, D, and F). 868

Image Analysis and Quantification

The American Journal of Pathology

-

clones in GL261-PDGF-BB and GL261-vector cell lines, respectively; t-test) (Figure 1, E and F).

ajp.amjpathol.org

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

7

Flox PDGFRα-KO

Blood Vessel No./Field

C

Claudin-5 Mean Gray Value (×10 3)

D

F

Collagen Type IV Positive Area (%)

Flox

I

L

Flox

Stratification of PDGFRβ+ Cell

PDGFRα-KO

Collagen Type IV Thickness (μm)

H

Collagen IV, Border

***

10 5 0 25 20 15 10 5 0

Flox

α-KO

Flox

α-KO

20

***

15 10 5 0

Flox

α-KO

J

G

CD31/PDGFRβ/Collagen Type IV

K

E

15

Flox

B

Blood Vessel Diameter (μm)

Claudin-5

PDGFRα-KO

CD31

MMP9

20 15 10

***

5 0

Flox

α-KO

Flox

A

(Figure 3, D and J). We examined the leakage of serum protein using immunostaining for IgG. IgG-positive area was observed in the marginal region of tumor, corresponding to the regions where many Olig2-positive cells

30 20 10 0

*** Flox

α-KO

4 3 2

***

1 0

PDGFRα-KO

suppressed around the tumor in PDGFRa-KO (Figure 3, B, C, H, and I). In addition, hemorrhage represented by extravasated red blood cells in the marginal region of tumor was significantly suppressed in PDGFRa-KO

print & web 4C/FPO

Flox

α-KO

M

PDGFRα-KO

Flox

α-KO

300 200

***

100 0 Flox

8

SOX10/MMP9

400 MMP9 Positive Cell No./Field

869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930

Zheng et al

Pro-MMP9

Pro-MMP2

α-KO

ajp.amjpathol.org

-

The American Journal of Pathology

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992

Cancer Stroma by PDGF Signal in Glioma 993 recruited, and the staining was more intense in Flox than with PDGFRa-KO in zymography (Figure 4M), whereas the 994 in PDGFRa-KO (Figure 3, E, F, and K). difference was not significant (P Z 0.3349, n Z 4 in Flox and 995 n Z 3 in PDGFRa-KO mice). The proeMMP-2 activity was 996 similar between Flox and PDGFRa-KO. Glioma-Associated OPC Induces Vessel Dilatation and 997 Permeability and Down-Regulates Claudin-5 998 Discussion 999 The diameter of blood vessel within GL261-PDGF-BB glioma 1000 1001 appeared to be larger in Flox than in PDGFRa-KO mice Herein, we implanted GL261-vector and GL261-PDGF-BB 1002 (Figure 3, A and B). Morphometrically, the diameter of CD31glioma cells into the brain of wild-type mouse, and found that 1003 positive blood vessels was significantly decreased in the volume of GL261-PDGF-BB glioma was extensively 1004 ½F4 PDGFRa-KO compared with Flox (Figure 4, A and B). The larger than that of GL261-vector glioma. The large tumor in 1005 density of the blood vessel was equivalent between Flox and GL261-PDGF-BB was accompanied by many Olig2/ 1006 PDGFRa-KO (Figure 4, A and C). When we focus on the PDGFRa-positive OPCs, and showed increased hemorrhage. 1007 blood vessels in the margin but not inside of the tumor, the Therefore, PDGF-BBeoverexpressing glioma cells showed 1008 immunofluorescence of claudin-5, a tight junction complex more high-grade malignant features, and these findings were 1009 protein that maintains the blood tumor barrier,27 was less in accordance with a previous report showing that the 1010 strong in Flox than in PDGFRa-KO (Figure 4, A and D). strength of PDGFR signaling is well correlated with high1011 1012 grade characteristics of glioma.30 Previously, the host 1013 tissueederived PDGF-CC was shown to recruit OPCs toward Glioma-Associated OPC Induces Remodeling of 1014 orthotopic GL261 glioma by inactivating host Pdgfc or Collagen 1015 Pdgfra genes.31 In our present study, an inactivation of host 1016 Pdgfra resulted in the substantial decrease of OPC recruitCollagen type IV is one of the major components of the 1017 ment in GL261-PDGF-BB glioma, and it was clearly shown extracellular membrane in glioma, and is mainly synthesized 1018 28,29 that glioma-derived paracrine signal of PDGF-BB recruited by vascular endothelial cells. Collagen type IV expression 1019 OPCs via PDGFRa activation in OPCs. Intriguingly, the was increased in the marginal region of GL261-PDGF-BB 1020 inactivation of host Pdgfra was accompanied by the decrease glioma, where many OPCs were recruited, but was low inside 1021 of hemorrhage and vascular leakage in PDGFRa-KO mouse, of the glioma in Flox mouse (some data not shown) (Figure 4, 1022 even though the glioma volume was comparable between E and G). Collagen type IV expression was at low level both 1023 1024 Flox and PDGFRa-KO mice. It was suggested that the outside and inside of glioma in PDGFRa-KO (Figure 4, E and 1025 paracrine PDGF signal of glioma could be causally related G). In the tumor border of Flox, but not PDGFRa-KO, triple 1026 with hemorrhage and brain edema that contribute to the staining for PDGFRb/collagen type IV/CD31 revealed that the 1027 serious prognostic factors of GBM, independently of the collagen type IVepositive basement membrane was aberrantly 1028 tumor volume.2 thick and multilayered (Figure 4, F and H), and that the 1029 PDGFRb-positive pericytes were detached from endothelial Activated PDGFRa after intracerebral hemorrhage 1030 cells and were interspersed within multilayer basement meminduced blood-brain barrier (BBB) dysfunction in 1031 brane (Figure 4, F and I). Next, we examined the expression of ischemic stroke and cerebral hemorrhage, in which MMP1032 4 9,32 glioma invasion-related protein, MMP-9. 9 was critically involved. Many recruited Similarly, OPCs were shown 1033 OPCs were highly immunoreactive for MMP-9 around tumor to release MMP-9 under acute phase of white matter 1034 of Flox, but not in PDGFRa-KO, whereas glioma cells were injury to initiate the early onset of BBB breakdown.33 We 1035 1036 similarly immunoreactive to MMP-9 in both genotypes of found frequent hemorrhage and increased IgG leakage in 1037 mice (Figure 4, J and K). We confirmed that most of the the marginal region of GL261-PDGF-BB glioma, where 1038 Sox10-positive OPCs expressed MMP-9 (Figure 4L). The MMP-9eexpressing OPCs were recruited. Specific to this 1039 proeMMP-9 activity tended to be higher in Flox compared area, aberrant vasculatures were found that were 1040 1041 1042 Figure 4 Morphological analyses of the parameters related with blood vessel formation and extracellular matrix in platelet-derived growth factor (PDGF)1043 BBeoverexpressing glioma. A: Immunohistochemistry for CD31 and immunofluorescence for claudin-5 (red) in the marginal region of tumor of Flox and PDGF 1044 receptor a knockout (PDGFRa-KO). B and C: Blood vessel diameter and numbers measured from the images of CD31 staining, respectively. D: Claudin-5 1045 immunofluorescence intensity per blood vessel was converted into gray color values, and mean gray value per blood vessel was calculated. E: Immunohistochemistry for collagen type IV in the border part of tumor of Flox and PDGFRa-KO mice. F: Triple immunofluorescence staining for PDGFRa (green), 1046 collagen type IV (magenta), and CD31 (red) in the border part of tumor of Flox and PDGFRa-KO. G: Percentage of collagen type IVepositive area per field. H: 1047 Thickness of the collagen type IVepositive basement membrane of blood vessel. I: Measurement of stratification of the PDGFRb-positive cells around the blood 1048 vessels. J: Immunohistochemistry for matrix metalloproteinase (MMP)-9 in the marginal region of the tumor of PDGFRa-KO and Flox mouse. K: Number of MMP1049 9epositive cells in the marginal region per field. L: Double-immunofluorescence staining for SOX10 (green) and MMP-9 (red) in the marginal region of the 1050 tumor of PDGFRa-KO and Flox mice. M: A representative image of the gelatin zymography gel. Dotted lines in A, J, and L indicate the edge of tumor. Error bars 1051 represent SEM (BeD, GeI, and K). n Z 16, each four fields from four mice (B and C); n Z 77 (D, blood vessels from nine Flox mice); n Z 83 (D, blood vessels 1052 from eight PDGFRa-KO mice); n Z 8, two fields each from four mice (G and K). ***P < 0.001. Scale bars: 100 mm (A, CD31); 50 mm [A, claudin-5, E, F (left 1053 panels), J, and L); 20 mm [F (right panels)]. 1054

The American Journal of Pathology

-

ajp.amjpathol.org

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

9

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 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 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178

Zheng et al accompanied by a multilayered thick collagen; they contain interspersed pericytes, whose vascular phenotypes were similar to those reported in previous experimentally induced glioma.34,35 These vascular abnormalities in the tumor border of GL261-PDGF-BB in Flox mice were mostly normalized in PDGFRa KO. Taken altogether into consideration, recruited OPCs by paracrine PDGF signal were indicated to be involved in the BBB dysfunction to cause hemorrhage and vascular leakage. Decreased expression of claudin-5, a tight junction complex protein, that maintains the blood-tumor barrier,27 could be an underlying cause of BBB dysfunction; however, the mechanism of decrease remains unknown. Glioma cellederived MMP-9 was not sufficient to induce hemorrhage within tumor, suggesting that OPC-derived additional factors were required for the vascular phenotype. MMPs strongly correlate with malignant progression of glioma,36,37 among which MMP-9 mediates glioma cell adhesion to the extracellular membrane and invasive growth along collagen type IV.38 In our study, a cancer stroma was established with abundant collagen type IV and MMP-9 expression around GL261-PDGF-BB glioma in wild-type and Flox mice, but not in that of PDGFRa-KO mice; however, the tumor volume was similar between Pdgfra-preserving mice and PDGFRa-KO mice. Different PDGF signal in glioma may differentially regulate invasive tumor growth and associating events, like hemorrhage and edema; further studies are needed. Colony-forming activities were significantly lower in GL261-PDGF-BB than in GL261-vector in vitro, which was compatible with low expression levels of PDGFRs in GL261-PDGF-BB glioma cells. The volume of either GL261-PDGF-BB or GL261-vector glioma was comparable between Flox and PDGFRa-KO mice. Accordingly, either of autocrine PDGF-BB signal in glioma or host PDGFRa signaling was not a major determinant of GL261 glioma volume in our study. These observations implied that activated host PDGFRb signaling might have been involved in the larger GL261-PDGF-BB than GL261-vector in wildtype mice. One of the subclasses of GBM was characterized by high levels of PDGF-B ligand expression and PDGFRb phosphorylation.12 The PDGFRb was insignificant in our GL261 cells. Further studies are required to explore the contribution of the two types of PDGFRs in PDGF-BBeoverexpressing glioma. Our study showed that glioma-derived paracrine signal of PDGF-B extensively recruits OPCs via the activation of PDGFRa, and that the recruited OPCs were implicated in the formation of cancer stroma that defines microenvironment, such as BBB dysfunction, hemorrhages, and vascular abnormality with extracellular membrane remodeling. PDGFRa inactivation could normalize vascular phenotype induced by PDGF-BB secreted from glioma, and vascular normalization is crucial for radiation therapy of GBM.34 Our findings may open prospects to develop a new

10

therapeutic target for GBM and other hypervascular brain tumors.

Acknowledgments We thank Dr. Atsushi Natsume (Graduate School of Medicine, Nagoya University) for the GL261 glioma cell line; Prof. Yoshimichi Ueda (Kanazawa Medical University) for the valuable scientific discussion; and members of the Department of Pathology and Life Science Research Center (University of Toyama) for thoughtful discussion and careful animal care. Y.I., S.Y., and M.S. conceived and designed the project; Y.Z., Y.I., S.Y., Y.S., N.V.D., H.N., H.M., and R.I. performed experiments; and Y.I., S.Y., and M.S. wrote the manuscript with significant contributions from Y.Z. and T.H.

References 1. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 2007, 114: 97e109 2. Velander AJ, DeAngelis LM, Navi BB: Intracranial hemorrhage in patients with cancer. Curr Atheroscler Rep 2012, 14:373e381 3. Jones TS, Holland EC: Standard of care therapy for malignant glioma and its effect on tumor and stromal cells. Oncogene 2012, 31: 1995e2006 4. Mammoto T, Jiang A, Jiang E, Panigrahy D, Kieran MW, Mammoto A: Role of collagen matrix in tumor angiogenesis and glioblastoma multiforme progression. Am J Pathol 2013, 183: 1293e1305 5. Curry RC, Dahiya S, Alva Venur V, Raizer JJ, Ahluwalia MS: Bevacizumab in high-grade gliomas: past, present, and future. Expert Rev Anticancer Ther 2015, 15:387e397 6. Zhao Y, Adjei AA: Targeting angiogenesis in cancer therapy: moving beyond vascular endothelial growth factor. Oncologist 2015, 20: 660e673 7. Shim AH, Liu H, Focia PJ, Chen X, Lin PC, He X: Structures of a platelet-derived growth factor/propeptide complex and a plateletderived growth factor/receptor complex. Proc Natl Acad Sci U S A 2010, 107:11307e11312 8. Nishiyama A, Komitova M, Suzuki R, Zhu X: Polydendrocytes (NG2 cells): multifunctional cells with lineage plasticity. Nat Rev Neurosci 2009, 10:9e22 9. Su EJ, Fredriksson L, Geyer M, Folestad E, Cale J, Andrae J, Gao Y, Pietras K, Mann K, Yepes M, Strickland DK, Betsholtz C, Eriksson U, Lawrence DA: Activation of PDGF-CC by tissue plasminogen activator impairs blood-brain barrier integrity during ischemic stroke. Nat Med 2008, 14:731e737 10. Yao H, Duan M, Buch S: Cocaine-mediated induction of plateletderived growth factor: implication for increased vascular permeability. Blood 2011, 117:2538e2547 11. Heldin CH: Autocrine PDGF stimulation in malignancies. Ups J Med Sci 2012, 117:83e91 12. Brennan C, Momota H, Hambardzumyan D, Ozawa T, Tandon A, Pedraza A, Holland E: Glioblastoma subclasses can be defined by activity among signal transduction pathways and associated genomic alterations. PLoS One 2009, 4:e7752 13. Heldin CH, Westermark B: Mechanism of action and in vivo role of platelet-derived growth factor. Physiol Rev 1999, 79:1283e1316

ajp.amjpathol.org

-

The American Journal of Pathology

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240

1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288

Cancer Stroma by PDGF Signal in Glioma

Q12

14. Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H: The brain tumor microenvironment. Glia 2012, 60:502e514 15. Appolloni I, Calzolari F, Tutucci E, Caviglia S, Terrile M, Corte G, Malatesta P: PDGF-B induces a homogeneous class of oligodendrogliomas from embryonic neural progenitors. Int J Cancer 2009, 124:2251e2259 16. Takeuchi T, Nomura T, Tsujita M, Suzuki M, Fuse T, Mori H, Mishina M: Flp recombinase transgenic mice of C57BL/6 strain for conditional gene targeting. Biochem Biophys Res Commun 2002, 293: 953e957 17. Kitayama K, Abe M, Kakizaki T, Honma D, Natsume R, Fukaya M, Watanabe M, Miyazaki J, Mishina M, Sakimura K: Purkinje cellspecific and inducible gene recombination system generated from C57BL/6 mouse ES cells. Biochem Biophys Res Commun 2001, 281: 1134e1140 18. Fukaya M, Tsujita M, Yamazaki M, Kushiya E, Abe M, Akashi K, Natsume R, Kano M, Kamiya H, Watanabe M, Sakimura K: Abundant distribution of TARP gamma-8 in synaptic and extrasynaptic surface of hippocampal neurons and its major role in AMPA receptor expression on spines and dendrites. Eur J Neurosci 2006, 24:2177e2190 19. Miya K, Inoue R, Takata Y, Abe M, Natsume R, Sakimura K, Hongou K, Miyawaki T, Mori H: Serine racemase is predominantly localized in neurons in mouse brain. J Comp Neurol 2008, 510: 641e654 20. Kanki H, Suzuki H, Itohara S: High-efficiency CAG-FLPe deleter mice in C57BL/6J background. Exp Anim 2006, 55:137e141 21. Hayashi S, McMahon AP: Efficient recombination in diverse tissues by a tamoxifen-inducible form of Cre: a tool for temporally regulated gene activation/inactivation in the mouse. Dev Biol 2002, 244:305e318 22. Yamamoto S, Yoshino I, Shimazaki T, Murohashi M, Hevner RF, Lax I, Okano H, Shibuya M, Schlessinger J, Gotoh N: Essential role of Shp2-binding sites on FRS2alpha for corticogenesis and for FGF2dependent proliferation of neural progenitor cells. Proc Natl Acad Sci U S A 2005, 102:15983e15988 23. Yamamoto S, Niida S, Azuma E, Yanagibashi T, Muramatsu M, Huang TT, Sagara H, Higaki S, Ikutani M, Nagai Y, Takatsu K, Miyazaki K, Hamashima T, Mori H, Matsuda N, Ishii Y, Sasahara M: Inflammation-induced endothelial cell-derived extracellular vesicles modulate the cellular status of pericytes. Sci Rep 2015, 5:8505 24. Shen J, Ishii Y, Xu G, Dang TC, Hamashima T, Matsushima T, Yamamoto S, Hattori Y, Takatsuru Y, Nabekura J, Sasahara M: PDGFR-beta as a positive regulator of tissue repair in a mouse model of focal cerebral ischemia. J Cereb Blood Flow Metab 2012, 32: 353e367 25. Sato H, Ishii Y, Yamamoto S, Azuma E, Takahashi Y, Hamashima T, Umezawa A, Mori H, Kuroda S, Endo S, Sasahara M: PDGFR-beta plays a key role in the ectopic migration of neuroblasts in cerebral stroke. Stem Cells 2015. doi:10.1002/stem.2212

The American Journal of Pathology

-

26. Xue Y, Lim S, Yang Y, Wang Z, Jensen LD, Hedlund EM, Andersson P, Sasahara M, Larsson O, Galter D, Cao R, Hosaka K, Cao Y: PDGF-BB modulates hematopoiesis and tumor angiogenesis by inducing erythropoietin production in stromal cells. Nat Med 2012, 18:100e110 27. Musch MW, Walsh-Reitz MM, Chang EB: Roles of ZO-1, occludin, and actin in oxidant-induced barrier disruption. Am J Physiol Gastrointest Liver Physiol 2006, 290:G222eG231 28. Huijbers IJ, Iravani M, Popov S, Robertson D, Al-Sarraj S, Jones C, Isacke CM: A role for fibrillar collagen deposition and the collagen internalization receptor endo180 in glioma invasion. PLoS One 2010, 5:e9808 29. Zamecnik J: The extracellular space and matrix of gliomas. Acta Neuropathol 2005, 110:435e442 30. Shih AH, Dai C, Hu X, Rosenblum MK, Koutcher JA, Holland EC: Dose-dependent effects of platelet-derived growth factor-B on glial tumorigenesis. Cancer Res 2004, 64:4783e4789 31. Huang Y, Hoffman C, Rajappa P, Kim JH, Hu W, Huse J, Tang Z, Li X, Weksler B, Bromberg J, Lyden DC, Greenfield JP: Oligodendrocyte progenitor cells promote neovascularization in glioma by disrupting the blood-brain barrier. Cancer Res 2014, 74:1011e1021 32. Ma Q, Huang B, Khatibi N, Rolland W 2nd, Suzuki H, Zhang JH, Tang J: PDGFR-alpha inhibition preserves blood-brain barrier after intracerebral hemorrhage. Ann Neurol 2011, 70:920e931 33. Seo JH, Miyamoto N, Hayakawa K, Pham LD, Maki T, Ayata C, Kim KW, Lo EH, Arai K: Oligodendrocyte precursors induce early blood-brain barrier opening after white matter injury. J Clin Invest 2013, 123:782e786 34. Winkler F, Kozin SV, Tong RT, Chae SS, Booth MF, Garkavtsev I, Xu L, Hicklin DJ, Fukumura D, di Tomaso E, Munn LL, Jain RK: Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell 2004, 6:553e563 35. Kamoun WS, Ley CD, Farrar CT, Duyverman AM, Lahdenranta J, Lacorre DA, Batchelor TT, di Tomaso E, Duda DG, Munn LL, Fukumura D, Sorensen AG, Jain RK: Edema control by cediranib, a vascular endothelial growth factor receptor-targeted kinase inhibitor, prolongs survival despite persistent brain tumor growth in mice. J Clin Oncol 2009, 27:2542e2552 36. Wang M, Wang T, Liu S, Yoshida D, Teramoto A: The expression of matrix metalloproteinase-2 and -9 in human gliomas of different pathological grades. Brain Tumor Pathol 2003, 20:65e72 37. Levicar N, Nuttall RK, Lah TT: Proteases in brain tumour progression. Acta Neurochir (Wien) 2003, 145:825e838 38. Zhao Y, Xiao A, diPierro CG, Carpenter JE, Abdel-Fattah R, Redpath GT, Lopes MB, Hussaini IM: An extensive invasive intracranial human glioblastoma xenograft model: role of high level matrix metalloproteinase 9. Am J Pathol 2010, 176:3032e3049

ajp.amjpathol.org

SCO 5.4.0 DTD  AJPA2278_proof  2 March 2016  6:43 pm  EO: AJP15_0315

11

1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336