15
International Scholarly Research Network ISRN Oncology Volume 2011, Article ID 409308, 14 pages doi:10.5402/2011/409308 Research Article Tumor Vascular Morphology Undergoes Dramatic Changes during Outgrowth of B16 Melanoma While Proangiogenic Gene Expression Remains Unchanged Elise Langenkamp, 1, 2 Franziska M. vom Hagen, 3 Peter J. Zwiers, 1 Henk E. Moorlag, 1 Jan P. Schouten, 4 Hans-Peter Hammes, 3 Annette S. H. Gouw, 5 and Grietje Molema 1 1 Medical Biology Section, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands 2 Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden 3 Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany 4 Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands 5 Department of Pathology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands Correspondence should be addressed to Grietje Molema, [email protected] Received 3 August 2011; Accepted 7 September 2011 Academic Editors: S. Mohanam and H. Rizos Copyright © 2011 Elise Langenkamp et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. In established tumors, angiogenic endothelial cells (ECs) coexist next to “quiescent” EC in matured vessels. We hypothesized that angio-gene expression of B16.F10 melanoma would dier depending on the growth stage. Unraveling the spatiotemporal nature thereof is essential for drug regimen design aimed to aect multiple neovascularization stages. We determined the angiogenic phenotype—represented by 52 angio-genes—and vascular morphology of small, intermediate, and large s.c. growing mouse B16.F10 tumors and demonstrated that expression of these genes did not dier between the dierent growth stages. Yet vascular morphology changed dramatically from small vessels without lumen in small to larger vessels with increased lumen size in intermediate/large tumors. Separate analysis of these vascular morphologies revealed a significant dierence in αSMA expression in relation to vessel morphology, while no relation with VEGF, HIF-1α, nor Dll4 expression levels was observed. We conclude that the tumor vasculature remains actively engaged in angiogenesis during B16.F10 melanoma outgrowth and that the major change in tumor vascular morphology does not follow molecular concepts generated in other angiogenesis models. 1. Introduction Tumors depend on the recruitment of new blood vessels to grow beyond a certain size and to metastasize to other tissues. This process represents an attractive target for the treatment of cancer, and several strategies have been pursued to inhibit neovascularization [1]. Yet, antiangiogenic drugs in clinical trials have not fully lived up to their expectations [2, 3]. The limited ecacy of antiangiogenic therapies may find its origin in heterogeneity of the molecular activation status of endothelial cells in the tumor vasculature [4, 5]. More than ten years ago, Bergers et al. demonstrated that the ecacy of antiangiogenic drugs in a multistage carcinogenesis model depends on the tumor growth stage being targeted [6]. Additional evidence for dierential susceptibility of tumor vasculature to antiangiogenic treatment was given by Wood et al. [7]. They showed that inhibition of vascular endothelial growth factor- (VEGF-) receptor 2 caused a reduced occur- rence of microvessels in the interior of the tumor, while larger, more mature vessels remained unaected [7]. We hypothesize that the tumor growth stage-specific responses to antiangiogenic drugs are related to the exis- tence of dierences in angiogenic status of the tumor microvasculature in the dierent stages of tumor growth. As tumor endothelial behavior results from the local pres- ence of growth factors, cytokines, and other molecules, which varies during tumor development [810], the dier- ences in angiogenic status would be reflected in variations

TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

International Scholarly Research NetworkISRN OncologyVolume 2011, Article ID 409308, 14 pagesdoi:10.5402/2011/409308

Research Article

Tumor Vascular Morphology Undergoes DramaticChanges during Outgrowth of B16 Melanoma WhileProangiogenic Gene Expression Remains Unchanged

Elise Langenkamp,1, 2 Franziska M. vom Hagen,3 Peter J. Zwiers,1 Henk E. Moorlag,1

Jan P. Schouten,4 Hans-Peter Hammes,3 Annette S. H. Gouw,5 and Grietje Molema1

1 Medical Biology Section, Department of Pathology and Medical Biology, University Medical Center Groningen,University of Groningen, 9713 GZ Groningen, The Netherlands

2 Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden3 Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany4 Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands5 Department of Pathology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands

Correspondence should be addressed to Grietje Molema, [email protected]

Received 3 August 2011; Accepted 7 September 2011

Academic Editors: S. Mohanam and H. Rizos

Copyright © 2011 Elise Langenkamp et al. This is an open access article distributed under the Creative Commons AttributionLicense, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properlycited.

In established tumors, angiogenic endothelial cells (ECs) coexist next to “quiescent” EC in matured vessels. We hypothesized thatangio-gene expression of B16.F10 melanoma would differ depending on the growth stage. Unraveling the spatiotemporal naturethereof is essential for drug regimen design aimed to affect multiple neovascularization stages. We determined the angiogenicphenotype—represented by 52 angio-genes—and vascular morphology of small, intermediate, and large s.c. growing mouseB16.F10 tumors and demonstrated that expression of these genes did not differ between the different growth stages. Yet vascularmorphology changed dramatically from small vessels without lumen in small to larger vessels with increased lumen size inintermediate/large tumors. Separate analysis of these vascular morphologies revealed a significant difference in αSMA expressionin relation to vessel morphology, while no relation with VEGF, HIF-1α, nor Dll4 expression levels was observed. We conclude thatthe tumor vasculature remains actively engaged in angiogenesis during B16.F10 melanoma outgrowth and that the major changein tumor vascular morphology does not follow molecular concepts generated in other angiogenesis models.

1. Introduction

Tumors depend on the recruitment of new blood vessels togrow beyond a certain size and to metastasize to other tissues.This process represents an attractive target for the treatmentof cancer, and several strategies have been pursued to inhibitneovascularization [1]. Yet, antiangiogenic drugs in clinicaltrials have not fully lived up to their expectations [2, 3].The limited efficacy of antiangiogenic therapies may find itsorigin in heterogeneity of the molecular activation status ofendothelial cells in the tumor vasculature [4, 5]. More thanten years ago, Bergers et al. demonstrated that the efficacyof antiangiogenic drugs in a multistage carcinogenesis modeldepends on the tumor growth stage being targeted [6].

Additional evidence for differential susceptibility of tumorvasculature to antiangiogenic treatment was given by Woodet al. [7]. They showed that inhibition of vascular endothelialgrowth factor- (VEGF-) receptor 2 caused a reduced occur-rence of microvessels in the interior of the tumor, whilelarger, more mature vessels remained unaffected [7].

We hypothesize that the tumor growth stage-specificresponses to antiangiogenic drugs are related to the exis-tence of differences in angiogenic status of the tumormicrovasculature in the different stages of tumor growth.As tumor endothelial behavior results from the local pres-ence of growth factors, cytokines, and other molecules,which varies during tumor development [8–10], the differ-ences in angiogenic status would be reflected in variations

Page 2: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

2 ISRN Oncology

in local expression patterns of angiogenesis- and vascularmaturation-related genes during tumor outgrowth. Thebasis for rational antiangiogenic therapy regimen designthroughout the different stages of tumor growth lies in theunderstanding of the nature and location of the expressionof angiogenic genes and local balances thereof in theirpathophysiological environment.

While a vast number of studies investigated the molecularcontrol of angiogenesis using transgenic animal models thateither overexpress specific genes or have a full gene knock-out, only limited studies describe the molecular angiogenicmake up of tumors that is endorsed by endogenouslycontrolled mechanisms. In the current study we investigatedthe angiogenic phenotype of the widely used, subcutaneously(s.c.) growing mouse B16.F10 melanoma model at an early,intermediate and late stage of tumor growth. By real-timeRT-PCR, we quantified the expression of 52 genes thathave previously been reported to be major controllers oftumor angiogenesis. Based on the morphological differencesobserved, we next zoomed in on the tumor vascular com-partment by laser microdissection, to obtain a more detailedview on the localization of angio-gene expression in thecomplexity of the in vivo tumor microenvironment. Lastly,we analyzed a number of vascular features, that is, vascularmorphology, pattern of pericyte association, and prolifera-tion status of tumor and endothelial cells in the differentgrowth stages, and related them to the observed local geneexpression patterns.

2. Materials and Methods

2.1. Tumor Cell Culture and Animal Studies. B16.F10 murinemelanoma cells were cultured in 75 cm2 culture flasks inDMEM (Biowhittaker, Verviers, Belgium) supplementedwith 10% fetal calf serum (FCS; Hyclone, Perbio Science,Etten-Leur, The Netherlands), 2 mM L-glutamine (Biowhit-taker), and 1% gentamycin (Biowhittaker) at 37◦C and 5%CO2/95% air in a humidified incubator.

Male C57bl/6 mice (20–25 g; Harlan, Zeist, The Nether-lands) were subcutaneously inoculated with 100,000 B16.F10melanoma cells in 100 μL phosphate-buffered saline underanesthesia by inhalation of isoflurane/O2. The mice weremonitored daily for behavior and every other day for bodyweight. Mice were randomly divided into three experimentalgroups from which tumors were harvested at three differentstages of tumor growth: 25 ± 0.7 (early stage/small volume,day 9 after tumor inoculation), 182 ± 1.5 (intermediatestage/volume, day 12–16), and 524 ± 42.3 (late stage/largevolume, day 14–25) mm3. Tumors were measured everyother day by calipers, and the volume was calculatedaccording to the formula: tumor volume = 0.52 ∗ length ∗width2, with “width” being the shorter of the two diameters.Animals were sacrificed under anesthesia by inhalation ofisoflurane/O2, and tumors were excised, immediately snapfrozen in liquid N2, and stored at −80◦C until further ana-lysis.

The animal experiments were approved by the local Ani-mal Care and Use Committee of the University of Groningen.

2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut sections of thetumors was carried out according to the protocol of RNeasyMini Plus kit (Qiagen, Leusden, The Netherlands). RNA wasanalyzed qualitatively by gel electrophoresis and quantita-tively by Nanodrop ND-100 spectrophotometry (NanoDropTechnologies, Rockland, DE, USA) and consistently foundto be intact and protein-free. Total RNA was subsequentlyreverse transcribed as described previously [11], using Super-script III Reverse Transcriptase (Invitrogen, Carlsbad, CA,USA) in a 20 μL final volume containing 250 ng of randomhexamers (Promega, Madison, WI, USA) and 40 unitsof RNase OUT inhibitor (Invitrogen). mRNA expressionanalysis was performed in a real-time PCR-based, custom-designed, Low-Density Array set-up (Applied Biosystems,Foster City, CA, USA). The Low-Density Array was designedwith exon-overlapping primers and minor groove-binding(MGB) probes of 46 genes (see Table 1 in supplemen-tary material available online at doi:10.5402/2011/409308)selected for their involvement in angiogenesis, inflammationand basic influence on endothelial cell behavior, as reportedin the literature [12–19], including GAPDH as a house-keeping gene. The Low-Density Array card was processedaccording to the suppliers’ protocol and analyzed in an ABIPRISM 7900HT Sequence Detector (Applied Biosystems).For a selection of genes and for PCNA, the Notch familygenes, αSMA and desmin (see Supplementary Table 1), real-time PCR was performed in duplicate per sample with1 μL cDNA per reaction in TaqMan PCR MasterMix ina total volume of 10 μL, with primer-probe sets (whereappropriate corresponding to the ones pre-spotted on theLow-Density Array cards) being purchased as Assay-on-Demand from Applied Biosystems (Nieuwekerk a/d IJssel,The Netherlands).

2.3. Laser Microdissection of B16 Tumor Vasculature. Five μmcryosections mounted on polyethylene-naphtalene mem-branes attached to normal glass slides (P.A.L.M. MicrolaserTechnology AG, Bernried, Germany) were fixed in acetoneand stained with Mayer’s haematoxylin, washed with diethylpyrocarbonate-treated water, and air-dried. Tumor vascularsegments (1-2 × 106 μm2 surface area, including vessellumen area) of both intermediate and large tumors andnormal mouse kidney postcapillary venule segments (0.6× 106 μm2) were microdissected using the Laser RobotMicrobeam System (P.A.L.M. Microlaser Technology), yield-ing isolated vascular tissue from vessels with a clearly visiblelumen. To isolate vascular segments from small profilesthat do not contain a visible lumen and are therefore notidentifiable based on haematoxylin staining, we appliedfluorescence guidance during laser dissection. Tumor vascu-lar segments representing small vascular profiles and largelumen-containing vessels were separately microdissectedusing an LMD6000 Laser Microdissection system (Leica,Wetzlar, Germany) from 9 μm cryosections of large tumorsmounted on polyethylene-terephthalate membranes on steelframes (Leica) that were stained with FITC-conjugatedGriffonia Simplicifolia Lectin I IsolectinB4 (GSLI-isolectinB4: Vector Labs, Burlingame, CA, USA). The two vascular

Page 3: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

ISRN Oncology 3

morphologies were distinguished based on the combinationof size of the vessel (small versus large) and the absencerespectively presence of a visible lumen.

Total RNA was extracted according to the protocol ofAbsolutely RNA Microprep kit (Stratagene, Amsterdam,The Netherlands) or RNeasy Micro Kit (Qiagen) for hae-matoxylin-stained lumen-containing vessels and fluorescent-stained vascular profiles/segments respectively, and reversetranscribed and analyzed by Low-Density Array and/orquantitative PCR as described above.

2.4. Immunohistochemical and Immunofluorescent Staining ofTumor Tissues. Immunohistochemical detection of antigens(CD31, ICAM-1, VCAM-1, Tie2, and vWF) in acetone-fixed,5 μm cryostat-cut sections was performed using the DakoEnvision System-HRP kit (Dako Cytomation, Glostrup,Denmark) according to the suppliers’ protocol. In brief,after rehydration, sections were incubated with endogenousperoxidase block diluted 1 : 1 with PBS for 5 minutes, washedwith PBS and subsequently incubated with primary antibodydiluted in 5% FCS in PBS for 60 minutes. The follow-ing primary antibodies were used: Rat-anti-Mouse CD31IgG2a (BD Pharmingen; clone MEC13.3), Rat-anti-ICAM-1IgG2b (YN1/1.7.4 hybridoma supernatant), Rat-anti-MouseVCAM-1 IgG2a (BD Pharmingen; clone 429), Rat-anti-Tie2(eBioscience; clone Tek4), and Rabbit-anti-vWF (Dako). Incase of rat primary antibodies, sections were incubated withunlabeled Rabbit-anti-Rat IgG diluted 1 : 500 (H+L, VectorLabs) for 45 minutes. After incubation with anti-rabbitHRP-conjugated polymer for 30 minutes, peroxidase activitywas detected with 3-amino-9-ethylcarbazole (AEC) complexand sections were counterstained with Mayer’s haematoxylin(Merck, Darmstadt, Germany).

Expression of the nuclear proliferation marker Ki67was assessed by immunohistochemical staining of acetone-fixed cryosections. Sections were incubated with rab-bit monoclonal antibody to Ki67 (Lab Vision, Duiven,The Netherlands) in 5% FCS/PBS for 60 minutes, afterAvidin/Biotin blocking (Dako). Sections were incubatedwith biotin-conjugated Goat-anti-Rabbit immunoglobulins(Dako) and subsequently incubated with Streptavidin-HRP(Dako) according to the manufacturers’ protocol. Peroxidaseactivity was detected with AEC (Sigma-Aldrich; Steinheim,Germany) and sections were counterstained with Mayer’shaematoxylin. Sections were embedded in Kaisers Glycerin(Merck) and examined using a Leica DMLB microscope andLeica Qwin V3 software.

The presence of pericytes was visualized by immunofluo-rescent double staining for α-Smooth Muscle Actin (αSMA)or desmin with CD31. Five μm cryosections were fixedwith acetone/methanol (1 : 1) on ice for 5 minutes andpreincubated with PBS supplemented with 1% BSA, 10%normal goat serum (NGS; Dako), and 0.20% Tween-20 for30 minutes at room temperature. Subsequently, sections wereincubated with Cy3-conjugated mouse-anti-αSMA (Sigma)or Rabbit-anti-desmin (Abcam, Cambridge, UK) in thepresence of Rat-anti-Mouse CD31 for 60 minutes at roomtemperature. All antibody incubations were performed inPBS supplemented with 1% BSA, 10% NGS, and 0.20%

Tween-20. Sections were washed with PBS and subsequentlyincubated with Alexa fluor 568-conjugated Goat-anti-RabbitIgG to detect desmin and Alexa fluor 488-conjugated Goat-anti-Rat IgG to detect CD31 (both H+L, Molecular ProbesInvitrogen Detection Technologies, Eugene, Oregon). Afterwashing with PBS, nuclear counterstaining was performedusing DAPI (F. Hoffmann-La Roche Ltd, Basel, Switzerland).Autofluorescence was reduced by incubating the sections in0.1% Sudan Black (Sigma-Aldrich) in 70% ethanol for 30minutes at room temperature. Sections were embedded inCitifluor (Citifluor Ltd., London, UK) and examined using afluorescence microscope (DM RXA, Leica) and Leica QwinV3 software.

2.5. Statistics. Linear Mixed Effects (LMEs) Models andAnalysis to account for multiple measurements within micewere used for data analysis to address the significance ofobserved effects [20]. Multiple testing was controlled by thestep-up False Discovery Rate-controlling procedure of Ben-jamini and Hochberg [21] in calculating adjusted P values(Padj < 0.05 indicated statistical significance) for the differentresearch questions. All analyses were performed using R [22].

Statistically significant differences in gene expressionin endothelium of the two different vascular morpholo-gies observed were assessed by means of Student’s t-test(GraphPad Prism, GraphPad Software, San Diego, CA) andconsidered to be significant when P < 0.05.

3. Results

3.1. Different B16.F10 Growth Stages Presented with a Dif-ferent Vascular Morphology, but with a Similar Gene Ex-pression Profile. We hypothesized that early-stage tumorgrowth would be characterized predominantly by angiogenicsprouting vasculature, while in late-stage tumors active an-giogenic vascular sprouts would exist next to “quiescent”matured neovessels. This would represent one mode of vas-cular heterogeneity that may affect efficacy of drug treat-ment. Therefore, we first investigated neovessel developmentduring tumor growth from a morphological point of viewand at a molecular level in s.c. growing B16 tumors, whichwere harvested at an early (palpable), intermediate, and latestage of tumor development (Figure 1).

Vascular morphology changed drastically throughouttumor development. While palpable tumors were charac-terized by small vessels, generally containing no or a smalllumen, the vascular network of intermediate and largetumors consisted of vessels with wide lumen that existednext to small vascular structures without lumen. The vesselswith the largest lumen surface were found in the largetumors (Figures 1(a)–1(d)). Strikingly, these morphologicaldifferences were not accompanied by a major differencein angio-gene expression profile (Figure 2), except for theexpression of integrin β3, which was statistically significantlyhigher in small tumors versus intermediate and large tumors(P < 0.05). Expression of molecules from the VEGF-familyand their receptors, regarded as key regulators of the firststep of the angiogenic cascade, was equal in the three stagesof tumor outgrowth. The ratio of Ang2/Ang1 mRNA levels

Page 4: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

4 ISRN Oncology

(a) B16 palpable (b) B16 intermediate

(c) B16 large (center) (d) B16 large (periphery)

0

100

200

300

400

500

600

Tum

orvo

lum

e(m

m3)

Palpable Intermediate Large

(e) B16 growth stages

Figure 1: Morphological appearance of tumor vasculature of B16.F10 melanoma growing subcutaneously: immunohistochemical detectionof CD31. Tumors were harvested at three different growth stages, based on tumor volumes as depicted in (e) (mean of 3 mice per group ±SEM). Vascular morphology differed per tumor growth stage, presenting vessels with increasing lumen size in intermediate and large tumorsnext to the presence of small vascular profiles, while small tumors were characterized by only small vessels that were generally lacking avisible lumen (Magnification 100x).

varied between 24 and 78, which is in line with the currentdogma that a dysbalance in Ang2/Ang1 in favor of Ang2 de-stabilizes the endothelium and presensitizes it for (VEGF-induced) proliferation and angiogenic sprouting [23].

Low mRNA levels of the adhesion molecules P- and E-selectin were associated with nondetectable protein levels(data not shown), while the mRNA expression levels ofVCAM-1 and ICAM-1 were higher and associated withprotein expression restricted to a subset of blood vessels(Figure 3). ICAM-1 was furthermore expressed by perivas-cular infiltrates of some vessels. Both VCAM-1 and ICAM-1 expression were more intense in palpable compared tointermediate and large tumors, a trend also observed at

the ICAM-1 mRNA level that was however statistically notsignificant. Interestingly, the mRNA level of von WillebrandFactor, which is like CD31 and VE-cadherin regarded asan endothelial marker molecule and often used as researchtool for immunofluorescence double staining protocols [24],was much lower than that of CD31, an observation thatwas confirmed at the protein level. In contrast to CD31,vWF protein expression appeared granular and covered onlyparts of the vessel wall of a small subset of vessels. AlsoTie2 expression was detected in a subset of vessels and oftendid not fully cover the endothelial lining within the vesselwall. Both vWF and Tie2 protein expression patterns did notchange during tumor growth.

Page 5: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

ISRN Oncology 5

NG

-2

Eph

B4

Eph

nB

2

vWF

VE

cad

CD

31 αv β3

0.01

0.1

1

10

mR

NA

expr

essi

onre

lati

veto

GA

PD

H(×

1000

)

(a) Vascular markers

CD

105

ALK

5

PD

GF-

B

Tie

2

An

g2

An

g1

FGF-

R2

FGF-

R1

FGF-

2

Nu

r77

VE

GF-

R2

VE

GF-

R1

PlG

F

VE

GF-

A

PD

GF-

TG

HIF

0.001

0.01

0.1

1

10

mR

NA

expr

essi

onre

lati

veto

GA

PD

H(×

1000

)

(b) Angiogenesis-associated

CD

45

ICA

M-1

VC

AM

E-s

el

P-se

l

MC

P-1

TN

F-R

2

TN

F-R

1

TN

F

eNO

S

iNO

S

PalpableIntermediateLarge

0.001

0.01

0.1

1

10

mR

NA

expr

essi

onre

lati

veto

GA

PD

H(×

1000

)

(c) Inflammation

PalpableIntermediateLarge

0.001

0.01

0.1

1

100

10

mR

NA

expr

essi

onre

lati

veto

GA

PD

H(×

1000

)

Bcl

2

PC

NA

CO

X-2

Tsp-

1

Pla

smin

ogen

t-PA

u-P

A

MM

P9

MM

P2

TIM

P-1

ND

(d) Miscellaneous

Figure 2: In the three different stages of s.c. B16.F10 tumor growth, mRNA expression of the 46 genes under study is similar. Tumorswere harvested at palpable, intermediate, and large volumes (early, intermediate and late stages). Values represent mRNA expression levelsmeasured by qRT-PCR-based Low-Density Array and adjusted to GAPDH. Mean + SD; n = 3. Expression of FGF-2, FGFR2, Ang1, P-selectin, E-selectin, and COX-2 was additionally measured twice by manual qRT-PCR. PCNA was measured by manual real-time RT-PCRalone in duplicate. ∗P < 0.05. ND: not detectable.

3.2. Gene Expression in the B16.F10 Tumor Vascular Com-partment That Represents Lumen-Containing Vessels. Wehypothesized that vascular behavior is the result of locallycontrolled balances of gene expression. As the majority of thegenes under study are not restricted to the endothelium, wezoomed in on the local balances of vascular gene expressionby isolating tumor vascular segments from their environ-ment by laser microdissection prior to gene expressionanalysis.

mRNA of microdissected tumor vasculature identifiedby light microscopy in haematoxylin-stained biopsies, hencerepresenting lumen-containing vasculature, showed a ∼20–60 fold enrichment in endothelial marker genes CD31, VE-cadherin, vWF, and EphrinB2, compared to mRNA isolatedfrom a tumor biopsy as a whole (Figure 4). Moreover, wecould assign VEGFR1 and -R2, FGFR1, Ang2, Tie2, PDGF-B,

PDGF-Rβ, eNOS, t-PA, and Tsp-1 as being tumor vascular-associated in this s.c. B16.F10 tumor model, as they exhibiteda similar 15–50-fold enrichment. In contrast, FGF-2, FGFR2,Ang1, MMP2, MMP9, plasminogen, TNFα, P-selectin, E-selectin, and COX-2 transcripts were not detectable in thiscompartment.

Of note is the observation that compartmentalization ofgenes revealed that the relative balances of gene expressionin the tumor vasculature differed from those observed whenanalyzing the tumor tissue as a whole. For example, whenanalyzing the mRNA levels in the tumors, the levels ofintegrin β3 were 2.5-fold lower than those of integrin αv,while in the tumor vasculature both integrin subunits wereexpressed at equal levels. Similarly, the ratio between the pro-apoptotic gene Tsp-1 [25] and the antiapoptotic Bcl2 shiftedfrom 1.9 in the tumor to 13.6 in the tumor vasculature, which

Page 6: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

6 ISRN Oncology

Palpable Intermediate Large

VC

AM

ICA

MvW

FT

ie2

CD

31C

D31

Figure 3: Vascular localization and expression of endothelial molecules at the protein level in B16.F10 tumors growing at three differentstages of tumor growth. Expression of VCAM-1 and ICAM-1 was highest in the early-stage tumors, as compared to intermediate and largetumors. A granular pattern of vWF expression was found in a small number of tumor vessels; the majority of them were negative for vWF.Tie2 expression remained the same throughout tumor development and was observed in only a subset of the vessels. The top three andbottom three rows of panels represent immunohistochemical staining of consecutive sections. Arrowheads point to the structures that areenlarged in the insets. Thin arrows point to vessels in the large tumor that are positive for Tie2, while the largest vessel in the same photographdid not express detectable levels of Tie2 protein. Pictures are representative examples of each experimental group (Magnification 100x).

Page 7: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

ISRN Oncology 7

CD31VEcadvWFEphnB2EphB4NG-2

Bcl2COX-2Tsp-1Plasminogent-PAu-PA

PCNA

MMP9MMP2CD45ICAM-1VCAME-selP-selMCP-1TNF-R2TNF-R1TNFeNOSiNOSCD105ALK5

PDGF-BTie2Ang2Ang1FGF-R2FGF-R1FGF-2Nur77VEGF-R2VEGF-R1PlGFVEGF-A

NDND

ND

NDNDND

NDND

NDND

ND

ND

00 55 1010 15 20 25

ND

TIMP-1

PDGF-RβTGFβ

αvβ3HIF1α

∗∗

∗∗∗

∗∗

∗∗

∗∗

∗∗

∗∗

∗∗

∗∗

∗∗

∗∗

∗∗

∗∗

Microdissected tumor vasculatureWhole tumor

mRNA expressionrelative to GAPDH (×1000)

Figure 4: Vascular compartmentalization of angio-gene expression in a large tumor. Tumor vasculature was laser microdissected and itsgene expression profile was compared to that of the tumor as a whole. Values represent mRNA levels adjusted to GAPDH. Mean values ±SD of 3 qPCR analyses, except for CD31, VE-cadherin, vWF, EphrinB2, EphB4, Integrin αv, VEGFR2, Tie2, iNOS, eNOS, TNFα, TNFR1,TNFR2 (a total of 5 qPCR analyses), and for VEGF (a total of 7 qPCR analyses) and PCNA (qPCR in quadruplicate). ∗P < 0.05. ND: notdetectable.

may point to a relatively higher degree of Tsp1-inducedproapoptotic signaling in the tumor vasculature comparedto the non-vascular tumor compartment.

3.3. Different Vascular Morphologies Are Associated with aSimilar Profile of Angiogenic Gene Expression. The vascu-lature of late-stage tumors was characterized by regionalvariations in vascular morphology. Regions with small vas-cular profiles present at a high density (Figure 5(a)) existednext to regions with large vessels containing a wide lumenthat were distributed at a much lower density (Figure 5(b)).Based on reports describing a role for HIF-1α and VEGF indetermining blood vessel morphogenesis, including lumenformation [26, 27], we hypothesized that a differential pat-tern of expression of these genes might underlie the different

vascular morphologies in this tumor model. To study this,we separately microdissected large, lumen-containing vesselsand small vascular segments without lumen, guided by flu-orescence staining with the FITC-labeled GSLI-isolectinB4(Figures 5(c) and 5(d)). Surprisingly, both vascular mor-phologies were characterized by an identical profile of mRNAexpression of those genes that were previously assigned asmajor controllers of vascular morphology and structure(Figure 5(e)). Also mRNA levels of members of the Notchfamily of genes and their ligands, which were recentlyidentified as being in control of angiogenic sprout formation(Notch1 and Dll4 [28]) on the one hand, and endothelial-pericyte adhesion (Notch3 and Jag1 [29]) on the other hand,did not differ in the two morphologically different vascularsegments (Figure 5(f)).

Page 8: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

8 ISRN Oncology

Small vascular profiles

(a)

Wide lumen-containing vessels

(b)

50μm

110

144143

142

141

(c)

80

50μm

(d)

CD

31

VE

cad

HIF

VE

GF-

A

PIG

F

VE

GF-

R1

VE

GF-

R2

An

g1

An

g2

Tie

2

PD

GF-

B

TG

ALK

5

CD

105m

RN

Aex

pres

sion

rela

tive

toG

AP

DH

(×10

00)

Small/no lumenWide open lumen

0.1

1

10

100

ND

Nu

r77

(e)

Not

ch1

Not

ch3

DII

4

Jag1

Small/no lumenWide open lumen

0.1

1

10

100

mR

NA

expr

essi

onre

lati

veto

GA

PD

H(×

1000

)

(f)

PD

GF-

Des

min

NG

-2

Small/no lumenWide open lumen

αSM

A

0.1

1

10

100

mR

NA

expr

essi

onre

lati

veto

GA

PD

H(×

1000

)

(g)

Figure 5: Different vascular morphologies displayed a similar pattern of gene expression. High variability of vascular morphology wasobserved in tumors of large volume. Large lumen-containing vessels ((b, d) vessel lumen size > 25 μm) existed next to much smaller bloodvessels that did not contain a visible lumen (a, c). (a, b) Immunohistochemical staining for CD31: pictures represent different regions withinthe same large tumor (Magnification 100x). (c, d) Cryosections from large tumors were stained with GSL-I IsolectinB4:FITC and the differentvascular morphologies were separately isolated by laser microdissection along the depicted lines, magnification 400x. Microdissection ofthese different vascular phenotypes followed by gene expression profiling did not reveal a differential pattern of mRNA expression ofangiogenesis- and vascular stability-regulating genes (e), nor of the Notch family of genes that has recently been identified to be importantregulators of both angiogenic sprouting and endothelial-pericyte adhesion (f), but demonstrated that the lumen-containing vessels wereassociated with significantly higher levels of αSMA mRNA compared to the small vessels without lumen (g). (e)–(g) Mean values + SD ofduplicate qPCR measurements of three large tumors. ND: not detectable. ∗P < 0.05.

Page 9: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

ISRN Oncology 9

3.4. Different Vascular Morphologies Were Associated withDifferent Pericyte Phenotypes. As pericyte coverage can affectvascular morphology, we next set out to investigate pericytemarker expression in these two segments. Of the threemarkers studied (αSMA, desmin, and NG-2), mRNA levelsof αSMA were found to be more than 3-fold higher inlarge lumen-containing vessels compared to small vascularprofiles, while the other two genes did not significantly differ(Figure 5(g)). To validate this observation we determinedthe extent and pattern of pericyte coverage of these twovascular profiles by immunofluorescent double staining forCD31 and either αSMA or desmin as pericyte markers.This demonstrated that lumen-containing B16 tumor vesselswere associated with pericytes that stained positive for bothdesmin and αSMA, while the majority of small vessels wereassociated with support cells that expressed only desmin.These protein data corroborated the difference in mRNAexpression in the two morphologically different vascular bedswithin the same tumor (Figure 6).

Absence of αSMA was previously shown to be a featureof a microvasculature that is actively engaging in angiogen-esis [30, 31]. This implies that αSMA-lacking vessels aremost likely to be found in areas where angiogenesis is aprerequisite for growth, that is, in areas where tumor cellsare highly proliferative. To address whether such a processindeed underlies the observed differences, we assessed theproliferative activity of endothelial cells of both types ofvascular morphologies, and of their surrounding tumorcells. While the majority of tumor cells were proliferativeactive, Ki67 expression was only sporadically detected in en-dothelial cells of both the small vascular profiles and thelumen-containing vessels. Ki67 expression by tumor cells,furthermore, appeared homogeneously distributed through-out the tumor, irrespective of the stage of tumor growth, andcould not be related to the observed differences in vascularmorphology nor vascular maturity. Similarly, PCNA mRNAlevels did not differ in small vascular profiles compared tolumen-containing large vessels (data not shown).

3.5. The Gene Expression Profiles of the Tumor VasculaturesRepresent an Active, Angiogenic Phenotype: Comparison withQuiescent Endothelium. To molecularly describe the dif-ference in phenotype between B16.F10 tumor vasculaturethat actively engages in angiogenesis, and quiescent, non-angiogenic vasculature, we profiled the expression of theangio-genes in normal postcapillary venule endotheliummicrodissected from a healthy mouse kidney (Figure 7).This analysis revealed that integrin β3, PlGF, Nur77, TIMP-1, and t-PA were absent in the majority of normal healthykidney endothelium samples, while being present in thetumor endothelium. Likewise, VEGFR1, Ang2, TGFβ, eNOS,and MCP-1 were more frequently detected in the tumorendothelium as compared to normal kidney endothelium.On the opposite, EphB4, FGFR1, Tie2, Alk5, TNFR2, andthe adhesion molecules VCAM-1 and ICAM-1 were morefrequently detected and at higher levels in normal kidneyendothelium as compared to tumor endothelium from in-termediate and large tumors.

4. Discussion

In this study we investigated the angiogenic make-up ofs.c. growing B16.F10 tumors at different stages of tumorgrowth. We furthermore mapped the molecular angiogenicmake-up of the tumor vascular compartment and relatedthat to the morphology of the vasculature. Our focus layon genes that were previously shown to be responsible forthe induction of angiogenic sprouting and vascular matu-ration. We showed that, although the vascular morphologychanged dramatically during tumor outgrowth, this was notassociated with major differences in expression of the 46genes analyzed. Moreover, morphologically highly differenttumor vessels exhibited similar expression profiles of themajor angio-genes studied. The expression of αSMA was adiscriminative parameter for large vessels; its high mRNAlevels and clear protein expression in large lumen-containingvessels contrasted with the much lower mRNA levels and theabsence of protein in small vascular profiles without a lumen.This difference in vascular support cell/pericyte characteris-tics may underlie the difference in vascular morphology inthis tumor model; yet the angio-genes accompanying thisphenotype remain until now unknown. We demonstratedthat the 46 genes chosen are associated with the angiogenicprocess by comparing their expression in the tumor vascularcompartment with that in a mature vessel of a healthy organ,which showed that many proangiogenic genes indeed exhib-ited a gain of function in the tumor vasculature. From thisstudy we conclude that the vasculature of s.c. B16.F10 tumorscontinues to actively engage in angiogenesis throughout thedifferent stages of tumor growth. The striking differencesin vascular morphology between small and large tumorsare associated with a difference in pericyte activation buthave no direct relation to the transcriptional activity ofthe angio-genes analyzed in this study. A similar pattern ofproangiogenic gene expression continues to exist in evenlater B16.F10 growth stages (tumor volumes ranging from461–1409 mm3; data not shown).

Our aim was to investigate how in a growing tumorof a widely employed mouse model the expression ofgenes important in the control of the different stages ofangiogenesis and vascular maturation would spatiotempo-rally change with ongoing tumor growth. Surprisingly, ourfindings demonstrated that the B16.F10 tumor does notchange its molecular angiogenic repertoire during tumordevelopment. This outcome contrasts findings in a rat C6glioma model growing in the brain [8] as well as in murineorthotopic KM12SM colon carcinoma growth [9] in whichVEGF expression varied during tumor growth progression.Furthermore, expression of Ang2 has been shown to beelevated during the early stages of tumor outgrowth in a U-87 MG glioma model growing intracranially in athymic mice,while being downmodulated once the vascular structuresof the tumor had established, as in the later stages oftumor outgrowth [32]. The here described elevated levels ofthe VEGF-receptors, Nur77, Ang2, and Tie2 in the tumorvascular compartment compared to the tumor compart-ment [16, 23], as well as the significantly higher level ofPDGF-B, a molecule extensively produced by angiogenic

Page 10: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

10 ISRN Oncology

CD31 Desmin Merge

Larg

elu

men

No/

smal

llu

men

(a)

CD31 MergeαSMA

Larg

elu

men

No/

smal

llu

men

(b)

Ki67Palpable Intermediate Large

(c)

Figure 6: Vessels with different morphological appearance are covered with pericytes presenting with a different pattern of pericyte markerexpression. (a, b) Immunofluorescent double staining for CD31 (green) and desmin (red) or α-SMA (red) and nuclear counter stain(DAPI; blue) of a representative tumor from the group of late-stage, large volume tumors (Magnification 200x). (c) Immunohistochemicaldetection of Ki67 revealed homogeneous distribution of proliferative activity throughout the whole tumor, in all three stages of tumor growth(Magnification 200x). Arrows point to Ki67-positive endothelial nuclei.

tip cells on a sprouting tip [33], suggest a VEGF/Ang2-driven proangiogenic phenotype of the B16.F10 modelemployed. Moreover, as compared to normal endothelium,tumor endothelium showed gain of function of numerous

proangiogenic molecules such as integrin β3, PlGF, VEGFR1,Nur77, Ang2, eNOS, TGFβ, MCP-1, TIMP-1, and t-PA,while loss of function was observed for vessel stability genessuch as Ang1 and Tie2. From this, it can be concluded

Page 11: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

ISRN Oncology 11

Bcl2COX-2Tsp-1Plasminogent-PAu-PATIMP-1MMP9MMP2CD45ICAM-1VCAME-selP-selMCP-1TNF-R2TNF-R1TNFeNOSiNOSCD105ALK5

PDGF-BTie2Ang2Ang1FGF-R2FGF-R1FGF-2Nur77VEGF-R2VEGF-R1PlGFVEGF-A

NG-2EphB4EphrinB2vWFVEcadCD31

Healthy organvenule EC

Tumor EC (B16 s.c.)

PDGF-

-

TGFβ

αvβ3

HIF 1α

Figure 7: Summary of gain and loss of gene expression inthe B16.F10 tumor vascular compartment of intermediate andlarge tumors compared to normal quiescent endothelium: geneexpression in microdissected tumor endothelium from s.c. growingintermediate and large B16.F10 tumors (a total of 6 animals) andmicrodissected kidney venule endothelium from healthy C57bl/6mice (10 animals). Red: detectable expression in more than 75%of all analyzed samples; yellow: detectable expression in 25–75%of all analyzed samples; green: expression of the gene could not bedetected in the majority of the samples (less than 25% of positivity).

that throughout the growth period studied, the majority ofvessels of B16.F10 tumors remain engaged in the angiogenicprocess. Interestingly, the same pattern of proangiogenic

gene expression was found when B16.F10 tumors weregrown in the highly vascularized brain (SupplementaryFigure 1), demonstrating that neither the host environmentwhere the tumor is growing changes the proangiogenicmolecular repertoire of the tumor vasculature.

Yet, during s.c. B16.F10 outgrowth, the vascular mor-phology underwent a major shift from small, lumen-lessvessels mainly covered by desmin-positive pericytes tolarge lumen-containing vessels mainly covered by pericytespositive for both αSMA and desmin. Recently, Helfrich etal. reported that spontaneously arising MT/ret transgenicmelanomas were characterized by a similar distribution ofαSMA- and desmin-positive vessels in relation to vascularlumen size and density and found that αSMA-coveredlarge vessels were less sensitive to anti-VEGF therapy [31],thereby illustrating the consequences of tumor vascularheterogeneity for the outcome of antiangiogenic therapy. Asimilar heterogeneity in vascular morphology as observedin our study was previously reported by Kashiwagi et al.in B16.F10 tumors grown in cranial windows. In theirstudy, changes in vascular morphology could not be relatedto transcriptional regulation of 96 angiogenesis-associatedgenes, including the Angiopoietins and Tie2, VEGF andits receptor, and PDGF [34]. Vascular morphology haspreviously been shown to be under the control of thesegenes, with low VEGF levels resulting in a reduction ofblood vessel lumen size in human gastric TMK-1 tumorsin athymic nude mice [26] and in the chick CAM assay[27]. In the B16.F10 tumor model in immune-competentmice employed here, vascular morphology did however notrelate to HIF-1α and VEGF mRNA levels. The expressionlevels of the VEGFR2 response gene Nur77 in the threegrowth stages as well as in the two different tumor vesselmorphologies furthermore corroborate the conclusion thatsimilar levels of VEGF-activity can exist while vasculaturesare morphologically highly variable. Another set of genespreviously shown to affect vascular morphology includesNotch and Notch ligand Dll4 [28, 35]. Enhanced Dll4expression in xenograft models was associated with increasedvessel size [36], but in our model, these genes were notdiscriminative between small vascular profiles and lumen-containing vessels. From these data it becomes clear thateach preclinical angiogenesis model needs to be appreciatedfor its own molecular repertoire underlying angiogenesisand vascular remodeling and that care should be taken inextrapolating observations from one model to the other.

Although we did not achieve the broader aim of thestudy, that is, identification of the molecular nature of tumorvascular heterogeneity during tumor outgrowth, a numberof issues were revealed that are of relevance when studyingangiogenesis. One important finding is that vWF expressionshowed a remarkable heterogeneity throughout the tumorvasculature. It was not expressed in a pan-endothelial man-ner as were CD31 and VE-cadherin. A similar loss of vWFexpression during angiogenesis was reported previously byus in a mouse model of inflammation [37], as well as by Yanoet al. in biopsies from Non-Small Cell Lung Cancer patients[38]. The use of vWF as a marker for tumor endothelium,as is employed in, for example, fluorescent double-labeling

Page 12: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

12 ISRN Oncology

strategies [24], hence can have important drawbacks. More-over, as endothelial cells are numerically underrepresentedin a tumor, their molecular profile will be masked in wholetumor mRNA isolates by that of the dominating tumorcells. By applying laser microdissection-assisted isolationof the tumor vasculature prior to transcriptional profiling,we were able to reveal local balances in gene expressionthat are controlled by the pathophysiological environmentwhich would have remained masked when analyzing wholetumor RNA samples. Although microdissection of tumorendothelium does not yield a pure endothelial isolate asvisualized by detection of mRNA impurities such as pericyte-associated genes NG-2, αSMA, and desmin, judging fromthe small enrichment of these molecules relative to that ofCD31 and VE-cadherin, their contribution to the populationof dissected cells is minor.

Our results indicate that B16.F10 melanoma vasculardevelopment is accompanied by major changes in the vas-cular morphology. This observation has clinical relevance,as variations in vascular architecture have been described inbiopsies from prostate carcinoma patients with metastasis inbone, liver, and lymph node [39]. Human clear-cell renalcell carcinoma is characterized by heterogeneous tumorvascular behavior, including variations in microvasculardensity, endothelial cell proliferation, and expression ofangiogenic growth factors [40]. Together, these examplesemphasize the clinical relevance of the occurrence of het-erogeneity of tumor vascularization and the need for in-depth studies on the molecular nature thereof. Ideally, thetechnique of sampling tumor vascular segments from thepathophysiological environment would allow to microdissectimmunohistochemically identified subsets of pericytes orendothelial cells that are differentially activated as observedby, for example, gain and loss of VEGFR2 and αvβ3. If sucha technique could be combined with genome-wide tran-scriptional profiling, unraveling the true molecular natureof morphological changes and tumor vascular heterogeneitywould come within reach. Immunofluorescence stainingwith the lectin as employed here is a first step, as it providesfor the first time the opportunity to dissect endothelial cellsthat are difficult to visualize, while so far in our handsantibody-based protocols did not allow for proper visual-ization with maintenance of RNA integrity. Combinationof laser microdissection with kinome and protein arrays todetect phosphorylation status of many kinases involved inangiogenic signaling, for example, VEGFR2, ERK, and Akt,would furthermore broaden the options to get a clearer viewon the molecular basis of tumor vascular heterogeneity [41];yet at present this is unfeasible due to the limited amount oftissue yielded by laser microdissection.

Future studies should focus on an extension of the mRNAexpression profiles in the tumor vascular microenvironment,on regulation and (posttranslational) modification thereof atthe protein level, and on determination of the consequenceof antiangiogenic drug treatment on these parameters. Inte-grating this knowledge will ultimately contribute to theestablishment of effective antiangiogenic therapy in the clinicand relevant biomarkers of efficacy.

Abbreviations

Ang1/2: Angiopoietin-1/2Alk5: Activin receptor-like kinase-5Bcl2: B-cell lymphoma protein-2COX-2: Cyclooxygenase-2eNOS: endothelial Nitrogen Oxide SynthaseFGF(-R1/2): Fibroblast Growth Factor (Receptor

1/2)HIF-1α: Hypoxia inducible factor-1αICAM-1: Intercellular adhesion molecule-1iNOS: inducible Nitrogen Oxide SynthaseMCP-1: Monocyte chemoattractant protein-1MMP2/9: Matrix metalloproteinases 2/9NG-2: Neuron glial-2PCNA: Proliferating cell nuclear antigenPDGF(-Rβ): Platelet-Derived Growth Factor

(Receptor-β)PlGF: Placental Growth FactorαSMA: α-Smooth Muscle ActinTGFβ: Transforming Growth Factor-βTIMP-1: Tissue inhibitor of metalloproteinase-1TNF(-R1/2): Tumor Necrosis Factor (Receptor 1/2)t-PA: tissue-type Plasminogen ActivatorTsp-1: Thrombospondin-1u-PA: urokinase-type Plasminogen ActivatorVCAM-1: Vascular cell adhesion molecule-1VEGF(-R1/2): Vascular Endothelial Growth Factor

(Receptor 1/2)vWF: von Willebrand Factor.

Conflict of Interests

The authors declare that they have no conflict of interests.

References

[1] J. Folkman, “Angiogenesis: an organizing principle for drugdiscovery?” Nature Reviews Drug Discovery, vol. 6, no. 4, pp.273–286, 2007.

[2] R. K. Jain, D. G. Duda, J. W. Clark, and J. S. Loeffler, “Lessonsfrom phase III clinical trials on anti-VEGF therapy for cancer,”Nature Clinical Practice Oncology, vol. 3, no. 1, pp. 24–40,2006.

[3] D. G. Duda, T. T. Batchelor, C. G. Willett, and R. K. Jain,“VEGF-targeted cancer therapy strategies: current progress,hurdles and future prospects,” Trends in Molecular Medicine,vol. 13, no. 6, pp. 223–230, 2007.

[4] E. Langenkamp and G. Molema, “Microvascular endothelialcell heterogeneity: general concepts and pharmacologicalconsequences for anti-angiogenic therapy of cancer,” Cell andTissue Research, vol. 335, no. 1, pp. 205–222, 2009.

[5] W. C. Aird, “Molecular heterogeneity of tumor endothelium,”Cell and Tissue Research, vol. 335, no. 1, pp. 271–281, 2009.

[6] G. Bergers, K. Javaherian, K. M. Lo, J. Folkman, and D.Hanahan, “Effects of angiogenesis inhibitors on multistagecarcinogenesis in mice,” Science, vol. 284, no. 5415, pp. 808–812, 1999.

Page 13: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

ISRN Oncology 13

[7] J. M. Wood, G. Bold, E. Buchdunger et al., “PTK787/ZK222584, a novel and potent inhibitor of vascular endothelialgrowth factor receptor tyrosine kinases, impairs vascular en-dothelial growth factor-induced responses and tumor growthafter oral administration,” Cancer Research, vol. 60, no. 8, pp.2178–2189, 2000.

[8] J. Holash, P. C. Maisonpierre, D. Compton et al., “Vesselcooption, regression, and growth in tumors mediated byangiopoietins and VEGF,” Science, vol. 284, no. 5422, pp.1994–1998, 1999.

[9] R. Kumar, H. Kuniyasu, C. D. Bucana, M. R. Wilson, andI. J. Fidler, “Spatial and temporal expression of angiogenicmolecules during tumor growth and progression,” OncologyResearch, vol. 10, no. 6, pp. 301–311, 1998.

[10] I. J. Fidler, “Angiogenic heterogeneity: regulation of neoplasticangiogenesis by the organ microenvironment,” Journal of theNational Cancer Institute, vol. 93, no. 14, pp. 1040–1041, 2001.

[11] J. M. Kułdo, J. Westra, S. A. Asgeirsdottir et al., “Differentialeffects of NF-κB and p38 MAPK inhibitors and combinationsthereof on TNF-α- and IL-1β-induced proinflammatory statusof endothelial cells in vitro,” American Journal of Physiology,vol. 289, no. 5, pp. C1229–C1239, 2005.

[12] A. W. Griffioen and G. Molema, “Angiogenesis: potentials forpharmacologic intervention in the treatment of cancer, cardio-vascular diseases, and chronic inflammation,” PharmacologicalReviews, vol. 52, no. 2, pp. 237–268, 2000.

[13] E. M. Conway, D. Collen, and P. Carmeliet, “Molecularmechanisms of blood vessel growth,” Cardiovascular Research,vol. 49, no. 3, pp. 507–521, 2001.

[14] R. K. Jain, “Molecular regulation of vessel maturation,” NatureMedicine, vol. 9, no. 6, pp. 685–693, 2003.

[15] P. Carmeliet, “Mechanisms of angiogenesis and arteriogene-sis,” Nature Medicine, vol. 6, no. 4, pp. 389–395, 2000.

[16] H. Zeng, L. Qin, D. Zhao et al., “Orphan nuclear receptorTR3/Nur77 regulates VEGF-A-induced angiogenesis throughits transcriptional activity,” Journal of Experimental Medicine,vol. 203, no. 3, pp. 719–729, 2006.

[17] G. D. Yancopoulos, S. Davis, N. W. Gale, J. S. Rudge, S. J.Wiegand, and J. Holash, “Vascular-specific growth factors andblood vessel formation,” Nature, vol. 407, no. 6801, pp. 242–248, 2000.

[18] P. Auguste, S. Lemiere, F. Larrieu-Lahargue, and A. Bikfalvi,“Molecular mechanisms of tumor vascularization,” CriticalReviews in Oncology/Hematology, vol. 54, no. 1, pp. 53–61,2005.

[19] C. F. Chantrain, P. Henriet, S. Jodele et al., “Mechanisms ofpericyte recruitment in tumour angiogenesis: a new role formetalloproteinases,” European Journal of Cancer, vol. 42, no. 3,pp. 310–318, 2006.

[20] J. Pinheiro, D. Bates, S. DebRoy, and D.a.t.R.C.t. Sarkar, “nlme:Linear and Nonlinear Mixed Effects Models,” R packageversion 3.1-892008.

[21] Y. Benjamini and Y. Hochberg, “Controlling the false discoveryrate: a practical and powerful approach to multiple testing,”Journal of the Royal Statistical Society B, vol. 57, pp. 289–300,1995.

[22] R Development Core Team, R: A Language and Environmentfor Statistical Computing, R Foundation for Statistical Com-puting, Vienna, Austria, 2008.

[23] M. Thomas and H. G. Augustin, “The role of the angiopoietinsin vascular morphogenesis,” Angiogenesis, vol. 12, no. 2, pp.125–137, 2009.

[24] J. Xu, D. Rodriguez, E. Petitclerc et al., “Proteolytic exposureof a cryptic site within collagen type IV is required forangiogenesis and tumor growth in vivo,” Journal of CellBiology, vol. 154, no. 5, pp. 1069–1079, 2001.

[25] J. E. Nor, R. S. Mitra, M. M. Sutorik, D. J. Mooney, V. P. Castle,and P. J. Polverini, “Thrombospondin-1 induces endothelialcell apoptosis and inhibits angiogenesis by activating thecaspase death pathway,” Journal of Vascular Research, vol. 37,no. 3, pp. 209–218, 2000.

[26] O. Stoeltzing, M. F. McCarty, J. S. Wey et al., “Role ofhypoxia-inducible factor 1α in gastric cancer cell growth,angiogenesis, and vessel maturation,” Journal of the NationalCancer Institute, vol. 96, no. 12, pp. 946–956, 2004.

[27] P. Parsons-Wingerter, U. M. Chandrasekharan, T. L. McKay etal., “A VEGF165-induced phenotypic switch from increasedvessel density to increased vessel diameter and increasedendothelial NOS activity,” Microvascular Research, vol. 72, no.3, pp. 91–100, 2006.

[28] M. Hellstrom, L. K. Phng, J. J. Hofmann et al., “Dll4 signallingthrough Notch1 regulates formation of tip cells duringangiogenesis,” Nature, vol. 445, no. 7129, pp. 776–780, 2007.

[29] H. Liu, S. Kennard, and B. Lilly, “NOTCH3 expressionis induced in mural cells through an autoregulatory loopthat requires Endothelial-expressed JAGGED1,” CirculationResearch, vol. 104, no. 4, pp. 466–475, 2009.

[30] L. E. Benjamin, I. Hemo, and E. Keshet, “A plasticity windowfor blood vessel remodelling is defined by pericyte coverage ofthe preformed endothelial network and is regulated by PDGF-B and VEGF,” Development, vol. 125, no. 9, pp. 1591–1598,1998.

[31] I. Helfrich, I. Scheffrahn, S. Bartling et al., “Resistance toantiangiogenic therapy is directed by vascular phenotype,vessel stabilization, and maturation in malignant melanoma,”Journal of Experimental Medicine, vol. 207, no. 3, pp. 491–503,2010.

[32] O. H. Lee, J. Fueyo, J. Xu et al., “Sustained angiopoietin-2expression disrupts vessel formation and inhibits gliomagrowth,” Neoplasia, vol. 8, no. 5, pp. 419–428, 2006.

[33] H. Gerhardt, M. Golding, M. Fruttiger et al., “VEGF guidesangiogenic sprouting utilizing endothelial tip cell filopodia,”Journal of Cell Biology, vol. 161, no. 6, pp. 1163–1177, 2003.

[34] S. Kashiwagi, Y. Izumi, T. Gohongi et al., “NO mediatesmural cell recruitment and vessel morphogenesis in murinemelanomas and tissue-engineered blood vessels,” Journal ofClinical Investigation, vol. 115, no. 7, pp. 1816–1827, 2005.

[35] G. Thurston and J. Kitajewski, “VEGF and Delta-Notch:Interacting signalling pathways in tumour angiogenesis,”British Journal of Cancer, vol. 99, no. 8, pp. 1204–1209, 2008.

[36] J. L. Li, R. C. A. Sainson, W. Shi et al., “Delta-like 4 Notchligand regulates tumor angiogenesis, improves tumor vascularfunction, and promotes tumor growth in vivo,” CancerResearch, vol. 67, no. 23, pp. 11244–11253, 2007.

[37] M. J. Van Amerongen, G. Molema, J. Plantinga, H. Moorlag,and M. J. A. Van Luyn, “Neovascularization and vascularmarkers in a foreign body reaction to subcutaneouslyimplanted degradable biomaterial in mice,” Angiogenesis, vol.5, no. 3, pp. 173–180, 2002.

[38] T. Yano, S. Tanikawa, T. Fujie, M. Masutani, and T. Horie,“Vascular endothelial growth factor expression and neovas-cularisation in non-small cell lung cancer,” European Journalof Cancer, vol. 36, no. 5, pp. 601–609, 2000.

[39] C. Morrissey, L. D. True, M. P. Roudier et al., “Differentialexpression of angiogenesis associated genes in prostate

Page 14: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

14 ISRN Oncology

cancer bone, liver and lymph node metastases,” Clinical andExperimental Metastasis, vol. 25, no. 4, pp. 377–388, 2008.

[40] M. M. Baldewijns, V. L. Thijssen, G. G. Van Den Eynden etal., “High-grade clear cell renal cell carcinoma has a higherangiogenic activity than low-grade renal cell carcinoma basedon histomorphological quantification and qRT-PCR mRNAexpression profile,” British Journal of Cancer, vol. 96, no. 12,pp. 1888–1895, 2007.

[41] G. Molema, “Angiogenesis: an integrative approach fromscience to medicine,” in Targeted Drug Delivery to the TumorNeovasculature, W. D. Figg and J. Folkman, Eds., pp. 283–298,Springer, New York, NY, USA, 2008.

Page 15: TumorVascularMorphologyUndergoesDramatic ...downloads.hindawi.com/archive/2011/409308.pdf2.2. Gene Expression Analysis by Quantitative RT-PCR. Ex-traction of total RNA from cryostat-cut

Submit your manuscripts athttp://www.hindawi.com

Stem CellsInternational

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Disease Markers

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation http://www.hindawi.com Volume 2014

Immunology ResearchHindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Parkinson’s Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttp://www.hindawi.com