Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Cancer
Research
Microenvironment and Immunology
Fibulin-5 Blocks Microenvironmental ROS in
Pancreatic Cancer
Miao Wang1, Mary Topalovski1, Jason E. Toombs1, Christopher M. Wright1,
Zachary R. Moore2, David A. Boothman2, Hiromi Yanagisawa3, Huamin Wang4,
Agnieszka Witkiewicz5, Diego H. Castrillon5, and Rolf A. Brekken1,6,7
Abstract
Elevated oxidative stress is an aberration seen in many solid
tumors, and exploiting this biochemical difference has the potential
to enhance the efficacy of anticancer agents. Homeostasis of reactive
oxygen species (ROS) is important for normal cell function, but
excessive production of ROS can result in cellular toxicity, and
therefore ROS levels must be balanced finely. Here, we highlight
the relationship between the extracellular matrix and ROS production by reporting a novel function of the matricellular protein
Fibulin-5 (Fbln5). We used genetically engineered mouse models
of pancreatic ductal adenocarcinoma (PDAC) and found that
mutation of the integrin-binding domain of Fbln5 led to decreased
tumor growth, increased survival, and enhanced chemoresponse to
standard PDAC therapies. Through mechanistic investigations, we
found that improved survival was due to increased levels of
oxidative stress in Fbln5-mutant tumors. Furthermore, loss of the
Fbln5–integrin interaction augmented fibronectin signaling, driving integrin-induced ROS production in a 5-lipooxygenase–dependent manner. These data indicate that Fbln5 promotes PDAC
progression by functioning as a molecular rheostat that modulates
cell–ECM interactions to reduce ROS production, and thus tip the
balance in favor of tumor cell survival and treatment-refractory
disease. Cancer Res; 75(23); 5058–69. 2015 AACR.
Introduction
an RGD sequence, but does not support integrin activation (8).
Thus, Fbln5 competes with structural ECM ligands, principally
fibronectin, that would otherwise activate these integrins.
ECM proteins stimulate the generation of reactive oxygen
species (ROS) in an integrin-dependent manner (2, 10, 11). ROS
generation in this context is generally transient and serves primarily as a signaling intermediate that enhances cellular activity
(12). We reasoned that a chronic increase in integrin-induced ROS
would negatively affect tumor growth. Fibronectin-driven ROS
generation is an attractive pathway to exploit for this strategy
because fibronectin ligation of b1 integrins is governed in part by
Fbln5 (8, 13). We reported previously that Fbln5 reduced fibronectin-mediated integrin-induced ROS production by competing
with fibronectin for binding to a5b1 integrin (13). Mutation of
the three amino acid RGD sequence in Fbln5 to RGE abolishes
integrin binding yet preserves other functions of Fbln5 (14, 15).
An essential function of Fbln5 is elastic fiber formation (16). As a
result, Fbln5 / mice exhibit disorganized elastic fibers throughout the body, leading to tortuous great vessels, emphysematous
lungs, and loose skin, resembling cutis laxa syndrome in humans
(17, 18). In contrast, Fbln5RGE/RGE (RGE) mice have intact elastic
fibers and are essentially indistinguishable from wild-type (WT)
littermates (15). However, RGE mice show increased levels of ROS
compared with WT animals in tissues where fibronectin is abundant (15). Given the high expression of fibronectin in the stroma
of PDAC and increasing evidence supporting enhanced ROS
production as an anticancer strategy (19), we evaluated the
consequence of ablating the integrin-binding ability of Fbln5 in
robust spontaneous models of PDAC. Our results show that
Fbln5–integrin interaction promotes aggressive tumor growth
and progression in mice and that 5-lipooxygenase (5-Lox) activity
was required for ROS induction in the absence of functional
Fbln5. In addition, we found that Fbln5 was expressed
Tumors develop and progress in the context of the extracellular
matrix (ECM). In fact, structural ECM proteins can promote
tumor cell survival and stimulate invasive tumor cell programs
(1–6). This is particularly evident in desmoplastic tumors, such as
pancreatic ductal adenocarcinoma (PDAC; 7), where ECM proteins, including fibronectin and collagen, activate signaling pathways that drive cell survival, proliferation, and migration (1, 2).
ECM-mediated signaling is governed by expression of the ECM
proteins, the presence of cell surface receptors and the expression
and activity of matricellular proteins that function as extracellular
adaptors to reduce ECM–cell interaction (1, 8, 9). Fibulin-5
(Fbln5), a member of the fibulin family of proteins, is particularly
important in this regard, as it binds to a4b1 and a5b1 integrins via
1
Hamon Center for Therapeutic Oncology Research, UT Southwestern
Medical Center, Dallas, Texas. 2Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas. 3Department of
Molecular Biology, UT Southwestern Medical Center, Dallas, Texas.
4
Department of Pathology, UT MD Anderson Cancer Center, Houston,
Texas. 5Department of Pathology, UT Southwestern Medical Center,
Dallas, Texas. 6Department of Surgery, UT Southwestern Medical Center, Dallas, Texas. 7Department of Pharmacology, UT Southwestern
Medical Center, Dallas, Texas.
Note: Supplementary data for this article are available at Cancer Research
Online (http://cancerres.aacrjournals.org/).
M. Wang and M. Topalovski contributed equally to this article.
Corresponding Author: Rolf A. Brekken, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry
Hines Boulevard MC 8593, Dallas, TX 75390-8593. Phone: 214-648-5151; Fax:
214-648-4940; E-mail:
[email protected]
doi: 10.1158/0008-5472.CAN-15-0744
2015 American Association for Cancer Research.
5058 Cancer Res; 75(23) December 1, 2015
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Fbln5 Reduces ROS in Pancreatic Cancer
abundantly in the stroma of human PDAC tumors. These data
provide insight into the function of Fbln5 in PDAC and reveal
how ECM signaling might be exploited to drive pro-oxidant
therapy.
Materials and Methods
Mouse models
Fbln5RGE/RGE (RGE), Fbln5 / (KO), LSL-KrasG12D/þ; Cdkn2aLox/Lox
(KI) and Cdkn2aLox/Lox; p48Cre (IC) mice were generated as previously
described (1, 15, 20, 21). RGE mice were used to breed with KI
and IC mice to generate genetically matched LSL-KrasG12D/þ;
Cdkn2aLox/Lox; p48Cre (KIC) mice and RGE-KIC mice. LSLTrp53R172H/þ mice were obtained from National Cancer Institute
(NCI) Mouse Repository (22). RGE mice were also used to breed
with LSL-KrasG12D/þ; LSL-Trp53R172H/þ (KP) and p48Cre mice to
generate genetically matched LSL-KrasG12D/þ; LSL-Trp53R172H/þ;
p48Cre (KPC) mice and RGE-KPC mice. All mice were housed in a
pathogen-free facility and all experiments were performed under
written protocols approved by the Institutional Animal Care and
Use Committee at the University of Texas Southwestern Medical
Center at Dallas.
Animal studies
For endpoint studies, KIC and RGE-KIC mice were sacrificed and
entire tissues, including pancreas/tumor, liver, and spleen, were
harvested and weighed at 1, 1.5, and 2-months-old. KPC and RGEKPC mice were sacrificed at 3 and 5 months, n 8 mice per time
point per group. For all survival studies, mice were carefully
monitored and sacrificed when they appeared moribund. For
antioxidant treatment, N-acetyl cysteine (NAC; Sigma-Aldrich) was
given to mice at 7 mg/mL in the drinking water from 4-weeks-old
until moribund. For endpoint therapy experiments, KIC and RGEKIC mice were treated for 3 weeks starting at 7 weeks (1.5 months)
of age with i.p. injection of low dose Gemcitabine (GemL; 12.5
mg/kg 3/week) or Abraxane (Abx; 5 mg/kg 2/week). Mice were
sacrificed and tissues were isolated for analysis, n 6 mice per
group. For survival studies with therapy, cohorts of KIC and RGEKIC mice were treated similarly with GemL, high dose Gemcitabine
(GemH; 50 mg/kg 1/week i.p.) or Abx until moribund.
Histology, immunohistochemistry, and immunofluorescence
staining
Tissues were snap frozen and embedded in OCT (Tissue-Tek) for
frozen sections or fixed with 4% paraformaldehyde overnight and
embedded in paraffin for sectioning. Frozen sections were fixed in
ice-cold acetone for 5 minutes, air dried for 10 minutes followed
by 10 minutes incubation with PBS to dissolve the OCT. Paraffin
sections were deparaffinized and rehydrated with xylene and serial
dilutions of ethanol followed by antigen retrieval with 0.01 mol/L
citric acid buffer (pH 6.0). Sections were blocked with 20% aquablock and incubated with primary antibodies in blocking solution
(5% BSA in TBST) at 4 C overnight. Primary antibodies used for
frozen sections were: rabbit polyclonal anti-mouse Fbln5 (1:100;
purified polyclonal IgG by our lab, 1.6 mg/mL; ref. 17), rat antiMeca32 (1:100; purified IgG from hybridoma by our laboratory;
1.0 mg/mL; ref. 23), goat anti-Amylase (1:2,000; sc-12821; Santa
Cruz Biotechnology), rabbit anti-fibronectin (1:100; DP3060;
Acris), and rabbit anti-gH2AX (1:50; NB100-2280; NOVUS). Primary antibodies used for paraffin sections were: rabbit anti-human
Fbln5 (1:75; HPA000868; Sigma-Aldrich), rabbit anti-Phospho-
www.aacrjournals.org
Histone H3 (PH3; 1:100; 06-570, Millipore), rabbit anti-Amylase
(1:2,000; 3796S; Cell Signaling Technology), and rat anti-endomucin (1:100; sc-65495, Santa Cruz Biotechnology). Fluorescein isothiocyanate (FITC)–conjugated donkey anti-rabbit, rat IgGs, Cyanine 3 (Cy3)–conjugated donkey anti-rat, mouse, rabbit IgGs
and horseradish peroxidase–conjugated donkey anti-rabbit IgGs
from Jackson ImmunoResearch were used as secondary antibodies.
Slides with sections of FFPE de-identified human pancreatic
cancer tissue were obtained from the UT Southwestern Tissue
Resource and the Department of Pathology, UT MD Anderson
Cancer Center. Human PDAC sections were stained for Fbln5
expression using rabbit anti-Fbln5 (HPA000868; Sigma-Aldrich)
as indicated above.
Western blot analysis
Western blots were performed as previously described (24). In
brief, protein lysates from cell culture or tumor tissues were extracted
in ice-cold RIPA buffer (50 mmol/L Tris-Cl, 150 mmol/L NaCl, 1%
NP-40, 0.5% sodium deoxycholate, 0.1% SDS) containing cocktails
of protease (Thermo Fisher Scientific) and phosphatase inhibitors
(Sigma-Aldrich), by centrifugation (13,000 g, 15 minutes) at 4 C
following three freeze-thaw cycles. Proteins were separated by
SDS-PAGE and transferred to methanol-activated polyvinylidene
difluoride membrane (VWR). The primary antibodies used include
the following: rabbit anti-mouse Fbln5 (1:1,000), rabbit anti-human
Fbln5 (1:500; HPA000868; Sigma-Aldrich), anti-Nqo1 (1:1,000;
ab34173; Abcam), anti–a-tubulin (1:1,000; ab4047; Abcam) and
anti–b-actin (1:5,000; A2066; Sigma-Aldrich). Horseradish peroxidase–conjugated donkey anti-rabbit IgG (1:10,000; Jackson Immunoresearch) as secondary antibodies were used.
Cell culture
Cell lines used include mouse endothelial cell (EC) line bEnd.3
(13), mouse pancreatic cancer cell lines Pan02 (13) and mPLR B9
(1), and human pancreatic cell lines MiaPaCa-2, AsPC-1, and
Panc1 (all purchased from the ATCC). NG2þ cells were isolated
using anti-NG2 antibody–conjugated magnetic beads from a KIC
tumor (25). Fbln5þ/þ (WT), Fbln5 / (KO) and Fbln5RGE/RGE
(RGE) mouse embryonic fibroblasts (MEF) were isolated from
embryonic day E12.5-E14.5 embryos and genotypes were confirmed by PCR. bEnd.3 cells were treated with 100 mmol/L H2O2
or 10 mg/mL a5b1 integrin–activating antibody and lysates were
collected for Western blot analysis. MEFs were cultured in reduced
serum medium Opti-MEM (Life Technologies) overnight before
being plated on plastic, fibronectin (Sigma-Aldrich) or collagen I
(Thermo Fisher Scientific), each at 10 mg/mL unless otherwise
noted. After plating, MEFs were grown in serum-free medium
supplemented with fibronectin, collagen, b1 integrin blocking
antibody (each at 10 mg/mL) or various chemicals. Inhibitors used
for various ROS sources include Rotenone (R8875-1G; Sigma
Aldrich), Diphenyleneiodonium chloride (DPI; D2926, SigmaAldrich) and nordihydroguaiaretic acid (NDGA; 479975; Millipore). All cells were maintained in DMEM (Mediatech, Inc.) with
10% FBS and were grown in 37 C humidified incubator with 5%
CO2. MEFs were used between passages 2–5 for all experiments.
ROS detection
The detailed protocol on ROS detection and quantification has
been described previously (13). In brief, for tissues, 5 mmol/L
dihydroethidium (DHE; D11347; Life Technologies) was applied
to freshly sectioned tissues and incubated at 37 C for 30 minutes.
Cancer Res; 75(23) December 1, 2015
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
5059
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Wang et al.
6–10 images were taken randomly from each tissue and at least
three tissues were included in each group. Fluorescence intensity
was quantified by the software NIS-Elements. To visualize ROS in
cells, 10 mmol/L of 20 -70 -dicholordihydrofluorescein diacetate
(DCF-DA; D-399, Life Technologies) was added to cells grown
on fibronectin-coated chamber slides. 8–10 pictures were taken
randomly from each condition and area fraction was quantified
and normalized to cell number by DAPI using the software NISElements. Three independent experiments for each condition
were evaluated.
qPCR array and real-time PCR
WT and RGE MEFs were cultured in reduced serum medium
Opti-MEM overnight before plating on fibronectin. Upon plating,
MEFs were grown in serum-free medium supplemented with
fibronectin for 4, 16, and 24 hours. RNA lysates were isolated
using the RNeasy Plus Mini Kit (Qiagen). The RT2 First Strand Kit
(Qiagen) was used for cDNA synthesis. Then cDNA samples were
subjected to RT2 Profiler PCR array to analyze gene-expression
changes related to mouse oxidative stress and antioxidant defense
pathways (Qiagen; PAMM-065A). Experiments were performed
and data were analyzed following the manufacturer's instructions.
All the candidate genes were further checked and confirmed by
real-time PCR. Ribosome protein S6 (RPS6) was used as the
internal control. Following primers were used for real-time PCR:
Nqo1 (forward): 50 -AGACCTGGTGATATTTCAGTTCCCATTG-30 ;
Nqo1 (reverse): 50 - CAAGGTCTTCTTATTCTGGAAAGGACCGT30 ; RPS6 (forward): 50 -AAGCTCCGCACCTTCTAT-30 ; RPS6R
(reverse): 50 -TGACTGGACTCAGACTTAGAAGTAGAAGC-30 .
Nqo1 activity assay
Nqo1 enzyme activity was measured in a reaction mixture
containing 200 mmol/L NADH (Sigma-Aldrich) as an electron
donor and 10 mmol/L menadione (Sigma-Aldrich) as an Nqo1
substrate and intermediate electron acceptor as described (26,
27). Cytochrome c serves as the terminal electron acceptor.
Therefore, the measured rate of cytochrome c reduction correlates
with Nqo1 enzymatic activity. To prepare lysates, cells were
scraped in PBS and samples were sonicated. Lysate was added to
the reaction mixture and the reduction of cytochrome c (SigmaAldrich) over two minutes was monitored by absorbance at 550
nm. Dicoumarol, a selective inhibitor of Nqo1, was added as a
negative control. Enzyme activity units were calculated as nmol of
cytochrome c reduced/min/mg lysate.
Statistical analysis
For statistical analysis, the unpaired t test was used for comparison between genotypes and various groups. The log-rank
Figure 1.
Expression pattern of Fbln5 in mouse
and human. A and B, protein lysates
from mouse ECs, cancer cells and
fibroblasts (A), and human PDAC
tissue and cancer cell lines (B) were
probed for indicated targets by
Western blot analysis. C and D, IHC
staining for Fbln5 on mouse (C) and
human (D) PDAC sections. E,
immunofluorescence staining of Fbln5
(red) and MECA-32 (green) on
subcutaneously grown pancreatic
tumor (Pan02) in Fbln5 WT and KO
mice. F, representative images of
FBLN5 expression in human PDAC,
showing heterogeneous expression
in the stroma. G and H,
immunofluorescence staining of
WT pancreas, KIC and RGE-KIC
tumors, KPC and RGE-KPC tumors
for Fbln5 (green), and EC marker
Meca32 (Meca; red) in G, acinar cell
marker amylase (Amy; green), and
fibronectin (red) in H. Nuclei were
counterstained with DAPI (blue).
Scale bars are presented as indicated.
5060 Cancer Res; 75(23) December 1, 2015
Cancer Research
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Fbln5 Reduces ROS in Pancreatic Cancer
(Mantel–Cox) test was used for all the mouse survival studies.
Overall, P value less than 0.05 was considered as statistically
significant; , P < 0.05; , P < 0.01; , P < 0.001; , P < 0.0001.
Results
Fbln5 expression in pancreatic cancer
The expression of Fbln5 is prominent in developing arteries and
is diminished in most adult organs, but can be reactivated in
injured vessels (14). We examined Fbln5 expression in multiple
mouse and human cell lines and tumor lysates. The mouse EC line
bEnd.3 and fibroblasts, including MEFs and fibroblasts isolated
from mouse PDAC (NG2þ cells), expressed Fbln5 (Fig. 1A and
Supplementary Fig. S1B) as did human and mouse PDAC lysates
(Fig. 1B and Supplementary Fig. S1C). In contrast, pancreatic
cancer cell lines, including three human (MiaPaCa-2, Panc1, and
AsPC-1; Fig. 1B) and four mouse lines (Pan02 and three isogenic
lines isolated from mouse PDAC), did not express detectable
levels of Fbln5 protein (Fig. 1A, Supplementary Fig. S1B and data
not shown). However, all cell lines and PDAC tumor lysates
examined express a5b1 integrin (Supplementary Fig. S1A and
S1C), which serves as the cell surface receptor for Fbln5 and
fibronectin (8).
Fbln5 IHC in syngenic pancreatic Pan02 tumors grown subcutaneously in Fbln5þ/þ (WT) or Fbln5 / (KO) mice revealed
that Fbln5 is produced by stromal cells (Fig. 1E). Costaining
of Fbln5 with the EC marker Meca32 shows that Fbln5 can be
produced by ECs within the tumor (Fig. 1E). IHC analysis of
Fbln5 expression in multiple mouse models of PDAC revealed
Fbln5 reactivity mainly in the stroma (Fig. 1C and D and data
not shown). We also examined FBLN5 in human PDAC by IHC
and found the protein was expressed in all human PDACs
examined (n ¼ 25). The staining pattern was confined largely
to the stromal compartment; however, not all regions of stroma
Figure 2.
RGE-KIC and RGE-KPC mice show
reduced tumor growth and prolonged
survival compared with KIC and KPC
mice. A and B, whole tumors were
isolated, weighed, and normalized
against body weight at 2 months for
KIC and RGE-KIC mice (A) or 3 and
5 months for KPC and RGE-KPC mice
(B). n 7 tumors per group. C and E,
immunofluorescence staining on
tumor sections for phospho-Histone
H3 (PH3; green). n 4 tumors per
group. D and F, quantification of PH3
positive (þ) cells per 20 field from 4
to 5 tumors per group with 8 to 10
pictures per tumor. Results are shown
as mean SEM. G and H, Kaplan–Meier
survival curve of KIC and RGE-KIC
mice (G), KPC and RGE-KPC mice (H).
Scale bars are presented as indicated.
For statistical analysis, the unpaired
t test was used for A, B, D, and F.
The log-rank test was used for G and H;
, P < 0.05; , P < 0.01; , P < 0.001;
, P < 0.0001.
www.aacrjournals.org
Cancer Res; 75(23) December 1, 2015
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
5061
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Wang et al.
Figure 3.
Increased oxidative stress and reduced MVD and EC proliferation in RGE KIC and RGE KPC tumors compared with KIC and KPC tumors. A and B, dihydroethidium
(DHE; red) staining on freshly cut frozen sections of KIC and RGE-KIC (A), KPC and RGE-KPC (B) tumors for in situ detection of ROS. Relative ROS level was
quantified by fluorescence intensity using the software NIS-Elements and are shown inside images. Quantification was from three tumors per group with 6 to 10 images
per tumor. C, immunofluorescence staining on KIC and RGE-KIC tumor sections for Meca32 (Meca; green) and g-H2AX (red). n ¼ 3 tumors per group. D, Kaplan–Meier
survival curve of KIC and RGE-KIC mice treated with the antioxidant NAC by drinking water starting at 4-weeks-old. E and G, MVD was counted per 20 field
from 4 to 5 KIC and RGE-KIC (E), KPC and RGE-KPC (G) tumors with 8 to 10 pictures per tumor. F and H, quantification of MVD for KIC and RGE-KIC (F), KPC and RGE-KPC
(H) tumors using the software NIS-Elements. Endomucin (Endo)-stained areas were counted as the percentage area fraction. I and L, immunofluorescence staining on
2-month-old KIC and RGE-KIC (I) and 3-month-old KPC and RGE-KPC (L) tumor sections for PH3 (green) and endomucin (red). (Continued on the following page.)
5062 Cancer Res; 75(23) December 1, 2015
Cancer Research
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Fbln5 Reduces ROS in Pancreatic Cancer
were positive for FBLN5 protein (Fig. 1F). The nature of the
heterogeneous stromal staining pattern is unclear but suggests
that FBLN5 expression is controlled tightly.
Characterization of KIC and KPC mice
To evaluate the contribution of Fbln5 to PDAC development
and progression, we used KIC and KPC mice, two well established
conditional genetically engineered mouse models (GEMM) of
PDAC based on the p48Cre (also known as Ptf1a) driver, which is
expressed in pancreatic bud progenitor cells (22, 28,29). KIC
animals express an active form of Kras and have biallelic inactivation of the Cdkn2a locus (LSL-KrasG12D/þ; Cdkn2aLox/Lox; p48Cre;
ref. 29). KPC animals express the same activating G12D mutation
in Kras and also harbor a R172H point mutation in p53, the LiFraumeni human ortholog (LSL-KrasG12D/þ; LSL-Trp53R172H/þ;
p48Cre; ref. 22). Histologic examination of KIC and KPC pancreatic
tissue by a pathologist revealed that each model developed early
pancreatic intraepithelial neoplasias (PanINs) and highly infiltrative adenocarcinomas ranging from well-differentiated areas
with clear malignant gland formation to areas that were more
poorly differentiated (Supplementary Fig. S2A). Similar to human
PDAC, Masson's trichrome staining showed extensive collagen
deposition in the area of PanINs and PDAC in KIC and KPC
tumors (Supplementary Fig. S2B).
Ablation of Fbln5–integrin interaction reduces tumor growth
and prolongs survival
Mutation of the Fbln5–integrin-binding sequence from RGD to
RGE renders the protein incapable of binding to integrins (14).
Fbln5RGE/RGE (RGE) mice are viable, fertile and phenotypically
normal compared with WT animals (15). To study the contribution of Fbln5 to PDAC development, we crossed RGE mice with
KIC or KPC animals to generate genetically matched KIC and RGEKIC, KPC and RGE-KPC mice. There was no difference in Fbln5
expression levels between KIC and RGE-KIC or KPC and RGE-KPC
tumors (Fig. 1G and Supplementary Fig. S1D). However, tumors
had significantly increased Fbln5 expression compared with normal pancreas (Fig. 1C and G and Supplementary Fig. S1D).
Pancreatic and mouse body weights were determined in KIC and
RGE-KIC mice at 1, 1.5, and 2 months (Supplementary Fig. S2C
and 2A). There is no significant difference for tumor/body weight
at 1 and 1.5 months (Supplementary Fig. S2C). RGE-KIC mice
exhibited significantly lower tumor/body weight at 2 months than
KIC mice, with tumor weights ranging from 0.24 to 0.86 gram for
RGE-KIC mice and 0.72 to 1.11 gram for KIC mice (Fig. 2A). This is
consistent with significantly reduced proliferating cells in RGEKIC tumors (Fig. 2C and D). Similar results were observed in the
KPC model, which were analyzed at 3 and 5 months of age
(Fig. 2B, E, and F).
Survival analysis revealed that RGE-KIC mice lived significantly
longer than KIC mice (Fig. 2G) with a median survival of 74 days
for RGE-KIC mice and 61.5 days for KIC mice. KIC and RGE-KIC
mice appeared normal with no obvious phenotype up to 1.5
months of age but later became moribund, usually accompanied
by weight loss. Moreover, some mice developed jaundice or
ascites. Autopsies revealed the presence of large solid tumors
with limited gross metastases. Liver micrometastasis was seen in
the majority of KIC and RGE-KIC mice in the survival study
necropsies (Supplementary Fig. S2D). Similarly, RGE-KPC
show significantly prolonged survival compared with KPC mice
(175 vs. 143.5 days; Fig. 2H). KPC and RGE-KPC mice appeared
healthy up to 3-month-old and were sacrificed between 3 and 9
months, commonly presenting with body weight loss, jaundice,
or ascites. Gross liver metastasis was seen in 30% to 40% of
animals (Supplementary Fig. S2E and S2F).
Increased oxidative stress in RGE-KIC and RGE-KPC tumors
We reported previously that the growth of subcutaneous
Pan02 tumors in Fbln5 / mice was significantly reduced compared with WT animals due to increased ROS (13). Dihydroethidium (DHE) staining of tumors from the GEMMs
showed that the Fbln5 RGE mutation significantly induced
ROS levels in KIC and KPC tumors (Fig. 3A and B). Accordingly,
the level of gH2AX, a commonly used marker for oxidative
stress-induced DNA damage (30), was higher in RGE-KIC
tumors than KIC tumors (Fig. 3C). However, in the context of
normal pancreatic tissue, the Fbln5 RGE mutation did not alter
ROS levels (Supplementary Fig. S3A and S3B). This is consistent
with the level of fibronectin expression, which was elevated in
PDAC compared with normal pancreatic tissue (Fig. 1H). To
determine whether ROS induction contributed to the prolonged survival in RGE animals, KIC and RGE-KIC mice were
treated with the antioxidant NAC and survival was examined
(Fig. 3D). Prolonged NAC treatment decreased survival of RGEKIC mice but did not affect the survival of KIC mice (Fig. 3D).
ROS production was also examined in NAC-treated KIC and
RGE-KIC tumors, which revealed no difference between the two
groups (Supplementary Fig. S3C and S3D). Collectively, ROS
induction driven by the Fbln5 RGE mutation resulted in
reduced tumor growth and prolonged survival.
Angiogenesis is reduced in RGE-KIC and RGE-KPC tumors
Prior studies indicate that Fbln5 can modulate angiogenesis
(31), and we reported that loss of Fbln5 resulted in decreased
angiogenesis in pancreatic tumors (13). Therefore, we examined microvessel density (MVD) in KIC and RGE-KIC, KPC, and
RGE-KPC tumors by co-immunostaining with the EC marker
endomucin and the acinar cell marker amylase. MVD was
significantly reduced in RGE-KIC compared with KIC in tumor
regions as marked by loss of amylase reactivity (Fig. 3E and F
and Supplementary Fig. S4A). Immunostaining and quantification of MVD from 3- to 5-month-old KPC and RGE-KPC
tumors also revealed significantly reduced MVD in RGE-KPC
tumors (Fig. 3G and H and Supplementary Fig. S4B and S4C).
We also examined the MVD of KIC and RGE-KIC tissues at
1-month-old. At this time point, more than 90% of the tissue
retained amylase expression. There was no difference in MVD
in amylase-positive areas between KIC and RGE-KIC tissues
(Continued.) Arrows indicate double-labeled ECs, one of which was enlarged and is shown in an inset box for each image. J and M, quantification of PH3 and
þ
endomucin costained cells (PH3 ECs) per 20 field in KIC and RGE-KIC (J) and KPC and RGE-KPC (M) tumors at indicated ages. K, quantification of the percentage
þ
of PH3 ECs over total number of ECs in 20 field in KIC and RGE-KIC tumors. Scale bars are presented as indicated. All the results shown are mean SEM.
For statistical analysis, the unpaired t test was used for E–H, J and K, and M; , P < 0.05; , P < 0.01; , P < 0.001; , P < 0.0001.
www.aacrjournals.org
Cancer Res; 75(23) December 1, 2015
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
5063
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Wang et al.
(Supplementary Fig. S5A–S5C). In addition, pancreas tissue
from nontumor-bearing WT and RGE mice were analyzed for
MVD (Supplementary Fig. S5D). Again, no significant difference between the two groups was observed (Supplementary Fig.
S5E and S5F). We found that the number of proliferating ECs
costained with phospho-histone H3 and endomucin was
decreased in tumors in RGE animals compared with tumors
in WT mice, supporting the reduction of MVD in RGE-KIC and
RGE-KPC tumors (Fig. 3I–M and Supplementary Fig. S4D and
S4E). Overall, the reduction of MVD correlated with tumorspecific induction of ROS in RGE animals (Fig. 3A and Supplementary Fig. S4A, 3B and Supplementary Fig. S4B and S4C).
These data suggest that the absence of functional Fbln5 impairs
EC survival specifically in the tumor microenvironment.
Induction of the oxidative stress–responsive gene Nqo1 by
fibronectin-induced ROS in vitro and in vivo
Our data suggested that Fbln5 controls ROS production
through fibronectin–b1 integrin interaction. To elucidate the
underlying molecular mechanism of ROS generation, we isolated
WT, KO, and RGE primary MEFs. Fbln5 expression levels were
equivalent between WT and RGE MEFs (Supplementary Fig. S6).
We found that ROS was elevated in KO and RGE MEFs but not in
WT MEFs when cells were plated on fibronectin (Fig. 4A). Next, we
performed qPCR arrays to screen for oxidative stress and antiox-
idant response pathway–related genes using RNA harvested from
WT and RGE MEFs. From these arrays, NADP(H):quinone oxidoreductase 1 (Nqo1) was a reproducible and reliable target that
was increased in RGE MEFs after plating on fibronectin. Nqo1 is
an antioxidant enzyme that is responsible for the reduction of
quinones to hydroquinones using NAD(P)H as an electron donor
(32). Reducing quinone levels lowers the occurrence of ROS
generation as a result of redox cycling (32). Induction of Nqo1
can be mediated by the Keap1–Nrf2–ARE pathway (33). The
induction of Nqo1 in RGE MEFs when plated on fibronectin was
confirmed by quantitative real-time PCR (Fig. 4B), enzymatic
activity (Fig. 4C), and Western blotting (Fig. 4D). Concordantly,
the induction of Nqo1 expression was reversed by antioxidant
treatment (with NAC) in a dose-dependent manner (Fig. 4E),
showing that the elevation of Nqo1 is a consequence of increased
ROS status in RGE MEFs. In addition, Nqo1 induction was
elevated in tumors from RGE animals (Fig. 4F and G).
Nqo1 induction is dependent on fibronectin–b1 integrin
interaction and 5-Lox activity
It has been reported that integrin activation by fibronectin
can induce ROS production (12). Accordingly, when the EC
line bEnd.3 was treated with a a5b1 integrin-activating antibody, Nqo1 levels were induced (Fig. 5A). The induction of
Nqo1 was specific to activation by fibronectin and was not
Figure 4.
Fbln5 RGE mutation induces ROS
production and oxidative stress–
responsive protein Nqo1 in vitro and in
vivo. A, MEFs harvested from Fbln5
WT, KO, and RGE mice were grown on
fibronectin for 16 hours and stained
with DCF-DA (green) to detect ROS.
Nuclei were counterstained in blue
with DAPI. B, real-time PCR result with
RNA isolated from WT or RGE MEFs
plated on fibronectin for 24 hours. C,
enzymatic activity of Nqo1 was
measured and normalized against
protein concentration with samples
isolated from WT or RGE MEFs plated
on fibronectin for 4 hours. D, Western
blot analysis using lysates harvested
from WT or RGE MEFs plated on
fibronectin for 1 or 4 hours.
E, Western blot analysis using lysates
harvested from WT or RGE MEFs
plated on fibronectin for 4 hours and
treated with increasing concentration
of antioxidant NAC. F and G, Western
blot analysis using lysates harvested
from several randomly selected KIC
and RGE-KIC tumors (F) or KPC and
RGE-KPC tumors (G). a-Tubulin or
b-actin was used as loading control.
Scale bars are presented as indicated.
All the results in B and C are mean
SEM. For statistical analysis, the
unpaired t test was used for B and C;
, P < 0.001; , P < 0.0001.
5064 Cancer Res; 75(23) December 1, 2015
Cancer Research
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Fbln5 Reduces ROS in Pancreatic Cancer
Figure 5.
5-Lox activation through fibronectin–
integrin interaction is responsible for
ROS induction in RGE MEFs. A, bEnd.3
cells were plated on fibronectin for 4
hours and treated with 100 mmol/L H2O2
or 10 mg/mL a5 integrin–activating
antibody at time of plating and probed
for Nqo1 by Western blot analysis. B, WT
or RGE MEFs were plated on plastic,
fibronectin, fibronectin þ b1 integrin–
blocking antibody (10 mg/mL) or
collagen (CN) for 4 hours. Lysates were
then harvested and subjected to
Western blot analysis. C, RGE MEFs were
plated on fibronectin and treated with
the NOX inhibitor DPI or the
mitochondrial electron transport chain
inhibitor Rotenone at the time of plating.
Lysates were harvested after 4 hours for
Western blot analysis. D, RGE MEFs
were plated on fibronectin and treated
with a 5-Lox inhibitor (NDGA) at the
time of plating and harvested 4 hours
later for Western blot analysis.
E, quantification results of relative Nqo1
protein levels from D using the software
Image Studio Digits. a-Tubulin or
b-actin was used as loading control for
all the Western blots.
present when cells were plated on plastic or collagen (Fig. 5B).
Induction was partially blocked by b1 integrin blockade (Fig.
5B). Given this data, we conclude that the induction of Nqo1 is
responsive to ROS production induced by fibronectin-mediated b1 integrin ligation.
To determine the cellular source of ROS production in the
absence of Fbln5–integrin interaction, we used inhibitors for
various ROS sources, including the mitochondrial respiratory
chain inhibitor Rotenone, NADPH oxidase (NOX) inhibitor
diphenyleneiodonium chloride (DPI), and 5-Lox inhibitor, nordihydroguaiaretic acid (NDGA). Treatment with Rotenone or DPI
did not suppress the induction of Nqo1, suggesting that mitochondria and NOX are not the intracellular source of ROS production (Fig. 5C). In addition, there was no induction of NOX
enzymatic activity in RGE MEFs compared with WT MEFs by
fibronectin (Supplementary Fig. S7). In contrast, inhibition of 5Lox by NDGA reduced Nqo1 levels, indicating 5-Lox as the
potential source of ROS (Fig. 5D and E). This is consistent with
a previous discovery that 5-Lox contributes to a strong burst of
ROS production by fibronectin–integrin engagement (11).
ROS induction has an additive therapeutic effect when
combined with standard chemotherapy agents
To determine whether increased integrin-induced ROS
improved response to chemotherapy, we compared the efficacy
of standard chemotherapy agents Gemcitabine (Gem) and Abraxane (Abx) in KIC and RGE-KIC mice. All therapy started at 1.5
month old when KIC and RGE-KIC mice had established solid
tumors (Supplementary Fig. S2C). We found that low-dose
Gemcitabine (GemL) and Abx were more effective in the context
of mutant Fbln5 (Fig. 6A and B). Survival studies were also
performed with cohorts of KIC and RGE-KIC mice treated with
www.aacrjournals.org
GemL, high-dose Gemcitabine (GemH) and Abx. RGE-KIC mice
consistently survived significantly longer in all three treatment
groups than similarly treated KIC mice (Fig. 6C–E). These data
suggest that increasing integrin-induced ROS augments the activity of standard chemotherapy.
Discussion
We demonstrated that Fbln5 expression is induced in a significant percentage of pancreatic cancers and that it promotes tumor
progression by competing with fibronectin for integrin ligation.
Global loss of Fbln5–integrin interaction resulted in decreased
tumor growth and prolonged survival of tumor-bearing mice with
no apparent adverse effects in normal tissues. The decrease in
tumor burden was dependent on increased fibronectin-mediated
integrin activation, which increased ROS production through 5Lox activity and resulted in reduced angiogenesis in the tumor
microenvironment. These findings are summarized in Fig. 7.
The ECM provides a structural framework in which tumors
develop and progress. ECM signaling contributes to cell survival,
proliferation, and migration; thus, regulation of cellular events
initiated by the ECM is critical for tumor progression. To date,
pharmacologic modification of the ECM in PDAC has not resulted
in improvement of chemoresponse or overall survival in patients
(34–36). However, preclinical studies focused on inhibiting pathways that stimulate ECM deposition (e.g., TGFb) have shown
promise in promoting tumor control (37). Here, we have
highlighted that increased integrin activation can result in
decreased tumor growth by elevating integrin-induced ROS production. The extent of cell–ECM interaction is regulated in
part by matricellular proteins, including Fbln5. Yet, the contribution of Fbln5 to cancer has been limited largely to expression
Cancer Res; 75(23) December 1, 2015
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
5065
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Wang et al.
Figure 6.
RGE KIC mice have increased survival
compared with KIC mice when given
chemotherapy. A and B, KIC and RGE-KIC
mice were treated with 12.5 mg/kg
gemcitabine (Gem) 3/wk (GemL; A) or
5 mg/kg Abraxane (Abx) 2/wk (B) for 3
weeks starting at 7-week-old. Mice were
then sacrificed and tissues were isolated
for analysis. Tumor size is presented as
the mean ratio of tumor/body weight
SEM. n 6 tumors per group. C–E,
Kaplan–Meier survival curve of KIC
control, KIC, and RGE-KIC mice treated
with GemL (C), GemH (D), or Abx (E). All
therapies were given to mice from
7-week-old until moribund. For GemL,
12.5 mg/kg Gem was given to mice
3/wk by i.p. injection. For GemH, 50
mg/kg Gem was given to mice 1/wk
by i.p. injection. For Abx, 5 mg/kg was
given to mice 2/wk by i.p. injection. For
statistical analysis, the unpaired t test was
used for A and B. The log-rank test was
used for C–E; , P < 0.05; , P < 0.01.
studies (38–41), which have not defined a clear function for Fbln5
in tumorigenesis. We found that FBLN5 protein was expressed in
all of the human PDAC samples (n ¼ 25) we evaluated. Expression was largely restricted to the stromal compartment, yet the
pattern of expression was not uniform as there were some areas of
stroma that were negative or only weakly reactive. This heterogeneity suggests that evaluation of FBLN5 and potentially other
matricellular proteins in human tissue microarrays could be
challenging. Additional studies on the expression of Fbln5 protein
in clinically annotated tumor samples are needed to elucidate
whether Fbln5 expression has predictive value.
To extend our studies, we sought to understand how Fbln5–
integrin interaction functions in the context of the microenvironment of PDAC. We used two distinct but related GEMMs of PDAC
that recapitulate common mutations observed in the human
disease (42–44). The expression of Fbln5 in each model is similar
to the expression level and pattern of Fbln5 expression in human
PDAC. Furthermore, fibronectin and a5b1 integrin are expressed
abundantly in animal models of PDAC as well as human PDAC
(45, 46). To study Fbln5–integrin interaction, we took advantage
of the fact that (i) Fbln5 binds but does not activate a5b1 (8, 14),
suggesting that it can function to reduce binding of other ligands
of the integrin; and (ii) knockin mice carrying a point mutation in
the integrin binding domain of Fbln5 (RGE mice) are viable and
fertile (15). The described essential function of Fbln5 is in elastic
fiber assembly as shown by Fbln5-deficient animals (17, 18) and
biochemical analysis (16, 47). However, RGE mice have intact
elastic fibers (15), indicating that Fbln5–integrin binding is not
required for elastic fiber assembly. These data strongly suggest that
the phenotype in the RGE animals is not due to changes in elastic
fiber assembly but a result of an increase in integrin activation by
ligands other than Fbln5. Given the dramatic increase in fibronectin expression as well as other stromal components in PDAC,
we postulated that the tumor microenvironment would provide a
biologically meaningful stress to ascertain the functional consequences of increased integrin ligation in RGE animals.
ROS production as a result of integrin ligation is a well established (11, 12), although underappreciated signaling pathway.
5066 Cancer Res; 75(23) December 1, 2015
Previously, we discovered that the loss of Fbln5–integrin interaction results in increased integrin-induced ROS production (13).
Cellular redox homeostasis is tightly regulated by the balance
between ROS scavenging and eliminating systems (19). Cancer
cells often generate higher levels of ROS due to metabolic abnormality, activation of oncogenes or loss of functional p53 (19). For
example, increased levels of ROS, particularly H2O2 are highly
mutagenic and contribute to elevated mutation levels and heterogeneity. Thus, an imbalance in ROS scavenging and eliminating systems is likely to result in acute consequences in the tumor
microenvironment. For example, increasing ROS levels might
result in inhibition of cell proliferation and ultimately cell death
(48). However, cancer cells have developed adaptive mechanisms
to manage increased ROS levels (19). One adaptive mechanism is
the induction of the antioxidant response transcription factor
Nrf2 to increase the expression of the ROS detoxification enzyme
Nqo1 (49, 50). We found that the loss of Fbln5–integrin interaction induces Nqo1, and that this response is ROS dependent.
Nqo1 levels as a result further validated the elevation of oxidative
stress in tumors grown in RGE mice and also provided a tractable
biochemical endpoint to evaluate the signaling cascade induced
by fibronectin in the absence of integrin-binding Fbln5. Furthermore, using Nqo1 levels as an endpoint, we discovered that
ablation of Fbln5–integrin interaction increased 5-Lox activity in
a fibronectin-dependent manner. Pharmacologic inhibition of 5Lox rescued the fibronectin-driven phenotype in vitro, implicating
that 5-Lox is downstream of integrin activation. This is consistent
with previous reports showing that integrin activation by fibronectin can stimulate ROS production primarily through 5-Lox
(11, 12). Fibronectin has also been reported to stimulate intracellular ROS in pancreatic cancer cells through NOX and the
mitochondria (10), although we did not find evidence of this in
our system.
We found that stromal and tumor cells express the integrin
profile required for fibronectin-induced ROS production. However, changes in ECs were the most apparent phenotype in tumors
from RGE mice. ECs are sensitive to elevated ROS (51) and this
was evident by the consistent reduction in MVD and reduction in
Cancer Research
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Fbln5 Reduces ROS in Pancreatic Cancer
Figure 7.
Fbln5 controls ROS production in the
tumor microenvironment. Fbln5 is
mainly secreted into the tumor
microenvironment by tumorassociated fibroblasts and ECs. Fbln5
competes with fibronectin for integrin
binding. In the absence of Fbln5–
integrin interaction (RGE), more
fibronectin will bind to integrin
receptors and increase ROS
production, resulting in increased
5-Lox activity and reduced
angiogenesis and tumor growth.
proliferating ECs in tumors grown in RGE animals. Fibroblasts
from RGE mice produce elevated levels of ROS in culture in a
fibronectin and integrin-dependent manner. Yet, surprisingly, we
found no significant changes in the presence or activation of
fibroblasts in tumors from RGE mice (data not shown). Global
analysis of ROS using DHE indicates that many cell types, including tumor cells, display elevated ROS levels in tumors from RGE
mice. However, in vitro studies suggest that Fbln5 does not affect
ECM-mediated ROS induction in tumor cells (data not shown). It
is plausible that long-lived ROS molecules (e.g., H2O2) travel
from stromal cells and increase oxidative stress in tumor cells,
resulting in decreased proliferation and reduced tumor growth. It
www.aacrjournals.org
is also feasible that ECs in the tumor microenvironment succumb
to elevated ROS induced by mutant Fbln5 and the decreased
tumor growth is akin to an antiangiogenic effect. In contrast,
Fbln5 null mice display an exaggerated vascular response after
subcutaneous implantation of polyvinyl alcohol sponges (31).
However, the mechanism of how Fbln5 directly affects EC function and the contribution of integrins in this phenotype is poorly
understood. In our model, nontumor-bearing pancreata of WT
and RGE mice show similar MVD (Supplementary Fig. S5).
However, in the context of the tumor microenvironment, the
basal level of ROS is increased compared with normal pancreas;
therefore, inducing further ROS by mutation of Fbln5 may explain
Cancer Res; 75(23) December 1, 2015
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
5067
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Wang et al.
the negative effect on EC function. If so, this suggests that the
mutation in Fbln5 functions as an endogenous inhibitor of
angiogenesis selectively in the tumor microenvironment. These
hypotheses are currently being evaluated.
Our studies show that Fbln5 functions as a rheostat to dampen
integrin-mediated ROS production. This function of reducing
cell–ECM interaction is similar to what has been observed for
other matricellular proteins. For example, SPARC reduces the
binding of fibrillar collagens to discoidin domain receptors,
thereby reducing collagen-induced cell signaling and attachment
(1). Current studies are focused on understanding factors that
drive Fbln5 expression in the tumor microenvironment and
identification of the integrin-mediated signaling pathway that
activates 5-Lox in the context of mutant Fbln5. Overall, our study
illustrates how the matricellular protein Fbln5 functions to reduce
fibronectin–integrin interaction and suggests that Fbln5 is a novel
therapeutic target for pancreatic cancer.
Disclosure of Potential Conflicts of Interest
Acquisition of data (provided animals, acquired and managed patients,
provided facilities, etc.): M. Wang, J.E. Toombs, Z.R. Moore, H. Yanagisawa,
H. Wang, A. Witkiewicz
Analysis and interpretation of data (e.g., statistical analysis, biostatistics,
computational analysis): M. Wang, Z.R. Moore, H. Wang, A. Witkiewicz,
D.H. Castrillon
Writing, review, and/or revision of the manuscript: M. Wang, M. Topalovski,
Z.R. Moore, D.A. Boothman, H. Yanagisawa, H. Wang, D.H. Castrillon,
R.A. Brekken
Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M. Wang, M. Topalovski, C.M. Wright,
D.A. Boothman
Study supervision: D.A. Boothman, R.A. Brekken
Acknowledgments
The authors acknowledge helpful discussions with Drs. Michael Dellinger
and Adi Gazdar and members of the Brekken laboratory.
Grant Support
Authors' Contributions
This work was supported in part by grants from the American Cancer Society
(ACS, RSG-10-244-01-CSM to R.A. Brekken), The Joe and Jessie Crump Medical
Research Foundation (R.A. Brekken), NIH (R01 CA118240 to R.A. Brekken; R01
CA137181 to D.H. Castrillon; and T32 GM008203 to M. Topalovski), the Effie
Marie Cain Scholarship in Angiogenesis Research (R.A. Brekken), and Remeditex Ventures. The UT Southwestern Tissue Resource is supported by the NCI
(5P30 CA142543).
The costs of publication of this article were defrayed in part by the payment of
page charges. This article must therefore be hereby marked advertisement in
accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Conception and design: M. Wang, M. Topalovski, R.A. Brekken
Development of methodology: M. Wang, M. Topalovski, Z.R. Moore,
D.H. Castrillon
Received March 19, 2015; revised July 23, 2015; accepted August 15, 2015;
published OnlineFirst November 17, 2015.
R.A. Brekken reports receiving a commercial research grant from Remeditex
Ventures and has ownership interest in patent on Fbln5 as a target for cancer
therapy. No potential conflicts of interest were disclosed by the other authors.
Disclaimer
The funders had no role in study design, data collection, data analysis,
decision to publish, or preparation of the article.
References
1. Aguilera KY, Rivera LB, Hur H, Carbon JG, Toombs JE, Goldstein CD, et al.
Collagen signaling enhances tumor progression after anti-VEGF therapy in
a murine model of pancreatic ductal adenocarcinoma. Cancer Res
2014;74:1032–44.
2. Aoudjit F, Vuori K. Integrin signaling in cancer cell survival and chemoresistance. Chemother Res Pract 2012;2012:283181.
3. Mantoni TS, Lunardi S, Al-Assar O, Masamune A, Brunner TB. Pancreatic
stellate cells radioprotect pancreatic cancer cells through beta1-integrin
signaling. Cancer Res 2011;71:3453–8.
4. Sebens Muerkoster S, Kotteritzsch J, Geismann C, Gast D, Kruse ML,
Altevogt P, et al. alpha5-integrin is crucial for L1CAM-mediated chemoresistance in pancreatic adenocarcinoma. Int J Oncol 2009;34:243–53.
5. Sethi T, Rintoul RC, Moore SM, MacKinnon AC, Salter D, Choo C, et al.
Extracellular matrix proteins protect small cell lung cancer cells against
apoptosis: a mechanism for small cell lung cancer growth and drug
resistance in vivo. Nat Med 1999;5:662–8.
6. Meredith JE Jr, Fazeli B, Schwartz MA. The extracellular matrix as a cell
survival factor. Mol Biol Cell 1993;4:953–61.
7. Maitra A, Hruban RH. Pancreatic cancer. Annu Rev Pathol 2008;3:157–88.
8. Lomas AC, Mellody KT, Freeman LJ, Bax DV, Shuttleworth CA, Kielty CM.
Fibulin-5 binds human smooth-muscle cells through alpha5beta1 and
alpha4beta1 integrins, but does not support receptor activation. Biochem J
2007;405:417–28.
9. Wong GS, Rustgi AK. Matricellular proteins: priming the tumour microenvironment for cancer development and metastasis. Br J Cancer
2013;108:755–61.
10. Edderkaoui M, Hong P, Vaquero EC, Lee JK, Fischer L, Friess H, et al.
Extracellular matrix stimulates reactive oxygen species production and increases pancreatic cancer cell survival through 5-lipoxygenase and NADPH
oxidase. Am J Physiol Gastrointest Liver Physiol 2005;289:G1137–47.
11. Taddei ML, Parri M, Mello T, Catalano A, Levine AD, Raugei G, et al.
Integrin-mediated cell adhesion and spreading engage different sources of
reactive oxygen species. Antioxid Redox Signal 2007;9:469–81.
5068 Cancer Res; 75(23) December 1, 2015
12. Chiarugi P, Pani G, Giannoni E, Taddei L, Colavitti R, Raugei G, et al.
Reactive oxygen species as essential mediators of cell adhesion: the oxidative inhibition of a FAK tyrosine phosphatase is required for cell
adhesion. J Cell Biol 2003;161:933–44.
13. Schluterman MK, Chapman SL, Korpanty G, Ozumi K, Fukai T, Yanagisawa
H, et al. Loss of fibulin-5 binding to beta1 integrins inhibits tumor growth
by increasing the level of ROS. Dis Model Mech 2010;3:333–42.
14. Nakamura T, Ruiz-Lozano P, Lindner V, Yabe D, Taniwaki M, Furukawa Y,
et al. DANCE, a novel secreted RGD protein expressed in developing,
atherosclerotic, and balloon-injured arteries. J Biol Chem 1999;274:
22476–83.
15. Budatha M, Roshanravan S, Zheng Q, Weislander C, Chapman SL, Davis
EC, et al. Extracellular matrix proteases contribute to progression of pelvic
organ prolapse in mice and humans. J Clin Invest 2011;121:2048–59.
16. Zheng Q, Davis EC, Richardson JA, Starcher BC, Li T, Gerard RD, et al.
Molecular analysis of fibulin-5 function during de novo synthesis of elastic
fibers. Mol Cell Biol 2007;27:1083–95.
17. Yanagisawa H, Davis EC, Starcher BC, Ouchi T, Yanagisawa M, Richardson
JA, et al. Fibulin-5 is an elastin-binding protein essential for elastic fibre
development in vivo. Nature 2002;415:168–71.
18. Nakamura T, Lozano PR, Ikeda Y, Iwanaga Y, Hinek A, Minamisawa S, et al.
Fibulin-5/DANCE is essential for elastogenesis in vivo. Nature 2002;415:
171–5.
19. Trachootham D, Alexandre J, Huang P. Targeting cancer cells by ROSmediated mechanisms: a radical therapeutic approach? Nat Rev Drug
Discov 2009;8:579–91.
20. Dineen SP, Roland CL, Greer R, Carbon JG, Toombs JE, Gupta P, et al. Smac
mimetic increases chemotherapy response and improves survival in mice
with pancreatic cancer. Cancer Res 2010;70:2852–61.
21. Ostapoff KT, Awasthi N, Cenik BK, Hinz S, Dredge K, Schwarz RE, et al.
PG545, an angiogenesis and heparanase inhibitor, reduces primary tumor
growth and metastasis in experimental pancreatic cancer. Mol Cancer Ther
2013;12:1190–201.
Cancer Research
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Fbln5 Reduces ROS in Pancreatic Cancer
22. Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH,
et al. Trp53R172H and KrasG12D cooperate to promote chromosomal
instability and widely metastatic pancreatic ductal adenocarcinoma in
mice. Cancer Cell 2005;7:469–83.
23. Hallmann R, Mayer DN, Berg EL, Broermann R, Butcher EC. Novel mouse
endothelial cell surface marker is suppressed during differentiation of the
blood–brain barrier. Dev Dyn 1995;202:325–32.
24. Ye R, Jung DY, Jun JY, Li J, Luo S, Ko HJ, et al. Grp78 heterozygosity
promotes adaptive unfolded protein response and attenuates diet-induced
obesity and insulin resistance. Diabetes 2010;59:6–16.
25. Rivera LB, Brekken RA. SPARC promotes pericyte recruitment via inhibition of endoglin-dependent TGF-beta1 activity. J Cell Biol 2011;193:
1305–19.
26. Pink JJ, Planchon SM, Tagliarino C, Varnes ME, Siegel D, Boothman DA.
NAD(P)H:Quinone oxidoreductase activity is the principal determinant of
beta-lapachone cytotoxicity. J Biol Chem 2000;275:5416–24.
27. Fitzsimmons SA, Workman P, Grever M, Paull K, Camalier R, Lewis AD.
Reductase enzyme expression across the National Cancer Institute Tumor
cell line panel: correlation with sensitivity to mitomycin C and EO9. J Natl
Cancer Inst 1996;88:259–69.
28. Kawaguchi Y, Cooper B, Gannon M, Ray M, MacDonald RJ, Wright CV. The
role of the transcriptional regulator Ptf1a in converting intestinal to
pancreatic progenitors. Nat Genet 2002;32:128–34.
29. Aguirre AJ, Bardeesy N, Sinha M, Lopez L, Tuveson DA, Horner J, et al.
Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic
pancreatic ductal adenocarcinoma. Genes Dev 2003;17:3112–26.
30. Mah LJ, El-Osta A, Karagiannis TC. gammaH2AX: a sensitive molecular
marker of DNA damage and repair. Leukemia 2010;24:679–86.
31. Sullivan KM, Bissonnette R, Yanagisawa H, Hussain SN, Davis EC. Fibulin5 functions as an endogenous angiogenesis inhibitor. Lab Invest 2007;87:
818–27.
32. Siegel D, Gustafson DL, Dehn DL, Han JY, Boonchoong P, Berliner LJ, et al.
NAD(P)H:quinone oxidoreductase 1: role as a superoxide scavenger. Mol
Pharmacol 2004;65:1238–47.
33. Lee JM, Calkins MJ, Chan K, Kan YW, Johnson JA. Identification of the NFE2-related factor-2-dependent genes conferring protection against oxidative stress in primary cortical astrocytes using oligonucleotide microarray
analysis. J Biol Chem 2003;278:12029–38.
34. Lee JJ, Perera RM, Wang H, Wu D-C, Liu XS, Han S, et al. Stromal response to
Hedgehog signaling restrains pancreatic cancer progression. Proc Natl Acad
Sci U S A 2014;111:E3091–E100.
35. Rhim Andrew D, Oberstein Paul E, Thomas Dafydd H, Mirek Emily T,
Palermo Carmine F, Sastra Stephen A, et al. Stromal elements act to restrain,
rather than support, pancreatic ductal adenocarcinoma. Cancer Cell
2014;25:735–47.
36. Provenzano Paolo P, Cuevas C, Chang Amy E, Goel Vikas K, Von Hoff
Daniel D, Hingorani Sunil R. Enzymatic targeting of the stroma ablates
physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer
Cell 2012;21:418–29.
www.aacrjournals.org
37. Ostapoff KT, Kutluk Cenik B, Wang M, Ye R, Xu X, Nugent D, et al.
Neutralizing murine TGFbR2 promotes a differentiated tumor cell
phenotype and inhibits pancreatic cancer metastasis. Cancer Res 2014;
74:1–12.
38. Yue W, Sun Q, Landreneau R, Wu C, Siegfried JM, Yu J, et al. Fibulin-5
suppresses lung cancer invasion by inhibiting matrix metalloproteinase-7
expression. Cancer Res 2009;69:6339–46.
39. Lee YH, Albig AR, Regner M, Schiemann BJ, Schiemann WP. Fibulin-5
initiates epithelial-mesenchymal transition (EMT) and enhances EMT
induced by TGF-beta in mammary epithelial cells via a MMP-dependent
mechanism. Carcinogenesis 2008;29:2243–51.
40. Hu Z, Ai Q, Xu H, Ma X, Li HZ, Shi TP, et al. Fibulin-5 is downregulated in
urothelial carcinoma of bladder and inhibits growth and invasion of
human bladder cancer cell line 5637. Urol Oncol 2011;29:430–5.
41. Wang Q, Li XG, Zhang Y, Cao LQ, Deng ZH, Chen Y. [Expression of EVEC in
ovarian carcinoma and its biological significance]. Zhonghua Zhong Liu Za
Zhi 2010;32:676–80.
42. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D,
Honess D, et al. Inhibition of Hedgehog signaling enhances delivery
of chemotherapy in a mouse model of pancreatic cancer. Science
2009;324:1457–61.
43. Singh M, Ferrara N. Modeling and predicting clinical efficacy for drugs
targeting the tumor milieu. Nat Biotechnol 2012;30:648–57.
44. Olive KP, Tuveson DA. The use of targeted mouse models for preclinical
testing of novel cancer therapeutics. Clin Cancer Res 2006;12:5277–87.
45. Ansari D, Aronsson L, Sasor A, Welinder C, Rezeli M, Marko-Varga G,
et al. The role of quantitative mass spectrometry in the discovery of
pancreatic cancer biomarkers for translational science. J Transl Med
2014;12:87.
46. Jagadeeshan S, Krishnamoorthy YR, Singhal M, Subramanian A, Mavuluri
J, Lakshmi A, et al. Transcriptional regulation of fibronectin by p21activated kinase-1 modulates pancreatic tumorigenesis. Oncogene
2015;34:455–64.
47. Hirai M, Ohbayashi T, Horiguchi M, Okawa K, Hagiwara A, Chien KR, et al.
Fibulin-5/DANCE has an elastogenic organizer activity that is abrogated by
proteolytic cleavage in vivo. J Cell Biol 2007;176:1061–71.
48. Kardeh S, Ashkani-Esfahani S, Alizadeh AM. Paradoxical action of reactive
oxygen species in creation and therapy of cancer. Eur J Pharmacol
2014;735C:150–68.
49. Awadallah NS, Dehn D, Shah RJ, Russell Nash S, Chen YK, Ross D, et al.
NQO1 expression in pancreatic cancer and its potential use as a biomarker.
Appl Immunohistochem Mol Morphol 2008;16:24–31.
50. DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A, Wei C, Frese K, et al.
Oncogene-induced Nrf2 transcription promotes ROS detoxification and
tumorigenesis. Nature 2011;475:106–9.
51. Kietadisorn R, Juni RP, Moens AL. Tackling endothelial dysfunction by
modulating NOS uncoupling: new insights into its pathogenesis and
therapeutic possibilities. Am J Physiol Endocrinol Metab 2012;302:
E481–95.
Cancer Res; 75(23) December 1, 2015
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.
5069
Published OnlineFirst November 17, 2015; DOI: 10.1158/0008-5472.CAN-15-0744
Fibulin-5 Blocks Microenvironmental ROS in Pancreatic Cancer
Miao Wang, Mary Topalovski, Jason E. Toombs, et al.
Cancer Res 2015;75:5058-5069. Published OnlineFirst November 17, 2015.
Updated version
Supplementary
Material
Cited articles
Citing articles
E-mail alerts
Reprints and
Subscriptions
Permissions
Access the most recent version of this article at:
doi:10.1158/0008-5472.CAN-15-0744
Access the most recent supplemental material at:
http://cancerres.aacrjournals.org/content/suppl/2015/12/10/0008-5472.CAN-15-0744.DC1
This article cites 51 articles, 22 of which you can access for free at:
http://cancerres.aacrjournals.org/content/75/23/5058.full#ref-list-1
This article has been cited by 3 HighWire-hosted articles. Access the articles at:
http://cancerres.aacrjournals.org/content/75/23/5058.full#related-urls
Sign up to receive free email-alerts related to this article or journal.
To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at
[email protected].
To request permission to re-use all or part of this article, use this link
http://cancerres.aacrjournals.org/content/75/23/5058.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
Rightslink site.
Downloaded from cancerres.aacrjournals.org on June 1, 2020. © 2015 American Association for Cancer Research.