You are on page 1of 11

Eur J Oral Sci 2017; 1–11 Ó 2017 Eur J Oral Sci

DOI: 10.1111/eos.12356 European Journal of


Printed in Singapore. All rights reserved
Oral Sciences

E. E. Hinsley1, C. E. de Oliveira2,
Angiotensin 1-7 inhibits angiotensin S. Hunt1, R. D. Coletta2,
D. W. Lambert1
II-stimulated head and neck cancer 1
Academic Unit of Oral and Maxillofacial
Pathology, School of Clinical Dentistry,

progression University of Sheffield, Sheffield, UK;


2
Department of Oral Diagnosis, School of
Dentistry, University of Campinas, Piracicaba,
Brazil

Hinsley EE, de Oliveira CE, Hunt S, Coletta RD, Lambert DW. Angiotensin 1-7
inhibits angiotensin II-stimulated head and neck cancer progression.
Eur J Oral Sci 2017; 00: 1–11. © 2017 Eur J Oral Sci
Angiotensin II (Ang II) is the product of the proteolytic action of angiotensin-con-
verting enzyme (ACE) on the precursor peptide, angiotensin I (Ang I). In addition
to its vasoactive properties, Ang II is able to stimulate angiogenesis and act as a
mitogen, promoting cellular proliferation. Recently, evidence has emerged that Ang
II is also able to promote tumour invasion, a key step in the metastatic cascade,
although the mechanisms by which it does so remain largely obscure. Here we show Daniel W. Lambert, School of Clinical
Dentistry, University of Sheffield, Sheffield
that Ang II is able to promote the invasion and migration of head and neck squa-
S10 2TA, UK
mous cell carcinoma (HNSCC) cells both in an autocrine manner and by triggering
stromal tumour–paracrine interactions. The effects of Ang II on autocrine and para- E-mail: d.w.lambert@sheffield.ac.uk
crine signalling pathways are mediated by angiotensin receptor 1 (AT1R) and inhib-
ited by angiotensin 1-7 (Ang 1-7), a peptide produced from Ang II by the action of Key words: a disintegrin and
angiotensin-converting enzyme 2 (ACE2). These data are the first to demonstrate a metalloproteinase; epidermal growth factor
role for the renin–angiotensin system in oral carcinogenesis and raise the possibility receptor; fibroblasts; head and neck
of utilizing AT1R receptor antagonists and/or Ang 1-7 as novel therapeutic agents squamous cell carcinoma; mas receptor
for HNSCC. Accepted for publication April 2017

Head and neck cancer, predominantly comprised of head (a-SMA) and matrix metalloproteinase-2 (MMP-2) (7).
and neck squamous cell carcinoma (HNSCC), is the Although fibroblasts are known to promote migration
sixth most common cancer worldwide. Although com- and invasion of HNSCC cells, little is known of the
monly associated with risk factors such as advanced age, factors that promote these interactions and direct
smoking, and alcohol consumption, the incidence of this remodelling of stromal fibroblasts. We have recently
cancer is increasing among younger and low-risk groups. shown that a small peptide, endothelin-1 (ET-1), is able
Head and neck squamous cell carcinoma has a poor to promote paracrine signalling between oral fibroblasts
prognosis, with surgery remaining the front-line thera- and HNSCC cells via epidermal growth factor receptor
peutic approach (1). Compared with other common can- (EGFR) transactivation (2).
cers, the molecular landscape of HNSCC is relatively Angiotensin II (Ang II), like ET-1, is a peptide
poorly understood, hampering the development of novel produced by the action of a metalloproteinase, in
therapeutic approaches. Recent studies have indicated this case angiotensin-converting enzyme (ACE).
that the tumour stroma plays a critical role in the pro- Angiotensin II is a key component of the renin–an-
gression of HNSCC (2–4), suggesting that factors within giotensin system (RAS) and exerts its actions on
the stroma may represent viable targets for a new genera- target cells by binding to one of two G protein–cou-
tion of therapeutic agents. pled receptors, angiotensin receptor 1 (AT1R) and
The tumour stroma is a complex milieu of extracellu- the less well-characterized angiotensin receptor 2
lar matrix components and a variety of cell types, of (AT2R) (8). It is becoming increasingly evident that
which fibroblasts are the most numerous (5). We have in addition to its systemic effects on blood pressure
recently demonstrated that fibroblast-derived factors and fluid homeostasis, Ang II is able to exert effects
are able to invoke phenotypic changes in adjacent at the local tissue level (9). A functional RAS has
epithelial cells, promoting tumour progression (6). In been detected in a number of tissues, including the
turn, factors in the stromal environment, including oral mucosa (10).
those generated by cancer cells and by fibroblasts them- Binding of Ang II to AT1R triggers receptor phospho-
selves, promote phenotypic changes in the fibroblasts rylation, activating a variety of intracellular and extracel-
(4). These include the acquisition of a more migratory, lular events, including arrestin recruitment (11) and
contractile phenotype and increased expression of EGFR transactivation (12). Angiotensin II-mediated
myofibroblast markers, such as a-smooth muscle actin EGFR transactivation, a result of AT1R-induced
2 Hinsley et al.

activation of a disintegrin and metalloproteinase consent and South Sheffield Ethics Committee approval,
(ADAM) proteinase-mediated EGFR ligand release, as described previously (25) and used within passages 3–
leading to autocrine or paracrine activation of EGFR 10] were routinely cultured in DMEM supplemented with
signalling, has been demonstrated to promote migration 2 mM L-glutamine and 10% (v/v) FBS. Normal oral ker-
of dermal keratinocytes and fibroblasts (13). Increased atinocytes (NOKs) [collected with informed consent and
South Sheffield Ethics Committee approval, as described
AT1R expression has been identified in a number of previously (26), and used up to passage 3] and D19 and
tumours, including ovarian (14), breast (15), and bladder D20 [derived from oral dysplasia lesions, described previ-
(16). Furthermore, Ang II is known to promote tumour ously (27)], B16 [derived from an HNSCC primary tumour
growth (17) and angiogenesis (18), and AT1R blockers and a cervical lymph node metastasis, respectively (28)],
and ACE inhibitors have been demonstrated to reduce and B22 cell lines were all routinely cultured in Green’s
growth and vascularization of a wide range of tumours, medium consisting of a 3:1 ratio of DMEM and Ham’s
including those of the lung, prostate, and oral cavity, F12 supplemented with 2 mM L-glutamine, 180 lM ade-
suggesting a role for Ang II in cancer pathogenesis [re- nine, 5 lg ml-1 of transferrin, 0.2 lM 3,30 ,5-triiodo-L-thyro-
viewed in (19)]. nine, 4 lg ml-1 of hydrocortisone, 10 ng ml-1 of epidermal
The discovery in 2000 of a homologue of ACE, termed growth factor, and 10% (v/v) FBS. All cells were grown in
antibiotic-free medium at 37°C and 5% (v/v) CO2. Oral
ACE2, revealed a new paradigm in angiotensin peptide fibroblasts were transfected at approximately 60% conflu-
biology (20). Angiotensin-converting enzyme 2 prote- ence using Oligofectamine reagent (Fisher Scientific,
olytically cleaves Ang II to produce angiotensin 1-7 (Ang Loughborough, UK) with small interfering RNA (siRNA)
1-7), a peptide that also binds to a G protein–coupled to ADAM17 or a negative-control non-targeting oligonu-
receptor, MasR (9). The cellular consequences of MasR cleotide (final concentration 50 nM; Fisher Scientific) in
activation remain unclear but it is apparent that Ang 1-7 Opti-MEM media (Fisher Scientific). Cells were incubated
appears able to counteract the effects of Ang II (8). for 24 h, after which media and cells were collected for
Recently, it was reported that administration of Ang 1-7 further experimentation and/or analysis.
reduces lung cancer growth (21) and invasion (22) and
breast cancer-associated fibrosis by inhibiting extracellu-
Cell treatments
lar signal-regulated kinase (ERK) activation (23).
Previous reports have reported a functional, tissue- Oral fibroblasts and HNSCC cells were serum starved for
level (as opposed to systemic) RAS in the oral mucosa 24 h before experimentation. For the preparation of con-
(10), and the ACE inhibitor perindopril has been ditioned media (CM), oral fibroblasts were treated with
shown to reduce the growth of head and neck carci- Ang II (100 nM; Sigma-Aldrich) and/or Ang 1-7 (100 nM;
Sigma-Aldrich) for 4 h at 37°C and 5% (v/v) CO2. Where
noma in vivo (24), suggesting a role for Ang II in dis-
indicated, fibroblasts were pretreated with telmisartan, an
ease aetiology. Here we examine, for the first time, the AT1R antagonist (1 lM; Sigma-Aldrich), A-779, a MasR
contribution of the local RAS to HNSCC pathogenesis. antagonist (1 lM; Sigma-Aldrich), or GM6001, an MMP
We identify aberrant expression of AT1R in HNSCC inhibitor (10 lM; Merck Millipore, Feltham, UK) for
cells and demonstrate that its ligand, Ang II, is able to 30 min before addition of and treatment with mitogenic
stimulate both HNSCC cells and oral fibroblasts to peptides for 4 h. Media was collected from fibroblasts
promote tumour cell chemotaxis and invasion. Further- after incubation, filtered, and added to the lower chamber
more, we demonstrate the ability of the Ang II metabo- of invasion or migration assay plates, before cells were
lite, Ang 1-7, to block the protumorigenic effects of seeded, as described, into the upper chamber. For prepara-
Ang II. Taken together, these findings suggest that tion of oral fibroblasts for immunoblotting, cells were trea-
ted with either Ang II (100 nM) or transforming growth
AT1R blockade and/or Ang 1-7 administration, may be
factor beta (TGF-b; 20 ng ml-1) for 48 h at 37°C and 5%
a novel therapeutic opportunity in HNSCC. (v/v) CO2.

RNA isolation and quantitative PCR


Material and methods
The RNeasy mini kit (Qiagen, Manchester, UK) was used
Materials to extract total RNA from all cell lines, and from primary
NOKs and oral fibroblasts, according to the manufac-
Dulbecco’s modified Eagle’s medium (DMEM), Roswell turer’s instructions. RNA from each sample was quantified
Park Memorial Institute medium (RPMI-1460), Ham’s using a NanoDrop spectrophotometer (Fisher Scientific).
F12 nutrient mix, fetal bovine serum (FBS), Dulbecco’s A High Capacity cDNA Reverse Transcription kit (Fisher
PBS, L-glutamine, adenine, transferrin, 3,30 ,5-triiodo-L- Scientific) was used for synthesis of cDNA according to
thyronine, hydrocortisone, epidermal growth factor and the manufacturer’s instructions. cDNA was subsequently
trypsin/ethylenediaminetetraacetic acid were all purchased analysed by SYBR Green or TaqMan quantitative PCR
from BioWhittaker (Wokingham, UK). Reagents were (qPCR). Primers used for SYBR Green quantification
purchased from Sigma-Aldrich (Poole, UK), unless other- were as follows: U6 forward, 50 -CTCGCTTCGGCAGCA
wise stated, and were of the highest analytical grade. CA-30 and U6 reverse, 50 -AACGTTCACGAATTTGCGT-
30 ; AT1R forward, 50 -ACCTGGCTATTGTTCACCCAA-30
Routine cell culture and transfection and AT1R reverse, 50 -ACAAGCATTGTGCGTCGAAG-30 ;
AT2R forward, 50 -GTTCCCCTTGTTTGGTGTAT-30 and
The HNSCC-derived cell lines SCC4, H357, and Cal27, AT2R reverse, 50 -CATCTTCAGGACTTGGTCAC-30 ;
and primary oral fibroblasts [collected with informed MasR forward, 50 -TTCATCCCGTTCAGAAGACA-30
Angiotensin peptides in head and neck cancer 3

and MasR reverse, 50 -CCAATGGCAAGCAGAAATA Immunoblotting


GA-30 ; a-SMA forward, 50 -GAAGAAGAGGACAGCAC
TG-30 and a-SMA reverse, 50 -TCCCATTCCCACCATC Cells were trypsinized, washed with PBS, and pelleted.
AC-30 . TaqMan probes for ACE, ACE2, and b2mi- Protein was extracted using radioimmunoprecipitation
croglobulin were obtained from Fisher Scientific. All val- assay (RIPA) lysis buffer (Merck Millipore, Feltham, UK)
ues were normalized to U6 (SYBR) or b2microglobulin containing Complete Mini Protease Inhibitor Cocktail
(TaqMan) expression levels. Where indicated, amplicons (Roche, Welwyn Garden City, UK; used according to the
were also analysed by agarose gel electrophoresis and manufacturer’s instructions) and Benzonase (Sigma-
visualized with ethidium bromide under ultraviolet (UV) Aldrich; used according to the manufacturer’s instruc-
transillumination. tions). Protein concentration was measured using the BCA
Protein Assay Kit (Fisher Scientific) with BSA as a
standard. Total protein extracts (50 lg) were separated by
Cell migration and invasion assays 3-8% (v/v) acrylamide SDS-PAGE and transferred to
nitrocellulose membrane. Following blocking of non-specific
SCC4 or H357 cells at 90% confluence were serum starved protein binding in 5% (w/v) dried milk and 3% (w/v) BSA in
for 24 h before experimentation. Then the cells were tryp- Tris-buffered saline containing 0.5% (v/v) Tween 20, mem-
sinized and resuspended in DMEM, containing 0.1% (w/ branes were incubated with antibodies directed to a-SMA
v) BSA, at 5 9 105 cells ml-1. Supplements including Ang (1:1,000; Sigma-Aldrich) or b-actin (1:4,000; Sigma-Aldrich).
II (100 nM) and/or Ang 1-7 (100 nM) were added and Horseradish peroxidase-conjugated secondary antibodies
200 ll of cell suspension was added to the top of the tran- (Sigma) were diluted 1:2,000. All antibodies were diluted in
swell chamber. Then, 500 ll of DMEM containing 10% 5% (w/v) dried milk and 3% (w/v) BSA in Tris-buffered
(v/v) FCS or CM from fibroblasts was added to the lower saline containing 0.5% (v/v) Tween 20. Immunoreactive
chamber (as indicated in individual figure legends). The bands were visualized using enhanced chemiluminescence
EGFR antagonist, AG 1478 (125 nM; Sigma-Aldrich), (ECL) (Fisher Scientific). Densitometry was performed using
was incubated with the SCC4 cell suspension for 30 min Adobe Photoshop (San Jose, CA, USA).
at 37°C and 5% (v/v) CO2 before the addition of the
SCC4 cell suspension to the chamber. After 24 h (migra-
tion) or 48 h (invasion), cells were swabbed away from the Statistical analyses
inside of the transwell chamber and cells adhering to the
underside of the chamber were fixed for 10 min in 100% Data are expressed as mean  standard error of the mean. Sta-
(v/v) methanol. Migrated cells were stained with 0.1% (w/ tistical analyses, where appropriate, were made between two
v) crystal violet and counted by light microscopy at 40x groups using the Student’s t-test or Mann–Whitney U-test. A
magnification. Three fields of view from each insert were value of P < 0.05 was considered statistically significant.
counted. For the scratch assay, oral fibroblasts were
seeded in a six-well plate (100,000 cells/well) and allowed
to adhere overnight. Cells were serum starved for 24 h
and a scratch was made in each well using a 200-ll pipette
Results
tip. Media was removed, and cells were washed in PBS Ang II stimulates migration and invasion of HNSCC
before the addition of peptides or inhibitors. Mitomycin C cells via AT1R
(Sigma-Aldrich), at 1 lg ml-1, was added to each well to
prevent proliferation. Each well was photographed at two We first examined the expression of AT1R and AT2R in
points along the scratch at 0 and 24 h. The distance a panel of primary human NOKs and fibroblasts, and in
between each edge of the scratch was measured. cell lines derived from oral dysplasia lesions, primary
Invasion assays were prepared using Matrigel (growth HNSCC, and a local metastasis. Expression of AT1R
factor depleted; Sigma-Aldrich) diluted 1/45 (v/v) with was found to be markedly elevated in a number of pri-
DMEM. In these assays, 100 ll of diluted Matrigel was mary carcinoma cell lines (Fig. 1A) compared with nor-
added to the top of each transwell chamber. The inserts mal, dysplastic, and metastatic cells. High levels of AT1R
were left overnight at 37°C and used the following day.
transcript were also detected in normal oral fibroblasts
(NOFs; Fig 1A). Angiotensin receptor 2 was detected in
Gel contraction assay all cell lines and NOKs examined, with no significant dif-
ferences in AT2R expression observed between cells from
Oral fibroblasts (250,000 cells/well) were resuspended in
normal and diseased tissue (Fig. 1B). Angiotensin recep-
DMEM containing 10% (v/v) FBS and were mixed with
rat tail collagen (7.5 mg ml-1 in 10x DMEM). The pH of tor 2 was undetectable in NOFs using this methodology.
the cell + collagen mixture was adjusted to pH 7 using Having established the expression of AT1R and AT2R in
0.1 M sodium hydroxide (NaOH). Then, 300 ll of the HNSCC cells, we next examined the effect of exogenous
cell + collagen mixture was added to 24-well plates. The Ang II on HNSCC cell motility. Two HNSCC cell lines,
cell + collagen mixture was left to incubate for 45 min H357 and SCC4 (both derived from squamous cell carci-
before the addition of 500 ll of DMEM containing 10% noma of the tongue), were selected on the basis of their
(v/v) FBS. This medium was removed after 4 h and high levels of AT1R expression. The ability of both cell
replaced with 500 ll of serum-free medium and matrices lines to migrate and invade was significantly increased
were incubated overnight at 37°C and 5% (v/v) CO2. following treatment with Ang II (Fig. 1C–F). The stimu-
Using a scalpel, the gels were carefully loosened from the
lation of migration was completely abrogated by the
edges of the well and treated with serum-free medium con-
taining thrombin (0.5 U ml-1) or Ang II (100 nM). Colla- prior application of a specific AT1R antagonist,
gen lattices were photographed and the distance telmisartan, but not by the AT2R antagonist, P123319
contracted by the gels was measured. (Fig. 1G,H).
4 Hinsley et al.

Fig. 1. Angiotensin receptor 1 (AT1R) mediates angiotensin II (Ang II)-stimulated migration and invasion of head and neck squa-
mous cell carcinoma (HNSCC). Total RNA was prepared from normal primary oral keratinocytes (NOKs), normal primary oral
fibroblasts (NOFs), and from a range of cell lines derived from oral dysplasias, squamous cell carcinomas, and a local lymph
node metastasis. Expression of AT1R (A) and AT2R (B) was assessed by quantitative PCR (qPCR). Migration (C) and invasion
(E) of SCC4 and migration (D) and invasion (F) of H357, in response to treatment with Ang II, were analysed as described in
the Material and methods. The effect of telmisartan, an AT1R antagonist, and PD123319, an AT2R antagonist, on migration of
SCC4 cells in response to Ang II was assessed (G and H, respectively). The results are plotted as mean values relative to
untreated control wells  standard error of the mean, of three separate fields of view from three independent experiments.
*P < 0.05 relative to control. ns, not significant.

Fibroblasts potentiate the effect of Ang II on HNSCC


signalling between fibroblasts and cancer cells. Treat-
motility
ment of SCC4 and H357 cells with CM collected from
Given the high levels of AT1R detected in NOFs, we NOFs incubated with Ang II significantly increased
next sought to determine whether interaction of Ang II migration and invasion (Fig. 2A–D) to a degree
with NOFs could trigger promigratory paracrine greater than that observed in the absence of such CM
Angiotensin peptides in head and neck cancer 5

Fig. 2. Fibroblasts potentiate the effects of angiotensin II (Ang II) on head and neck squamous cell carcinoma (HNSCC) migra-
tion and invasion. Conditioned media (CM) was collected and filtered from fibroblasts treated with Ang II or PBS for 4 h, before
being added to the lower chamber of a transwell plate, as described in the Material and methods. Migration (A) and invasion (C)
of SCC4 cells, and migration (B) and invasion (D) of H357 cells, were then assessed. The ability of telmisartan to block the
observed migration of SCC4 cells was also analysed (E). The results are plotted as mean values relative to untreated control wells
 standard error of the mean, of three separate fields of view from three independent experiments. *P < 0.05 relative to control,
**P < 0.05 relative to Ang II treated.

Ang II invokes myofibroblast transdifferentiation of


(~three-fold increase in the presence of CM vs. 1.5-fold
NOFs
increase in the absence of CM, for SCC4). Pretreatment
of fibroblasts with telmisartan blocked the stimulation As myofibroblast transdifferentiation has previously
of migration and invasion (Fig. 2E), as expected given been shown to promote HNSCC cell motility, we next
the absence of AT2R in these cells. Pretreatment of analysed whether Ang II treatment of NOFs resulted in
fibroblasts with GM6001 blocked the Ang II-mediated the acquisition of an activated, myofibroblast-like phe-
stimulation of migration, implicating the ADAM- notype. Treatment with TGF-b1 was used in these
mediated release of a soluble factor (Fig. 3A,B). The experiments as a positive control as TGF-b1 is widely
ability of AG 1478 treatment of SCC4 cells to block reported to induce myofibroblast differentiation in vitro
the stimulatory effect of CM collected from Ang II- (29). qRT-PCR analysis of RNA extracted from NOFs
treated fibroblasts implicates the involvement of EGFR treated with Ang II revealed a significant increase in
signalling (Fig. 3C). We have previously shown a role the expression of a-SMA, a widely utilized marker of
for ADAM17, a membrane-anchored protease responsi- myofibroblasts (Fig. 4A). A significant increase was
ble for the proteolytic activation of a number of physi- also observed in the expression of MMP-2, previously
ologically significant substrates, in stromal-epithelial reported to be up-regulated in myofibroblasts (Fig. 4B).
signalling in HNSCC (2). We therefore examined the Immunoblotting also indicated a small increase in
effect of abrogating ADAM17 expression on the non- a-SMA protein (Fig. 4C). The stimulation of a-SMA
cell autonomous actions of Ang II; siRNA-mediated expression by Ang II was completely blocked by prior
knockdown of ADAM17 in fibroblasts blocked the application of telmisartan, suggesting the involvement
Ang II-mediated increase in SCC4 migration on indi- of AT1R (Fig. 4D). Collagen contraction assays indi-
rect co-culture (Fig. 3D). A significant reduction in cated the acquisition of a more contractile phenotype
ADAM17 transcript confirmed the efficacy of the upon Ang II treatment, of a similar magnitude to that
siRNA-mediated knockdown (Fig. 3E). induced by thrombin, a known stimulator of fibroblast
6 Hinsley et al.

Fig. 3. Angiotensin II (Ang II) stimulates head and neck squamous cell carcinoma (HNSCC) migration via epidermal growth factor
receptor (EGFR) transactivation. Angiotensin II is known to induce phenotypic changes by transactivating EGFR signalling by a mech-
anism involving a disintegrin and metalloproteinase (ADAM)-mediated proteolysis of EGFR ligands (A). The effect of inhibiting both
ADAM proteolysis, by treating normal oral fibroblasts (NOFs) with the broad-spectrum ADAM/matrix metalloproteinase (MMP) inhi-
bitor, GM6001 (A, B), and EGFR signalling in SCC4 cells, using the EGFR antagonist, AG1478 (A, C), was assessed using migration
assays, as described in the Material and methods. Small interfering RNA (siRNA) targeted to siRNA (siA17) or a non-targeting negative
control oligonucleotide (-ve) were transiently transfected into NOFs treated with Ang II or left untreated, and the conditioned medium
was used in a migration assay to assess chemotaxis of SCC4 cells, as described in the Materials and methods (D). Results are plotted rela-
tive to untreated control wells  standard error of the mean of three separate fields of view from three independent experiments. Total
RNA was extracted from transfected NOFs and subjected to quantitative RT-PCR using probes for ADAM17 and B2M (as a reference
gene) to assess siRNA-mediated knockdown (E; n = 3). *P < 0.05 relative to control, **P < 0.05 relative to Ang II treated.

Ang 1-7 inhibits Ang II-induced cancer cell motility


contraction (Fig. 4E). Angiotensin II-treated NOFs
were also observed to be more migratory than their Angiotensin 1-7, the product of the proteolytic action
vehicle-treated counterparts (Fig. 4F). of ACE2 on Ang II, is reported to oppose the
Angiotensin peptides in head and neck cancer 7

Fig. 4. Angiotensin II (Ang II) provokes myofibroblast transdifferentiation of normal oral fibroblasts (NOFs). NOFs were treated
with Ang II or transforming growth factor beta (TGF-b) for 48 h before cells were harvested and RNA extracted or lysates pre-
pared as described in the Material and methods. Alpha smooth muscle actin (a-SMA) (A) and matrix metalloproteinase 2 (MMP-
2) (B) transcript levels were assessed by quantitative PCR (qPCR); values are shown normalized to U6 expression levels in the
same samples. Cell lysates were separated by SDS-PAGE and immunoblotted for a-SMA and b-actin (as a loading control). A
representative blot is shown (C) and the intensity of the band corresponding to a-SMA, determined by densitometry and normal-
ized to b-actin levels in the same sample, is indicated under each lane. qPCR for a-SMA was also carried out on RNA extracted
from NOFs pretreated with telmisartan before addition of Ang II for 48 h (D). Results are plotted relative to untreated control
wells, as the mean value  standard error of the mean of three independent experiments, carried out in triplicate. *P < 0.05. Oral
fibroblasts were seeded into collagen as described in the Material and methods. Following overnight incubation in serum-free
medium, NOFs were treated with Ang II or thrombin for 30 min. The gels were photographed and the distance contracted was
measured (E). A scratch was made in oral fibroblasts cultured as a monolayer. Cells were treated with Ang II or TGF-b, and the
diameter of the scratch was measured before treatment and after 24 h. Photographs were taken (F) and percentage migration rela-
tive to control was calculated. The data represent the mean value of at least three experiments  standard error of the mean.
*P < 0.05.

profibrotic actions of Ang II in animal models of renal invasion, both in the presence and the absence of Ang
and cardiovascular disease, acting through its receptor, II. Co-application of Ang 1-7 with Ang II significantly
MasR. Therefore, we next analysed the expression of abrogated the ability of Ang II to promote cell motility
ACE2 and MasR in our panel of cells to determine and invasion directly (Fig. 5C,D).
whether the oral mucosa may be capable of generating Given that detectable levels of MasR transcript were
and/or responding to Ang 1-7. The highest ACE2 levels observed in NOFs, albeit low compared with HNSCC
were observed in NOKs, with lower levels observed in cells (Fig. 5B, inset), we next analysed the effect of
a number of HNSCC cell lines, and ACE2 was unde- exogenous Ang 1-7 on Ang II-stimulated paracrine sig-
tectable in NOFs (Fig. 5A). MasR was readily detect- nalling between NOFs and HNSCC cells. As observed
able in HNSCC-derived cells, with lower levels present for direct application, treatment of SCC4 cells with
in NOFs and NOKs (Fig. 5B). Having established that CM from fibroblasts incubated with Ang 1-7 and Ang
HNSCC cells express both ACE2 and MasR, we next II significantly inhibited the stimulation of cell migra-
examined the effect of Ang 1-7 on their motility and tion and invasion observed in response to Ang II alone
8 Hinsley et al.

Fig. 5. Angiotensin 1-7 (Ang 1-7) abrogates angiotensin II (Ang II)-stimulated migration and invasion of head and neck squa-
mous cell carcinoma (HNSCC) cells. Total RNA was prepared from normal primary oral keratinocytes (NOKs), normal primary
oral fibroblasts (NOFs), and from a range of cell lines derived from oral dysplasias, squamous cell carcinomas, and a local lymph
node metastasis. Expression of angiotensin-converting enzyme 2 (ACE2) (A) and a G protein–coupled receptor, MasR (B), was
assessed by quantitative PCR (qPCR). Migration (C) and invasion (D) of SCC4 in response to treatment with Ang II and Ang 1-
7 was analysed as described in the Material and methods. The results are plotted as mean values relative to untreated control
wells  standard error of the mean, of three separate fields of view from three independent experiments. *P < 0.05 relative to
control; **P < 0.05 relative to Ang II treated. ns, not significant.

(Fig. 6A,B). Angiotensin 1-7 was also able to block the (the most common form of HNSCC) and observed a
Ang II-stimulated increase in a-SMA expression in marked up-regulation of AT1R expression in all the
NOFs, suggesting that Ang 1-7 is able inhibit the HNSCC cell lines examined compared with NOKs,
fibroblast-specific actions of Ang II (Fig. 6C) and that cells propagated from dysplastic lesions, and those
MasR is functional in these cells. from a metastatic lesion. Alterations in AT1R expres-
sion have been detected in other malignancies,
including prostate and breast (15, 33); in the latter,
meta-analysis of a large number of gene-expression pro-
Discussion filing data sets recently identified AT1R as one of the
Evidence supporting a role for the RAS in carcinogene- most prominently overexpressed genes. This is the first
sis and tumour progression has been mounting in report, however, of aberrant AT1R expression in
recent years and is currently the focus of considerable HNSCC and suggests that malignant cells may become
interest (19). Changes in the levels of components of sensitive to the mitogenic effects of local Ang II. This is
the RAS have been observed in a number of tumour given credence by the finding that treatment of two
types, including lung and prostate cancer, and inhibi- HNSCC-derived cell lines with Ang II resulted in a
tion of various elements of the pathway has been modest, but significant, increase in motility and inva-
shown to reduce tumour growth and metastasis [re- sive capacity; the ability of telmisartan, but not the
viewed previously (30)]. Here we have examined, for AT2R antagonist, PD123319, to inhibit this provides
the first time, the role of the RAS in head and neck further evidence that Ang II mediates these effects via
cancer progression with the aim of identifying novel AT1R.
therapeutic targets for this frequently fatal disease. In the current study, high levels of AT1R were also
A recent report identified the presence of all the com- detected in NOFs; CM collected from NOFs treated
ponents required for a functional local tissue RAS in with Ang II was able to stimulate the motility of oral
the human oral mucosa (10), in keeping with previous cancer cells, suggesting that AT1R in oral fibroblasts is
reports in other species (31, 32). In light of these find- functional and promotes stromal-tumour cell interac-
ings, we examined the levels of the two receptors for tions. The ability of an ADAM/MMP inhibitor and an
the major bioactive peptide generated by the RAS, Ang EGFR antagonist to block the promigratory effects of
II, in a panel of NOKs, NOFs, and cell lines derived CM from Ang II-treated oral fibroblasts indicates, as
from oral dysplasias and oral squamous cell carcinomas we recently reported for another mitogenic peptide,
Angiotensin peptides in head and neck cancer 9

Fig. 6. Angiotensin 1-7 (Ang 1-7) blocks stimulation of normal oral fibroblasts (NOFs) by angiotensin II (Ang II). NOFs were
pretreated for 30 min with Ang 1-7 before addition of Ang II or vehicle control for 4 h. Conditioned media (CM) was harvested
and applied to the lower chamber of migration (A, C) or matrigel invasion (B) assay plates, before addition of SCC4 cells to the
upper chamber. SCC4 migration and invasion was quantified as described in ‘Materials and methods’. Results are mean values
plotted relative to untreated control wells  standard error of the mean, of three separate fields of view from three independent
experiments. *P < 0.05 relative to control, **P < 0.05 relative to Ang II treated. Quantitative reverse transcription PCR (RT-
qPCR) for a-SMA was carried out on RNA extracted from NOFs pretreated with Ang 1-7 before addition of Ang II for 48 h;
values are expressed relative to U6 in the same samples (C).

ET-1 (2), that EGFR transactivation may play a role in demonstrate that TGF-b treatment of oral fibroblasts
the effects of Ang II on HNSCC motility. Epidermal provokes a myofibroblast transdifferentiation, which
growth factor receptor transactivation mediates patho- increases pro-invasive paracrine signalling (3). Here we
physiological signalling triggered by Ang II in other tis- show that Ang II provokes the acquisition of a myofi-
sues, such as aortic smooth muscle cells (34), and broblast phenotype in oral fibroblasts and increases
aberrant EGFR signalling is a frequently observed fea- their ability to stimulate cancer cell migration and inva-
ture of head and neck cancers (35). Further work is sion in a paracrine manner. Although the ability of
required to elucidate Ang II-induced signalling path- Ang II to stimulate myofibroblast transdifferentiation
ways in HNSCC. in pathologies, such as lung fibrosis, is known (36), this
Invasion and local metastatic spread of head and is, to our knowledge, the first report of Ang II-
neck cancer is increasingly recognized to involve com- mediated fibroblast remodelling influencing cancer cell
plex interactions between tumour cells and the sur- behaviour. The ability of an AT1R antagonist to block
rounding stroma. The most numerous cells of the the fibroblast phenotypic changes induced by Ang II,
stroma, fibroblasts, frequently adopt an activated, at least in vitro, raises the possibility of utilizing such
myofibroblastic phenotype in response to contextual antagonists in the clinic to inhibit metastasis and to
signals such as TGF-b in the tumour microenvironment reduce fibrosis to improve penetration of existing
(5). These cancer-associated fibroblasts (CAFs) have chemotherapeutic agents.
altered morphology resulting from the expression of Evidence is mounting in the literature for the possi-
a-SMA and secrete elevated levels of ECM compo- ble clinical applications of Ang 1-7, the product of
nents, such as MMP-2, which promote cancer cell inva- ACE2 action on Ang II, with beneficial effects of Ang
sion. Indeed, our unpublished data and a recent study 1-7 administration observed in a number of pathologies
10 Hinsley et al.

[reviewed previously (8)]. Angiotensin 1-7 has been of myofibroblast cell lines from oral squamous cell carcinoma.
shown to reduce lung cancer proliferation, angiogene- Oncol Rep 2011; 25: 1013–1020.
8. LAMBERT DW, HOOPER NM, TURNER AJ. Angiotensin-con-
sis, and invasion (21, 22); it is also reported to reduce verting enzyme 2 and new insights into the renin-angiotensin
fibrosis associated with breast cancers (23). In HNSCC system. Biochem Pharmacol 2008; 75: 781–786.
cells, we observed beneficial effects only in the presence 9. HAMMING I, COOPER ME, HAAGMANS BL, HOOPER NM, KOR-
of Ang II; no significant changes in cancer cell motility STANJE R, OSTERHAUS AD, TIMENS W, TURNER AJ, NAVIS G,
VAN GOOR H. The emerging role of ACE2 in physiology and
or fibroblast phenotype were observed in response to
disease. J Pathol 2007; 212: 1–11.
Ang 1-7 treatment alone. This is unsurprising given the 10. NAKAMURA T, HASEGAWA-NAKAMURA K, SAKODA K, MAT-
well-documented ability of Ang 1-7 to oppose the SUYAMA T, NOGUCHI K. Involvement of angiotensin II type 1
actions of Ang II, but is in apparent contradiction to receptors in interleukin-1beta-induced interleukin-6 produc-
previous findings in lung cancer cells (22), which tion in human gingival fibroblasts. Eur J Oral Sci 2011; 119:
345–351.
reported inhibition of invasion by Ang 1-7 in the 11. LEE MH, EL-SHEWY HM, LUTTRELL DK, LUTTRELL LM. Role
absence of Ang II, suggesting that the effects of Ang 1- of beta-arrestin-mediated desensitization and signaling in the
7 may be tissue specific. Our finding, that the response control of angiotensin AT1a receptor-stimulated transcrip-
to Ang 1-7 is dependent on the actions of Ang II, raises tion. J Biol Chem 2008; 283: 2088–2097.
12. BHOLA NE, GRANDIS JR. Crosstalk between G-protein-
the possibility that application of Ang 1-7 may be tar-
coupled receptors and epidermal growth factor receptor in
geted to those tumours (and associated CAFs) express- cancer. Front Biosci 2008; 13: 1857–1865.
ing AT1R and Mas. A recent phase I clinical trial in 13. YAHATA Y, SHIRAKATA Y, TOKUMARU S, YANG L, DAI X,
patients with solid tumours, including one with head TOHYAMA M, TSUDA T, SAYAMA K, IWAI M, HORIUCHI M,
and neck cancer, indicated that Ang 1-7 (delivered sub- HASHIMOTO K. A novel function of angiotensin II in skin
wound healing. Induction of fibroblast and keratinocyte
cutaneously) is well tolerated and with some clinical migration by angiotensin II via heparin-binding epidermal
benefit (37). Although further studies are required to growth factor (EGF)-like growth factor-mediated EGF
determine its efficacy, these data, together with the receptor transactivation. J Biol Chem 2006; 281: 13209–
in vitro evidence presented here, suggest that Ang 1-7 13216.
14. SUGANUMA T, INO K, SHIBATA K, KAJIYAMA H, NAGASAKA T,
and/or AT1R receptor antagonists are promising new
MIZUTANI S, KIKKAWA F. Functional expression of the angio-
therapeutic agents for HNSCC. tensin II type 1 receptor in human ovarian carcinoma cells
and its blockade therapy resulting in suppression of tumor
Acknowledgements – We thank Dr Craig Murdoch, Dr Helen Col- invasion, angiogenesis, and peritoneal dissemination. Clin
ley and Dr Keith Hunter (University of Sheffield, UK) for kindly Cancer Res 2005; 11: 2686–2694.
providing cells and Prof. Paula Farthing, Dr Simon Whawell 15. RHODES DR, ATEEQ B, CAO Q, TOMLINS SA, MEHRA R, LAX-
(University of Sheffield, UK) for helpful suggestions. Funding was MAN B, KALYANA-SUNDARAM S, LONIGRO RJ, HLEGESON BE,
obtained from British Society of Oral and Maxillofacial Patholo- BHOJANI MS, REHEMTULLA A, KLEER CG, HAYES DF, LUCAS
gists (to D.W.L.). E.E.H. was supported by a University of Shef- PC, VARAMBALLY S, CHINNAIYAN AM. AGTR1 overexpression
field PhD Scholarship. defines a subset of breast cancer and confers sensitivity to
losartan, an AGTR1 antagonist. Proc Natl Acad Sci U S A
Conflicts of interest – None declared. 2009; 106: 10284–10289.
16. TANAKA N, MIYAJIMA A, KOSAKA T, MIYAZAKI Y, SHIROTAKE
S, SHIRAKAWA H, KIKUCHI E, OYA M. Acquired platinum
resistance enhances tumour angiogenesis through angiotensin
References II type 1 receptor in bladder cancer. Br J Cancer 2011; 105:
1331–1337.
1. ARGIRIS A, KARAMOUZIS MV, RABEN D, FERRIS RL. Head 17. AMAYA K, OHTA T, KITAGAWA H, KAYAHARA M, TAKAMURA
and neck cancer. Lancet 2008; 371: 1695–1709. H, FUJIMURA T, NISHIMURA G, SHIMIZU K, MIWA K. Angio-
2. HINSLEY EE, HUNT S, HUNTER KD, WHAWELL SA, LAMBERT tensin II activates MAP kinase and NF-kappaB through
DW. Endothelin-1 stimulates motility of head and neck squa- angiotensin II type I receptor in human pancreatic cancer
mous carcinoma cells by promoting stromal-epithelial interac- cells. Int J Oncol 2004; 25: 849–856.
tions. Int J Cancer 2012; 130: 40–47. 18. FUJITA M, HAYASHI I, YAMASHINA S, FUKAMIZU A, ITOMAN
3. MARSH D, SUCHAK K, MOUTASIM KA, VALLATH S, HOPPER C, M, MAJIMA M. Angiotensin type 1a receptor signaling-depen-
JERJES W, UPILE T, KALAVREZOS N, VIOLETTE SM, WEINREB dent induction of vascular endothelial growth factor in
PH, CHESTER KA, CHANA JS, MARSHALL JF, HART IR, HACK- stroma is relevant to tumor-associated angiogenesis and
SHAW AK, PIPER K, THOMAS GJ. Stromal features are predic- tumor growth. Carcinogenesis 2005; 26: 271–279.
tive of disease mortality in oral cancer patients. J Pathol 19. GEORGE AJ, THOMAS WG, HANNAN RD. The renin-angioten-
2011; 223: 470–481. sin system and cancer: old dog, new tricks. Nat Rev Cancer
4. KELLERMANN MG, SOBRAL LM, DA SILVA SD, ZECCHIN KG, 2010; 10: 745–759.
GRANER E, LOPES MA, KOWALSKI LP, COLETTA RD. Mutual 20. LAMBERT DW, CLARKE NE, TURNER AJ. Not just angiotensi-
paracrine effects of oral squamous cell carcinoma cells and nases: new roles for the angiotensin-converting enzymes. Cell
normal oral fibroblasts: induction of fibroblast to myofibrob- Mol Life Sci 2010; 67: 89–98.
last transdifferentiation and modulation of tumor cell prolifer- 21. GALLAGHER PE, TALLANT EA. Inhibition of human lung can-
ation. Oral Oncol 2008; 44: 509–517. cer cell growth by angiotensin-(1–7). Carcinogenesis 2004; 25:
5. KALLURI R, ZEISBERG M. Fibroblasts in cancer. Nat Rev Can- 2045–2052.
cer 2006; 6: 392–401. 22. NI L, FENG Y, WAN H, MA Q, FAN L, QIAN Y, LI Q, XIANG
6. KABIR TD, LEIGH RJ, TASENA H, MELLONE M, COLETTA RD, Y, GAO B. Angiotensin-(1-7) inhibits the migration and inva-
PARKINSON EK, PRIME SS, THOMAS GJ, PATERSON IC, ZHOU sion of A549 human lung adenocarcinoma cells through inac-
D, MCCALL J, SPEIGHT PM, LAMBERT DW. A miR-335/COX- tivation of the PI3K/Akt and MAPK signaling pathways.
2/PTEN axis regulates the secretory phenotype of senescent Oncol Rep 2012; 27: 783–790.
cancer-associated fibroblasts. Aging 2016; 8: 1608–1635. 23. COOK KL, METHENY-BARLOW LJ, TALLANT EA, GALLAGHER
7. SOBRAL LM, ZECCHIN KG, NASCIMENTO DE, AQUINO S, LOPES PE. Angiotensin-(1–7) reduces fibrosis in orthotopic breast
MA, GRANER E, COLETTA RD. Isolation and characterization tumors. Cancer Res 2010; 70: 8319–8328.
Angiotensin peptides in head and neck cancer 11

24. YASUMATSU R, NAKASHIMA T, MASUDA M, ITO A, KURATOMI angiotensin II- and endothelin-1-stimulated proliferation by
Y, NAKAGAWA T, KOMUNE S. Effects of the angiotensin-I con- selective MEK inhibitor in cultured rabbit gingival fibrob-
verting enzyme inhibitor perindopril on tumor growth and lasts. Fundam Clin Pharmacol 2005; 19: 677–685.
angiogenesis in head and neck squamous cell carcinoma cells. 32. OHUCHI N, KOIKE K, SANO M, KUSAMA T, KIZAWA Y, HAYA-
J Cancer Res Clin Oncol 2004; 130: 567–573. SHI K, TANIGUCHI Y, OHSAWA M, IWAMOTO K, MURAKAMI H.
25. HEARNDEN V, LOMAS H, MACNEIL S, THORNHILL M, MURDOCH Proliferative effects of angiotensin II and endothelin-1 on gui-
C, LEWIS A, MADSEN J, BLANAZS A, ARMES S, BATTAGLIA G. nea pig gingival fibroblast cells in culture. Comp Biochem
Diffusion studies of nanometer polymersomes across tissue Physiol C Toxicol Pharmacol 2002; 132: 451–460.
engineered human oral mucosa. Pharm Res 2009; 26: 1718– 33. UEMURA H, ISHIGURO H, NAKAIGAWA N, NAGASHIMA Y,
1728. MIYOSHI Y, FJINAMI K, SAKAGUCHI A, KUBOTA Y. Angioten-
26. COLLEY HE, HEARNDEN V, JONES AV, WEINREB PH, VIOLETTE sin II receptor blocker shows antiproliferative activity in pros-
SM, MACNEIL S, THORNHILL MH, MURDOCH C. Development tate cancer cells: a possibility of tyrosine kinase inhibitor of
of tissue-engineered models of oral dysplasia and early inva- growth factor. Mol Cancer Ther 2003; 2: 1139–1147.
sive oral squamous cell carcinoma. Br J Cancer 2011; 105: 34. YANG X, ZHU MJ, SREEJAYAN N, REN J, DU M. Angiotensin
1582–1592. II promotes smooth muscle cell proliferation and migration
27. WASEEM A, ALI M, ODELL EW, FORTUNE F, TEH MT. Down- through release of heparin-binding epidermal growth factor
stream targets of FOXM1: CEP55 and HELLS are cancer and activation of EGF-receptor pathway. Mol Cells 2005; 20:
progression markers of head and neck squamous cell carci- 263–270.
noma. Oral Oncol 2010; 46: 536–542. 35. POMERANTZ RG, GRANDIS JR. The role of epidermal growth
28. MCGREGOR F, MUNTONI A, FLEMING J, BROWN J, FELIX DH, factor receptor in head and neck squamous cell carcinoma.
MACDONALD DG, PARKINSON EK, HARRISON PR. Molecular Curr Oncol Rep 2003; 5: 140–146.
changes associated with oral dysplasia progression and acqui- 36. MARSHALL RP, GOHLKE P, CHAMBERS RC, HOWELL DC, BOT-
sition of immortality: potential for its reversal by 5-azacyti- TOMS SE, UNGER T, MCANULTY RJ, LAURENT GJ. Angioten-
dine. Cancer Res 2002; 62: 4757–4766. sin II and the fibroproliferative response to acute lung injury.
29. HINSLEY EE, KUMAR S, HUNTER KD, WHAWELL SA, LAMBERT Am J Physiol Lung Cell Mol Physiol 2004; 286: L156–L164.
DW. Endothelin-1 stimulates oral fibroblasts to promote oral 37. PETTY WJ, MILLER AA, MCCOY TP, GALLAGHER PE, TALLANT
cancer invasion. Life Sci 2012; 91: 557–561. EA, TORTI FM. Phase I and pharmacokinetic study of angio-
30. AGER EI, NEO J, CHRISTOPHI C. The renin-angiotensin system tensin-(1-7), an endogenous antiangiogenic hormone. Clin
and malignancy. Carcinogenesis 2008; 29: 1675–1684. Cancer Res 2009; 15: 7398–7404.
31. OHSAWA M, OHUCHI N, TANIGUCHI Y, KIZAWA Y, KOIKE K,
IWAMOTO K, HAYASHI K, MURAKAMI H. Inhibition of

You might also like