ArticlePDF Available

Promotion of Cancer Cell Invasiveness and Metastasis Emergence Caused by Olfactory Receptor Stimulation

PLOS
PLOS ONE
Authors:

Abstract and Figures

Olfactory receptors (ORs) are expressed in the olfactory epithelium, where they detect odorants, but also in other tissues with additional functions. Some ORs are even overexpressed in tumor cells. In this study, we identified ORs expressed in enterochromaffin tumor cells by RT-PCR, showing that single cells can co-express several ORs. Some of the receptors identified were already reported in other tumors, but they are orphan (without known ligand), as it is the case for most of the hundreds of human ORs. Thus, genes coding for human ORs with known ligands were transfected into these cells, expressing functional heterologous ORs. The in vitro stimulation of these cells by the corresponding OR odorant agonists promoted cell invasion of collagen gels. Using LNCaP prostate cancer cells, the stimulation of the PSGR (Prostate Specific G protein-coupled Receptor), an endogenously overexpressed OR, by β-ionone, its odorant agonist, resulted in the same phenotypic change. We also showed the involvement of a PI3 kinase γ dependent signaling pathway in this promotion of tumor cell invasiveness triggered by OR stimulation. Finally, after subcutaneous inoculation of LNCaP cells into NSG immunodeficient mice, the in vivo stimulation of these cells by the PSGR agonist β-ionone significantly enhanced metastasis emergence and spreading.
Content may be subject to copyright.
Promotion of Cancer Cell Invasiveness and Metastasis
Emergence Caused by Olfactory Receptor Stimulation
Guenhae
¨
l Sanz
1,2
*, Isabelle Leray
3,4
, Aure
´
lie Dewaele
1,2
, Julien Sobilo
5
, Ste
´
phanie Lerondel
5
,
Ste
´
phan Bouet
6,7,8
, Denise Gre
´
bert
1,2
,Re
´
gine Monnerie
1,2
, Edith Pajot-Augy
1,2
, Lluis M. Mir
3,4
*
1 INRA, UR1197 Neurobiologie de l’Olfaction et Mode
´
lisation en Imagerie, Jouy-en-Josas, France, 2 IFR 144 NeuroSud Paris, Gif-sur-Yvette, France, 3 CNRS, UMR8203
Vectorologie et the
´
rapeutiques anti-cance
´
reuses, Institut Gustave-Roussy, Villejuif, France, 4 Univ. Paris-Sud, UMR8203, Orsay, France, 5 CNRS-TAAM, UPS44, Centre
d’Imagerie du Petit Animal, Orle
´
ans, France, 6 INRA, UMR 1313, Ge
´
ne
´
tique Animale et Biologie Inte
´
grative, Jouy-en-Josas, France, 7 CEA, DSV, IRCM, SREIT, Laboratoire de
Radiobiologie et Etude du Ge
´
nome, Jouy-en-Josas, France, 8 AgroParisTech, UMR 1313, Ge
´
ne
´
tique Animale et Biologie Inte
´
grative, Jouy-en-Josas, France
Abstract
Olfactory receptors (ORs) are expressed in the olfactory epithelium, where they detect odorants, but also in other tissues
with additional functions. Some ORs are even overexpressed in tumor cells. In this study, we identified ORs expressed in
enterochromaffin tumor cells by RT-PCR, showing that single cells can co-express several ORs. Some of the receptors
identified were already reported in other tumors, but they are orphan (without known ligand), as it is the case for most of
the hundreds of human ORs. Thus, genes coding for human ORs with known ligands were transfected into these cells,
expressing functional heterologous ORs. The in vitro stimulation of these cells by the corresponding OR odorant agonists
promoted cell invasion of collagen gels. Using LNCaP prostate cancer cells, the stimulation of the PSGR (Prostate Specific G
protein-coupled Receptor), an endogenously overexpressed OR, by b-ionone, its odorant agonist, resulted in the same
phenotypic change. We also showed the involvement of a PI3 kinase c dependent signaling pathway in this promotion of
tumor cell invasiveness triggered by OR stimulation. Finally, after subcutaneous inoculation of LNCaP cells into NSG
immunodeficient mice, the in vivo stimulation of these cells by the PSGR agonist b-ionone significantly enhanced metastasis
emergence and spreading.
Citation: Sanz G, Leray I, Dewaele A, Sobilo J, Lerondel S, et al. (2014) Promotion of Cancer Cell Invasiveness and Metastasis Emergence Caused by Olfactory
Receptor Stimulation. PLoS ONE 9(1): e85110. doi:10.1371/journal.pone.0085110
Editor: Aamir Ahmad, Wayne State University School of Medicine, United States of America
Received April 24, 2013; Accepted December 1, 2013; Published January 8, 2014
Copyright: ß 2014 Sanz et al. This is an open-access arti cle distributed under the terms of the Creative Commons Attribution License, which permits unrestricted
use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work was supported by INRA and CNRS (The National Centre for Scientific Research (France). This research was conducted in the scope of the LEA
EBAM (The European Associated Laboratory (LEA) entitled ‘‘Pulsed Electric Fields Applications in Biology and Medicine"). The funders had no role in study design,
data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: Guenhael.Sanz@jouy.inra.fr. (GS); Luis.MIR@gustaveroussy.fr (LMM)
Introduction
Olfactory receptors (ORs) are G protein-coupled receptors
mainly expressed in olfactory sensory neurons (OSNs) of the
olfactory epithelium, where they detect and discriminate myriads
of odorants according to a combinatorial code in which an OR
can be activated by various odorants and an odorant can stimulate
various ORs [1,2]. Moreover, ORs are expressed in non-olfactory
tissues [3–5] where they can play additional roles. They notably
govern sperm chemotaxis, regulate migration and adhesion of
muscle cells, and control serotonin secretion by enterochromaffin
(EC) cells [6–10]. Several studies also reported that some ORs can
be tumor marker, one of them modifying in vitro the proliferation
of LNCaP prostate cancer cells [11,12,13,14].
In particular, EC cells can acquire a tumoral phenotype and
differentially express ORs depending on the neuroendocrine
carcinoma evolution [15]. The BON cells, a human EC cell line
derived from a metastasis of a pancreatic carcinoma [16,17], were
described to endogenously express ORs [8] which could be tumor
markers when overexpressed [15]. Because BON cells were
derived from a metastasis, we explored whether activation of
ORs by agonist odorants could have a role in tumor progression.
To this end, we decided to identify the ORs expressed in BON
cells. However the agonist or antagonist odorants specific of BON
cells ORs are unknown, like for most of the hundreds of identified
human ORs. We thus tried to develop a model by transfecting
these cells with deorphanized ORs. The heterologous expression
achieved allowed us to assess the in vitro invasiveness of these cells
upon stimulation with the odorant ligand of the transfected
receptor. Furthermore, we identified PI3 kinase cPI3Kc as a
component of the signaling pathway induced by OR stimulation
and promoting cell invasiveness. A more physiological model was
also used in vitro, the LNCaP prostate cancer cells which
overexpress the PSGR (Prostate Specific G protein-coupled
Receptor), an endogenous and deorphanized OR considered as
a tumor marker, and wich was described to inhibit the
proliferation of these cells in vitro [12]. This model was then used
in vivo to analyze the role of ORs stimulation in tumor progression,
that is in metastasis emergence and spreading.
Materials and Methods
Ethics Statement
The animals were handled in conformity with the Guidelines of
the French government regarding operative procedures and animal
care. Protocol was approved by the ethics comity for experiments
with animals named ‘‘Comite´ d’Ethique en Expe´rimentation
Animale de l’IRCIV’’ CEEA-26 (protocol number 2012-043).
PLOS ONE | www.plosone.org 1 January 2014 | Volume 9 | Issue 1 | e85110
Reverse transcription (RT)-PCR, cloning and sequencing
Total RNAs were extracted using TRIzol reagent (Invitrogen)
and treated with DNase I. RT was performed with the «
SuperScript First-StrandH, Synthesis System for RT-PCR» kit
(Invitrogen).
For single cell RT-PCR, single cells were collected by aspiration
into a glass pipette and RT was performed using the « Single Cell
Superscript
TM
III Cells Direct cDNA Synthesis System » kit
(Invitrogen) after cell disruption, protein denaturation and DNAse
treatment.
Nested PCR was carried out starting from 1
mL of RT products
and using degenerate primers targeting OR conserved regions, or
primers specifically targeting OR identified with the degenerate
primers. Degenerate primers sequences were kindly provided by
Stephan Bieri (Givaudan, Switzerland). Absence of genomic DNA
was controlled using human GAPDH or b-actin primers on
DNase I-treated RNAs without reverse transcriptase.
PCR products amplified with degenerate primers were cloned
into the pGEM-T vector (Promega) and sequenced by Beckman
Coulter Genomics. PCR products amplified with specific primers
were directly sequenced (Beckman Coulter Genomics).
Chemicals
Odorants, DMSO and mineral oil (M3516) were purchased
from Sigma-Aldrich, Fluka or Acros Organics at the highest purity
available. AS605240 was purchased from Euromedex (Selleck,
S1410) and gallein from TOCRIS bioscience. Paraffin (CellWax)
was obtained from CML, and hemalun, eosin, and safran from
RAL.
Mammalian expression vectors
OR1G1 or OR17-40 coding sequences were introduced into
the pCMV-Tag3B mammalian expression vector (Stratagene) in a
way resulting in the fusion of a cmyc epitope at the receptor N-
terminus. The resulting vectors were named pCMV-TagOR1G1
and pCMV-TagOR17-40.
Cell culture and transfection
BON cells (subclone #7) were kindly provided by Dr Courtney
M. Townsend (Department of Surgery UTMB, Galveston, TX
77551, USA) [16,17]. They were grown in DMEM/F-12 (Ham)
without phenol red (GIBCO, Invitrogen Corporation) supple-
mented with 10% fetal bovine serum (Hyclone, Perbio) and
antibiotics (100 U penicillin/mL and 100
mg streptomycin/mL,
Invitrogen), at 37uC in a humidified incubator with 5% CO
2
. Cells
were transiently transfected with pCMV-TagOR1G1 or pCMV-
TagOR17-40 using jetPEI
TM
(Polyplus-transfection) according to
the manufacturer’s instructions. OR expression at the cell surface
was checked by immunofluorescence microscopy using the
monoclonal anti-cmyc-Cy3 antibody (C6594, Sigma-Aldrich) on
non-permeabilised cells.
LNCaP cells were purchased from ATCC (Clone FGC, No.
CRL-1740
TM
) at passage 19, and grown in RPMI 1640 medium
(ATCC, No. 30-2001) supplemented with 10% fetal bovine serum
(ATCC, No. 30-2021), at 37uC in a humidified incubator with 5%
CO
2
.
Calcium imaging
BON and LNCaP cells were seeded onto a 96-well culture plate
(black microtiter plate, Greiner Bio-one), respectively at a density
of 10
5
and 0.5610
5
cells per well. 24 hours later, cells were loaded
with 2.5
mM of fluo-4 acetoxymethyl ester (Molecular Probes), as
previously described [18]. Calcium imaging was performed using
an inverted epifluorescence microscope (CK40 Olympus)
equipped with a digital camera (ORCA-ER, Hamamatsu
Photonics). Ca
2+
reponses were observed at 460–490 nm excita-
tion and $ 515 nm emission wavelengths. Data acquisition and
analysis was performed using the SimplePCI software (Hama-
matsu, Compix). Odorants and mineral oil were prepared
extemporaneously by a first dilution into DMSO and then serial
dilutions into Hanks’ salt solution (Eurobio) supplemented with
20 mM Hepes, pH 7.2. Stimuli were tested at concentrations that
do not elicit calcium responses in mock-transfected cells. 1
mM
isoproterenol (Sigma-Aldrich) was applied as a positive control.
The Ca
2+
signal was measured as the relative change in
fluorescence intensity DF/F = (F–F
0
)/F
0
, where F
0
is the
fluorescence level before stimulation. Results were expressed as
the mean of the DF/F of at least twenty cells.
In vitro assessment of cell invasion
Collagen type I gels were prepared as described by De Wever
[19]. Cells were cultured for 48 hours before seeding them as a
suspension of single cells deposited on top of the collagen type I
gels. To stimulate ORs, odorants and mineral oil were first diluted
into DMSO and then into the collagen I solution or the culture
medium. The effects of specific inhibitors of PI3Kc (AS605240)
and bc subunits of G proteins (gallein) were also assessed by
adding them to the collagen gel and culture medium. For controls,
DMSO was added at the same final concentration used to dilute
these chemicals. The amount of added DMSO did not exceed
0.2% and did not modify the number of invasive cells compared to
tests without DMSO (data not shown). 24 hours after their
seeding, invasive cells presenting invasive extensions into the
collagen gel and non-invading cells were counted in 10–15
microscope fields randomly selected. Results were expressed as the
percentage of invasive cells (invasion index). Morerover, F-actin
cytoskeleton was observed using rhodamine-conjugated phalloidin
(Invitrogen). For this purpose, cells were fixed for 20 min with 3%
paraformaldehyde in PBS at room temperature, then permeabi-
lized for 15 min with 0.5% Triton X-100 in PBS and blocked for
30 min with 2% BSA, 1% glycine in PBS. Cells were incubated
with rhodamine-phalloidin (1:300) in PBS for 30 min at room
temperature and extensively washed with PBS before observation
with an inverted epifluorescence microscope.
Mice
Nod Scid Gamma (NSG) male mice were bred in the animal
housing facilities of the Institut Gustave Roussy, with free access to
food and water. Plastic cages were connected to controlled
ventilated racks. The cages with the animals exposed to the
odorant b-ionone were connected to a separated ventilation unit.
In vivo assessment of cell invasion and metastases
LNCaP cells at passage 25 were inoculated into 8 week-old
castrated male NSG mice (castration was performed two weeks
before cell inoculation). 10
6
cells were suspended in 75 mLof
RPMI 1640 plus 75
mL of Matrigel (BD Biosciences) and injected
with a needle (26G) into the subcutaneous space, at 2 sites in each
flank of the mice. The odorant b-ionone was first diluted into
DMSO at a concentration of 100 mM and then into the RPMI +
Matrigel mixture at the final concentration of 100
mM. DMSO
was also added at the same dose to the RPMI + Matrigel mixture
without odorant. A first group of 5 mice was inoculated with
LNCaP cells (in the presence of DMSO) and received no further
treatment. Five other mice were inoculated with LNCaP cells (in
the presence of DMSO) and brushed with mineral oil three times a
day during 6 weeks and then three times a week until sacrifice. A
Olfactory Receptors Promote Tumor Cell Invasion
PLOS ONE | www.plosone.org 2 January 2014 | Volume 9 | Issue 1 | e85110
third group of 5 mice was inoculated with LNCaP cells in the
presence of b-ionone in DMSO. These mice were brushed with
1mMb-ionone directly diluted in mineral oil three times a day
during 6 weeks and then three times a week until sacrifice. Before
sacrifice, some animals were first examined by tomoscintigraphy
(SPECT, NanoSPECT/CT Bioscan) using
99m
Tc-MDP, a classi-
cal bone scintigraphy agent for functional imaging of the bone.
This analysis was not performed on all animals because it
appeared less informative than X rays in our study. Thus all mice
were explored in vivo by microcomputed tomography (
mCT)
(CT120, General Electric Healthcare) to detect bone metastasis.
360 X ray projections were collected in 1u increments (100 kVp,
50 mA, 20 msec exposure) for about 5 min total scan time. Images
were reconstructed into 3D volumes (50
mm resolution) on a
reconstruction cluster using a modified tent-FDK conebeam
algorithm (GE reconstruction software). 3D data were processed
using MicroView (GE Healthcare). Data analysis was performed
first on individual slices (axial, coronal, sagittal) then on
reconstructed volumes and MIP images (Maximum Intensity
Projection). Animals were sacrificed when tumor size exceeded
1,500 mm
3
. Upon autopsy, tumors and tissues known to harbor
metastases from prostate tumors such as lymph nodes, lungs and
spines, were sampled. Livers and Tyson glands were also sampled,
some livers appearing anomalous and some Tyson glands
surprisingly large. Tissues were fixed for 24 hours in formaldehyde
then stored in 70% ethanol at 4uC. For spines, decalcification was
realized by an additional incubation in 10% EDTA, pH 7.4, at
4uC during one week. All samples were dehydrated in ethanol and
included in paraffin. Serial sections of 5
mm thickness were
prepared and dewaxed in toluene and rehydrated in ethanol and
then water. Some sections were stained with hemalun (RAL), eosin
and safran (HES staining). Immunohistochemistry was performed
on other sections using anti-PSGR (LS-A6332, Cliniscience), anti-
PSA (ab9537, abcam), or rabbit serum as a negative control, the
Vectastain Elite ABC-Peroxidase Kits Rabbit IgG (Cliniscience),
and a DAB revelation (SK-4100, Vector).
Results
ORs endogenously expr essed by BON cells
Since BON cells display an heterogeneous morphology, we
isolated homogeneous subclones. OR expression was investigated
by nested PCR on cDNAs from nine clones using degenerate
primers targeting OR conserved regions, and PCR products
sequencing. We detected ORs transcripts in six of the clones
(Table S1). Among them, five displayed expression of more than
one OR gene or pseudogene, and the panel of ORs identified
varied from clone to clone. To confirm these results, we performed
nested PCR with primers specifically targeting the previously
identified ORs. Actually all nine clones expressed ORs transcripts
(Table 1) and some of them (OR7D2, OR1F1) were found in most
of the clones. It must be highlighted that OR7A17, OR7D2 and
OR2A1 transcripts are also found in several tumors (ESTs listed in
the HORDE database).
To further assess that, contrary to OSNs, BON cells co-express
several ORs, we analyzed OR expression at the single-cell level.
We succeeded in amplifying cDNAs corresponding to GAPDH or
b-actin for most tested cells, but OR cDNAs could be amplified
only for a few cells, probably because of the very low level of OR
mRNAs at the single-cell level. Our data show that some single
BON cells do co-express more than one OR transcript (Figure
S1a).
Heterologous functional expression of ORs in BON cells
Since BON cells endogenously express ORs, we infered that
they could also heterologously express functional ORs after
transfection of the OR1G1 and OR17-40 genes. BON cells
appeared to express these heterologous receptors and to expose
them at the plasma membrane (Figure S1b). We also found in
BON cells the transcript of REEP1, a protein which facilitates OR
expression in OSNs [20] (Figure S1c). We then demonstrated that
the heterologously expressed receptors are functional, inducing a
calcium response when they are stimulated with their respective
ligand (1-nonanol for OR1G1 and helional for OR17-40 [18,21])
(Figure 1). The calcium response induced by stimulation of the
OR17-40 receptor is less pronounced than that induced by
stimulation of the OR1G1 receptor, but it remains significant.
Differences between OR response levels can be due to different
expression levels of the receptors, to a different coupling efficiency
with the endogenous G-proteins of heterologous cells, or to a
different efficiency of the ligands used. Mock-transfected cells did
not respond to nonanol nor helional, showing that the odorants
tested are not agonists of the ORs endogenously expressed in BON
cells.
OR-induced enhancement of cell invasiveness
Using BON cells heterologously expressing OR1G1 or OR17-
40 receptors, we assessed the invasiveness of collagen type I gels
[19] by these cells, stimulated or not with the odorant agonists of
these ORs. In absence of odorant stimulation, the invasiveness of
BON cells was not modified by heterologous expression of ORs
(the invasion index remains around 3%, Figure 2a). Nonanol
stimulation increased significantly the invasion index of OR1G1-
expressing cells (OR1G1 cells) by a factor of 2.7, whereas helional
stimulation increased the invasion index of OR17-40 cells by a
factor of 2.5 (Figure 2a). We observed that 10
26
and 10
27
Mof
nonanol induced the same invasion level, whereas 10
26
M
appeared more efficient in activating OR1G1 in calcium imaging
experiments. This may be due to the inability of BON cells to
reach larger invasion levels (around 10% invasive cells). Nonanol
and helional had no significant impact on mock-transfected
control cells. Nonanol had no significant effect on OR17-40
expressing cells, nor helional on OR1G1 expressing cells.
Furthermore, vanillin, an antagonist of the OR1G1 receptor
[22], was able to specifically counteract the invasiveness induced
by nonanol in OR1G1 cells. The invasion index of control cells
stimulated by nonanol alone or by a mixture of nonanol and
vanillin was unchanged (Figure 2a). Invasive cellular extensions
into collagen type I gels, characterizing the invasive cells, were also
observed after immunolabeling of the F-actin cystoskeleton (Figure
2b). All together, these results demonstrate that, in vitro, ORs
stimulation by odorants can specifically promote invasiveness of
the OR-expressing cancer cells.
We confirmed this result using the LNCaP prostate cancer cells
which endogenously express an OR, the PSGR. This receptor has
known agonist and antagonist odorants [12], respectively the b-
ionone and a-ionone. We used a 100
mM concentration of b-
ionone to stimulate LNCaP cells, since this dose was already
reported to stimulate the PSGR [12] and it induced the highest
invasiveness of LNCaP cells in our hands (data not shown). As
shown in Figure 2c, stimulation of PSGR with 100
mM b-ionone
increased invasiveness of LNCaP cells by a factor of 2.75 and this
effect was totally abrogated by the antagonist a-ionone. Alone, this
antagonist had no effect on LNCaP cells invasion level. While
there is no negative control with LNCaP cells that would not
express PSGR, the drastic pharmacological effect of a-ionone
argues in favor of a specific effect of b-ionone through PSGR
Olfactory Receptors Promote Tumor Cell Invasion
PLOS ONE | www.plosone.org 3 January 2014 | Volume 9 | Issue 1 | e85110
stimulation. Exclusion of a non specific chemical effect of b-ionone
on LNCaP cells inducing invasiveness is also supported by the fact
that a-ionone, which is very similar to b-ionone and was applied at
twice the b-ionone dose, did not induce invasiveness of LNCaP
cells. Moreover, we tested the effect of 100
mM b-ionone on the
invasiveness of PC3 cells, other prostate cancer cells that do not
express the PSGR [12], and we did not observe an increased
invasiveness in these cells. Experimental results detailed below also
support the idea that LNCaP invasiveness can be enhanced
through PSGR stimulation.
Involvement of PI3Kc in cell invasiveness induced by ORs
PI3Kc activation through GPCRs can be involved in
transforming functions such as invasion [23], and a crosstalk
between odorant signaling and PI3Kc was described in olfactory
sensory neurons [24,25]. We thus explored whether PI3K c could
be part of the signaling pathway which is triggered by the odorant
activation of ORs and promotes cell invasiveness. First we showed
the expression of PI3Kc in BON and LNCaP cells by crude lysates
immunoblotting with an antibody targeting PI3Kc (data not
shown). We then assessed the invasiveness of BON cells
hetorologously expressing OR1G1 or of LNCaP cells upon
stimulation with agonists of OR1G1 or PSGR, in the presence
of a specific inhibitor of PI3Kc (AS605240). 10
26
M of AS605240
have been reported to completely inhibit PI3Kc [26]. Concerning
BON cells, using 10
26
M and 10
27
M of AS605240, we found a
similarly large (about 80%) but not total reduction of the cell
invasiveness promoted by OR1G1 upon nonanol stimulation
(Figure 3), indicating that the maximal effect is observed at 10
27
M
of AS605240. Thus, PI3Kc appears to play a major role in
mediating BON cell invasiveness promoted by the OR stimulation
by its specific odorant, even if other signaling pathways might also
be involved. Involvement of PI3Kc was confirmed for LNCaP
cells (Figure 3). However, contrary to BON cells, PI3Kc inhibitor
AS605240 induced a reduction of LNCaP invasiveness even in
absence of PSGR stimulation. Therefore PI3Kc seems to be also
involved in the basal invasiveness of LNCaP cells. Moreover, since
PI3Kc can be activated by the G
bc
subunit of the G proteins
through GPCR activation [27], we used gallein, a G
bc
subunits
inhibitor that interferes with the interaction of G
bc
subunits with
PI3Kc [28], and showed that it counteracted the enhancement of
LNCaP cell invasiveness induced by PSGR stimulation (Figure 3).
This result also supports the involvement of PI3Kc in the
invasiveness of tumor cells induced by OR stimulation.
OR activation-induced enhancement of cancer cell
invasiveness in vivo
Since in vitro enhancement of cell invasiveness by ORs activation
suggests a possible role of (at least some) ORs in metastasis
emergence in vivo, we inoculated LNCaP prostate tumor cells
subcutaneously into immunodeficient NSG (NOD scid gamma)
mice. Animals were either left untreated, or daily brushed on skin
with PSGR agonist b-ionone diluted in mineral oil (an oily
excipient needed to apply the lipophilic odorants over the mice
skin), or with mineral oil alone as a control. Tumor size was
measured and metastases were detected by in vivo imaging and by
post-mortem immunohistochemistry using antibodies targeting
PSGR or PSA (Prostate Specific Antigen) (examples of spine and
lung metastases are displayed in Figure 4). PSGR expression was
detected in primary tumors and in all metastases (see other
examples in Figure S2), confirming that this receptor was present
Table 1. Targeted search of ORs expressed by subclones of BON cells, using nested RT-PCR with primers specific to previously
identified ORs.
Clone OR4F16 OR13H1 OR7A17 OR10Q1 OR7D2 OR2A1 OR6V1 OR1F1 OR13A1
1X X X X X
3X X X
4X X X
8X X X X
9XXX
10 X X X X
14XXXXX X
19 X X X X
23XX XXXX
(X) indicates identified ORs.
doi:10.1371/journal.pone.0085110.t001
Figure 1. Functional response of ORs heterologously ex-
pressed in BON cells. BON cells were transiently transfected to
express OR1G1 or OR17-40 receptors. 72h later, cells were loaded with
fluo-4 and stimulated with the respective odorant ligands of the
transfected ORs (1-nonanol and helional). Calcium responses due to the
interaction between the OR and its specific odorant agonist are
expressed as the mean fluorescence variation DF/F (%). (open circles)
OR1G1 cells, 1-nonanol ; (filled diamonds) OR17-40 cells, helional ; bars
indicate standard deviation (n = 3). Mock-transfected cells did not
respond to 1-nonanol nor helional.
doi:10.1371/journal.pone.0085110.g001
Olfactory Receptors Promote Tumor Cell Invasion
PLOS ONE | www.plosone.org 4 January 2014 | Volume 9 | Issue 1 | e85110
and possibly activated during our experiments. Without treatment,
metastases emerged mainly in the inguinal nodes and occasionn-
ally in spine and liver (Figure 4a). Metastases located in the
inguinal nodes were well developed while those located in spine
and liver were micrometastases. The number of metastases
increased upon treatment with mineral oil and their localization
was more diverse in the presence of b-ionone. Actually, metastases
appeared in lungs and Tyson glands only for mice treated with b-
ionone (3 out of 5 animals for Tyson glands and 2 out of 5 animals
for lungs). Moreover, metastases located in Tyson glands were
highly developed, with sizes approaching 1,000 mm
3
. In lungs,
only micrometastases were detected, like in spine and liver. Since
mice were not sacrificed at the same time, but depending on tumor
size, we show in Figures 5b and 5c the evolution with time of the
number of metastases according to the number of sacrificed mice
and the average number of metastases per mouse at the time of
sacrifice for each experimental group. We observed that mice
treated with mineral oil or b-ionone had to be sacrificed earlier,
due to faster tumor growth, and that they displayed a significant
increase in metastases number compared to untreated mice. In
parallel we also observed that tumors developed at 85% of the
inoculated sites in untreated mice, while they were less numerous
(50%) in mineral oil or b-ionone treated mice.
The results with the mineral oil were intriguing. Since ORs can
be activated by different molecules [2,12,18], we investigated
whether mineral oil can stimulate the PSGR in in vitro cell invasion
assays and calcium imaging experiments. Mineral oil increased
both the invasiveness and the calcic response of LNCaP cells, and
the PSGR antagonist a-ionone abrogated these effects (Figure 6
a,b). These results demonstrate that a mineral oil component
stimulated LNCaP cells via the PSGR, promoting their invasive-
ness. Moreover, LNCaP cells invasiveness induced by mineral oil
was inhibited by PI3Kc or G
bc
subunits inhibitors (respectively
AS605240 and gallein), demonstrating that PI3Kc is involved in
this process, as it is involved in cell invasiveness induced by b-
ionone. However, even if mineral oil enhanced metastasis
emergence, metastasis occurence was even more pronounced in
the presence of b-ionone, resulting in the cells spreading to lungs
and Tyson glands that occurred only in the presence of b-ionone.
All these data converge to demonstrate that stimulation of an OR,
Figure 2. Promotion of cancer cells invasiveness upon odorant stimulation. (a) BON cells were transiently transfected to express OR1G1 or
OR17-40 receptors or mock-transfected. Cells were seeded on collagen type I gels and stimulated by the respective odorant ligands of OR1G1 and
OR17-40 receptors (nonanol: OR1G1 agonist, vanillin: OR1G1 antagonist, helional: OR17-40 agonist). Invasive cells were counted 24 hours later.
Results are presented as the invasion index. (b) Modification of the F-actin cytoskeleton of BON cells in collagen type I matrices. F-actin was revealed
by rhodamine-conjugated phalloidin. Invasive extensions into collagen gels characterizing invasive cells are indicated by arrows. (c) LNCaP cells were
seeded onto collagen type I gels and stimulated by PSGR ligands (b-ionone: agonist, a-ionone: antagonist). Invasive cells were counted 24 hours later.
Results are presented as the invasion index relative to control cells without odorant stimulation. Standard deviation of the control was 13,42%.
Statistics were performed using a two-tailed Student test and bars indicate standard deviation (n = 3).
doi:10.1371/journal.pone.0085110.g002
Olfactory Receptors Promote Tumor Cell Invasion
PLOS ONE | www.plosone.org 5 January 2014 | Volume 9 | Issue 1 | e85110
the PSGR, can contribute to the dissemination of prostate tumor
cells and in metastases generation.
Discussion
In this study, we showed that contrary to OSNs, individual
BON tumor cells (BON) co-express various ORs transcripts.
Genes encoding the co-expressed ORs are not located in the same
cluster nor on the same chromosomes, excluding a simultaneous
expression controlled by a common promoter. OR co-expression
by a same cell and differences in the set of expressed ORs between
cells could ensure a wide detection of luminal odorants in the
gastro-intestinal tract. Indeed normal and neoplastic EC cells were
previously reported to detect odorants through OR activation,
which participate in the control of their serotonin secretion [8,9].
Deregulation of serotonin release can induce pathological disor-
ders [29-32] and thus it could be expected that such disorders
could result from changes in the levels and variety of the ORs
expression. We also unexpectedly found that BON tumor cells
express OR pseudogenes at the mRNA level. Some of them
(OR7E38P and OR2A9P) and several functional receptors
expressed by BON cells (OR7A17, OR7D2 and OR2A1) have
been found in other tumors. Moreover, other ORs are overex-
pressed in EC tumor cells [15] and prostate tumor cells [12]. Thus,
the expression of some OR genes and pseudogenes in BON cells,
which originate from a pancreatic carcinoma metastasis, could be
part of their tumor phenotype.
Since BON cells endogenously express ORs, we also demon-
strated their ability to efficiently express transfected functional
heterologous ORs with known odorant ligands. In particular, we
found that the transcript of REEP1, a protein described to
improve OR expression [20], was present in BON cells. These
cells are thus a promising system for deorphanizing ORs, which
still remains a challenge due to the well-known low and variable
Figure 3. PI3Kc involvement in cell invasiveness promoted by
ORs. BON cells heterologously expressing the OR1G1 receptor were
seeded onto collagen type I gels and were stimulated by nonanol
(OR1G1 agonist) in the presence of a specific PI3Kc inhibitor
(AS605240). LNCaP cells were seeded onto collagen type I gels and
were stimulated by b-ionone (PSGR agonist) in the presence of a
specific PI3Kc inhibitor (AS605240) or an inhibitor of bc subunits of G
proteins (gallein). Invasive cells were counted 24 hours later. Results are
presented as the invasion index relative to that of control cells (with no
odorant nor AS605240 nor gallein). Standard deviation of the control
was 4,74% for the control BON cells and 13,86% for the control LNCaP
cells. Statistics were performed using a two-tailed Student test and bars
indicate standard deviation (n = 3).
doi:10.1371/journal.pone.0085110.g003
Figure 4. Examples of metastases found in the spine or the lungs of immunodeficient mice inoculated with LNCaP cells. Metastases
are indicated by arrows. In the spine, metastases were observed using microcomputed tomography (Maximum Intensity Projection) and confirmed
post-mortem by HES staining and immunohistology using anti-PSA (prostate specific antigen) and anti-PSGR antibodies (only one metastasis is
presented). In the lungs, metastases were characterized by HES staining and immunohistology using anti-PSA and anti-PSGR antibodies.
doi:10.1371/journal.pone.0085110.g004
Olfactory Receptors Promote Tumor Cell Invasion
PLOS ONE | www.plosone.org 6 January 2014 | Volume 9 | Issue 1 | e85110
OR expression in heterologous cells. To date, even using HEK
293 cells engineered to improve OR expression, only 4% of 245
human ORs investigated were deorphanized [20,33,34]. Identify-
ing OR ligands and studying OR pharmacology is of increasing
interest given the high number of ORs (several hundreds in
humans) and their involvement in various physiological functions,
and possibly in physiopathological functions such as tumor
progression.
Furthermore, we demonstrate, for the first time, that ORs play a
role in tumor progression by promoting cell invasiveness and
metatasis emergence. Taking advantage of the ability of BON cells
to heterologously express ORs for which odorant ligands are
known, we showed that odorant stimulation of heterologously
expressed ORs enhanced BON cell invasiveness in vitro. We further
assess this result using LNCaP prostate cancer cells, which
overexpress an OR, the PSGR (also named OR51E2), described
as a prostate tumor marker [11,12]. Stimulation of LNCaP cells
with the PSGR agonist b-ionone promoted their invasiveness, and
this phenomenon was inhibited by a-ionone, a PSGR antagonist.
Recently, ORs were reported to participate in early cytokinesis by
exerting a regulatory role on actin cystoskeleton, and particularly
in cancer cell lines [35]. This suggests that ORs could favor cell
invasion by regulating actin cytoskeleton. Yet, the signaling
pathway triggered by OR stimulation and inducing cell invasive-
ness remains to be explored. Our results provide a first evidence
for a major role of PI3Kc, which is also supported by the following
data. The G
aolf
protein subunit, activated by ORs in OSNs, was
reported to promote invasion in human digestive and urogenital
epithelial cells, and in particular in LNCaP cells, through PI3
kinase, Rho GTPases and PKC dependent pathways [36]. Yet, in
our study, we found that PI3K is activated by bc subunits of G
proteins. Thus, in LNCaP cells, two PI3K dependent pathways
originating from PSGR activation could coexist. Moreover,
GPCRs activation of PI3Kc is involved in transformed cell
functions such as invasion and alteration of homotypic cell-cell
adhesion [23], and a crosstalk between odorant signaling and
PI3Kc was reported in OSNs [24,25]. All together, these data
suggest that an interplay between ORs, G proteins and PI3Kc in
tumor cells might promote the cell invasiveness phenotype.
We also demonstrate in vivo that stimulation of some ORs
expressed in tumor cells could facilitate cells dissemination and
metastasis generation. Indeed, we stimulated xenografted LNCaP
cells in NSG mice with a PSGR ligand. Without stimulation, mice
developed tumors at 85% of the inoculation sites, but also some
metastases in inguinal nodes, spine and liver, which is seldom
reported in the literature concerning prostate cancer models using
immunodeficient mice and LNCaP cells [37–39]. When treated
with mineral oil alone or containing b-ionone, mice showed a
significantly increased number of metastases, despite the small
number of animals in the experiment. Noteworthy, only the b-
ionone treated mice developed metastases in other tissues, namely
in lungs and Tyson glands, and those in Tyson glands were
particularly well developed. In addition, we showed that the
mineral oil used as an excipient in our experiments induces
LNCaP cell invasiveness by activating the PSGR and involving a
PI3Kc inhibitor pathway, like b-ionone. These results corroborate
that metastases emergence enhancement observed in mineral oil
treated mice can be due to PSGR activation, at least partly, since
we cannot exclude the presence of other ORs in LNCaP cells and
of their specific ligands in the mineral oil. It would be interesting to
identify the mineral oil component activating the PSGR, but this
appears difficult due to the complex and unavailable detailed
composition of mineral oil. Moreover, while in vitro there was no
additive effect of mineral oil and b-ionone in promoting cell
invasion, in vivo, addition of b-ionone to mineral oil not only
slightly boosted metastasis emergence in the same tissues but
moreover induced metastasis spreading to additional tissues. Also,
mice treated with b-ionone had to be sacrificed earlier due to
faster tumor growth, and the number of metastases detected in
Figure 5. Metastases detection in immunodeficient mice
inoculated with LNCaP cells. (a) The number of metastases
observed in various tissues after mice sacrifice (white: untreated control
mice, gray: mice treated with mineral oil, black: mice treated with 1 mM
b-ionone in mineral oil). (b) Cumulative number of metastases,
regardless of their location, as a function of the time elapsed between
LNCaP cells inoculation and mice sacrifice (white: untreated control
mice, gray: mice treated with mineral oil, black: mice treated with 1 mM
b-ionone in mineral oil). Mice were sacrificed when tumor size exceeded
1,500 mm
3
. The number of sacrificed mice is indicated in brackets for
each time point and each mice group. (c) Curves reporting the ratios of
the number of metastases over the number of sacrificed mice as a
function of the time (black dashed: untreated control mice, gray: mice
treated with mineral oil, black: mice treated with 1 mM b-ionone in
mineral oil).
doi:10.1371/journal.pone.0085110.g005
Olfactory Receptors Promote Tumor Cell Invasion
PLOS ONE | www.plosone.org 7 January 2014 | Volume 9 | Issue 1 | e85110
those mice was sligthly higher. Thus, in the limit of our
experimental conditions, where the real amount of mineral oil
and b-ionone stimulating the LNCaP cells cannot be controlled,
the presence of b-ionone appeared to further promote metastasis
emergence. We cannot totally exclude that the observed effect on
metastases emergence could also be partly due to an increased
tumor growth rate in the presence of b-ionone. Indeed, b-ionone
seemed to accelerate tumor growth, as suggested by the early
sacrifice of some animals of the b-ionone treated group (actually,
all the animals were sacrificed when their largest lesion the
inoculated tumor or a metastasis reached 1500 mm
3
). However,
the treated animals sacrificed earlier presented more metastases
than untreated animals sacrificed later, whereas it could be
expected the contrary since the number of metastases usually
increase with age. Moreover, there were no significant differences
in tumor size between mice groups at sacrifice time. So, the
observed differences in the number and localization of the
metastases cannot be attributed just to differences in tumor
growth. We can also notice that the faster tumor growth observed
in vivo in the presence of b-ionone is not in agreement with the
results of Neuhaus et al. [12] demonstrating a reduced in vitro
proliferation of LNCaP cells in the presence of b-ionone. Finally,
we observed that tumor engraftement was less important in treated
mice than in untreated mice. This could be due to a greater
migration of the cells stimulated with mineral oil or b-ionone from
the inoculation site, detrimental to the local tumor development.
In humans, the PSGR can be activated by endogenous ligands
such as steroid hormones [12]. Nevertheless, we performed this
study in an androgen-depleted context (that is in castrated male
mice) and therefore our findings are of major relevance concerning
the androgen-independent progression of prostate cancer, which is
still poorly understood. Since mineral oil and b-ionone are present
in various products of our close environment, namely cosmetics,
food and beverages, it is conceivable that these exogenous
molecules could be found in the body and our findings might
help defining prostate cancer prevention measures. An important
prospect of future studies would be also to identify PSGR odorant
antagonists (other than a-ionone which is irritant and harmful,
side effects that would limit its use) as potential new anti-cancer
agents probably possessing low side effects since the PSGR is not
widely expressed in normal tissues.
Finally this study should be extended to other cancer types.
Indeed, not only a genomics approach has associated the olfactory
transduction pathway with an increased pancreas cancer risk [40]
but moreover overexpression of 34 ORs genes has been reported
in patients bearing breast tumors caused by CHEK2 1100delC-
mutation [41]. Hence, it appears of great importance to continue
addressing the role of ORs in tumor progression, in hormono-
dependent tumors as well as in non hormono-dependent ones.
Supporting Information
Figure S1 ORs expression in BON cells. (a) Example of
nested PCR performed on RT products obtained from a single
BON cell from clone 14, using two pairs of primers specifically
targeting OR10Q1 or OR4F16. Each amplification product was
was about 400 bp long and was verified by sequencing. Lane 1: 2-
Log DNA Ladder (BioLabs), lane 2: OR4F16 amplification, lane
3: OR10Q1 amplification, lanes 4 and 5: controls without cell with
each pair of primers. (b) Heterologous expression of ORs by BON
cells. BON cells were transiently transfected to express OR1G1 or
OR17-40 receptors tagged by the cmyc epitope at their
Figure 6. Mineral oil effect on LNCaP cells invasiveness and calcium response. (a) LNCaP cells were seeded onto collagen type I gels and
were stimulated by mineral oil or mineral oil mixed with 10
24
M b-ionone, 2.10
24
M a-ionone, 10
27
M AS605240 or 10
25
M gallein. Mineral oil was first
diluted 1/4 in DMSO (mineral oil was not totally solubilized at this dose, but this allowed us to expect a very large solubilization of the mineral oil
components) and the solution was then diluted 1/1000 in the culture medium or collagen I gel. Invasive cells were counted 24 hours later. Results are
presented as the invasion index relative to that of control cells (with only the DMSO). Bars indicate standard deviation. Standard deviation of the
control was 3,67%. Results obtained with mineral oil alone and with mineral oil containing b-ionone are not significantly different. Statistics were
performed using a two-tailed Student test (n = 3). (b) LNCaP cells were loaded with fluo-4 and stimulated with mineral oil or a mixture of mineral oil
and 10
24
M a-ionone. Calcium responses are expressed as the mean fluorescence variation DF/F relative to that of control, which corresponds to cells
stimulated with buffer containing the same DMSO concentration than the odorants and mineral oil samples. Bars indicate standard deviation.
Standard deviation of the control was 15,96%. Statistics were performed using a two-tailed Student test (n = 7).
doi:10.1371/journal.pone.0085110.g006
Olfactory Receptors Promote Tumor Cell Invasion
PLOS ONE | www.plosone.org 8 January 2014 | Volume 9 | Issue 1 | e85110
extracellular N-terminal end. 72h later, non-permeabilised cells,
labeled with an anti-cmyc-Cy3 antibody, were observed by
immunofluorescence microscopy. Mock-transfected cells are
shown as a negative control. (c) REEP1 transcripts in BON cells.
RT-PCRs were performed using total RNA from BON cells and
primers targeting human RTP1, RTP2 or REEP1 cDNAs.
Expected PCR products lengths were 686 bp for RTP1, 564 bp
for RTP2 and 524 bp for REEP1. Negative controls were obtained
with the same primers on RNAs treated without reverse
transcriptase. Only REEP1 cDNA was amplified at the expected
length (boxed in white).
(TIF)
Figure S2 PSGR expression in various tissues of mice
inoculated with LNCaP cells. PSGR expression in primary
tumors, inguinal nodes, Tyson glands and livers was detected using
an anti-PSGR antibody (LS-A6332, Cliniscience) and negative
controls were performed using rabbit serum.
(TIF)
Table S1 Blind search of ORs expressed by subclones of
BON cells, using nested RT-PCR with degenerate
primers. Alternative ORs denominations are given in brackets.
« P » indicates pseudogenes.
(DOCX)
Acknowledgments
We thank Elena Legenre for her contribution to this work during a training
period, Patrice Congar (INRA, UR1197, Jouy-en-Josas) for his help in
collecting single BON cells, Christophe Calvet (CNRS, UMR8203, Institut
Gustave-Roussy) for taking part in mice treatment and tumor growth
evaluation, and Maryline Le Me´e (CNRS, TAAM-CIPA, UPS44, Orle´ans)
for performing mice necropsies. We also thank the staff of the Service
Commun d’Expe´rimentation Animale de Gustave Roussy for mice feeding
and nursing. This work was partly performed in the scope of the European
Associated Laboratory EBAM.
Author Contributions
Conceived and designed the experiments: GS IL JS SL SB LMM.
Performed the experiments: GS IL AD JS SB DG RM. Analyzed the data:
GS IL JS SL LMM. Wrote the paper: GS IL JS SL EPA LMM. Obtained
permission for use of cell line: EPA.
References
1. Buck L, Axel R (1991) A novel multigene family may encode odorant receptors:
a molecular basis for odor recognition. Cell 65: 175–187.
2. Malnic B, Hirono J, Sato T, Buck LB (1999) Combinatorial receptor codes for
odors. Cell 96: 713–723.
3. Zhang X, Rogers M, Tian H, Zhang X, Zou DJ, et al. (2004) High-throughput
microarray detection of olfactory receptor gene expression in the mouse. Proc
Natl Acad Sci U S A 101: 14168–14173.
4. Zhang X, De la Cruz O, Pinto JM, Nicolae D, Firestein S, et al. (2007)
Characterizing the expression of the human olfactory receptor gene family using
a novel DNA microarray. Genome Biol 8: R86.
5. Flegel C, Manteniotis S, Osthold S, Hatt H, Gisselmann G (2013) Expression
profile of ectopic olfactory receptors determined by deep sequencing. PLoS One
8: e55368.
6. Spehr M, Schwane K, Heilmann S, Gisselmann G, Hummel T, et al. (2004)
Dual capacity of a human olfactory receptor. Curr Biol 14: R832–R833.
7. Fukuda N, Touhara K (2006) Developmental expression patterns of testicular
olfactory receptor genes during mouse spermatogenesis. Genes Cells 11: 71–81.
8. Braun T, Voland P, Kunz L, Prinz C, Gratzl M (2007) Enterochromaffin cells of
the human gut: sensors for spices and odorants. Gastroenterology 132: 1890–
1901.
9. Kidd M, Modlin IM, Gustafsson BI, Drozdov I, Hauso O, et al. (2008) Luminal
regulation of normal and neoplastic human EC cell serotonin release is mediated
by bile salts, amines, tastants, and olfactants. Am J Physiol Gastrointest Liver
Physiol 295: G260–272.
10. Griffin CA, Kafadar KA, Pavlath GK (2009) MOR23 promotes muscle
regeneration and regulates cell adhesion and migration. Dev Cell 17: 649–661.
11. Weng J, Wang J, Cai Y, Stafford LJ, Mitchell D, et al. (2005) Increased
expression of prostate-specific G-protein-coupled receptor in human prostate
intraepithelial neoplasia and prostate cancers. Int J Cancer 113: 811–818.
12. Neuhaus EM, Zhang W, Gelis L, Deng Y, Noldus J, et al. (2009) Activation of an
olfactory receptor inhibits proliferation of prostate cancer cells. J Biol Chem 284:
16218–16225.
13. Cui T, Tsolakis AV, Li SC, Cunningham JL, Lind T, et al. (2013) Olfactory
receptor 51E1 protein as a potential novel tissue biomarker for small intestine
neuroendocrine carcinomas. Eur J Endocrinol 168: 253–261.
14. Giandomenico V, Cui T, Grimelius L, Oberg KE, Pelosi G, et al. (2013)
Olfactory Receptor 51E1 as a Novel Target in Somatostatin Receptor Negative
Lung Carcinoids. J Mol Endocrinol. PMID: 23969981
15. Leja J, Essaghir A, Essand M, Wester K, Oberg K, et al. (2009) Novel markers
for enterochromaffin cells and gastrointestinal neuroendocrine carcinomas. Mod
Pathol 22: 261–272.
16. Evers BM, Ishizuka J, Townsend CM, Jr., Thompson JC (1994) The human
carcinoid cell line, BON. A model system for the study of carcinoid tumors. Ann
N Y Acad Sci 733: 393–406.
17. von Mentzer B, Murata Y, Ahlstedt I, Lindstrom E, Martinez V (2007)
Functional CRF receptors in BON cells stimulate serotonin release. Biochem
Pharmacol 73: 805–813.
18. Sanz G, Schlegel C, Pernollet JC, Briand L (2005) Comparison of odorant
specificity of two human olfactory receptors from different phylogenetic classes
and evidence for antagonism. Chem Senses 30: 69–80.
19. De Wever O, Hendrix A, De Boeck A, Westbroek W, Braems G, et al. (2010)
Modeling and quantification of cancer cell invasion through collagen type I
matrices. Int J Dev Biol 54: 887–896.
20. Saito H, Kubota M, Roberts RW, Chi Q, Matsunami H (2004) RTP family
members induce functional expression of mammalian odorant receptors. Cell
119: 679–691.
21. Wetzel CH, Oles M, Wellerdieck C, Kuczkowiak M, Gisselmann G, et al. (199 9)
Specificity and sensitivity of a human olfactory receptor functionally expressed in
human embryonic kydney 293 cells and Xenopus laevis oocytes. J Neurosci 19:
7426–7433.
22. Sanz G, Thomas-Danguin T, Hamdani EH, Le Poupon C, Briand L, et al.
(2008) Relationships Between Molecular Structure and Perceived Odor Quality
of Ligands for a Human Olfactory Receptor. Chem Senses 33: 639–653.
23. Attoub S, De Wever O, Bruyneel E, Mareel M, Gespach C (2008) The
transforming functions of PI3-kinase-gamma are linked to disruption of
intercellular adhesion and promotion of cancer cell invasion. Ann N Y Acad
Sci 1138: 204–213.
24. Brunert D, Klasen K, Corey EA, Ache BW (2010) PI3Kgamma-dependent
signaling in mouse olfactory receptor neurons. Chem Senses 35: 301–308.
25. Ukhanov K, Brunert D, Corey EA, Ache BW (2011) Phosphoinositide 3-kinase-
dependent antagonism in Mammalian olfactory receptor neurons. J Neurosci 31:
273–280.
26. Camps M, Ruckle T, Ji H, Ardissone V, Rintelen F, et al. (2005) Blockade of
PI3Kgamma suppresses joint inflammation and damage in mouse models of
rheumatoid arthritis. Nat Med 11: 936–943.
27. Vanhaesebroeck B, Leevers SJ, Ahmadi K, Timms J, Katso R, et al. (2001)
Synthesis and function of 3-phosphorylated inositol lipids. Annu Rev Biochem
70: 535–602.
28. Lin Y, Smrcka AV (2011) Understanding molecular recognition by G protein
betagamma subunits on the path to pharmacological targeting. Mol Pharmacol
80: 551–557.
29. Podolsky DK (2002) Inflammatory bowel disease. N Engl J Med 347: 417–429.
30. van der Horst-Schrivers AN, Wymenga AN, Links TP, Willemse PH, Kema IP,
et al. (2004) Complications of midgut carcinoid tumors and carcinoid syndrome.
Neuroendocrinology 80 Suppl 1: 28–32.
31. Gershon MD (2004) Review article: serotonin receptors and transporters
roles in normal and abnormal gastrointestinal motility. Aliment Pharmacol Ther
20 Suppl 7: 3–14.
32. Modlin IM, Kidd M, Latich I, Zikusoka MN, Shapiro MD (2005) Current status
of gastrointestinal carcinoids. Gastroenterology 128: 1717–1751.
33. Saito H, Chi Q, Zhuang H, Matsunami H, Mainland JD (2009) Odor coding by
a Mammalian receptor repertoire. Sci Signal 2: ra9.
34. Zhuang H, Matsunami H (2007) Synergism of accessory factors in functional
expression of mammalian odorant receptors. J Biol Chem 282: 15284–15293.
35. Zhang X, Bedigian AV, Wang W, Eggert US (2012) G protein-coupled
receptors participate in cytokinesis. Cytoskeleton (Hoboken) 69: 810–818.
36. Regnauld K, Nguyen QD, Vakaet L, Bruyneel E, Launay JM, et al. (2002) G-
protein alpha(olf) subunit promotes cellular invasion, survival, and neuroendo-
crine differentiation in digestive and urogenital epithelial cells. Oncogene 21:
4020–4031.
37. Sato N, Gleave ME, Bruchovsky N, Rennie PS, Beraldi E, et al. (1997) A
metastatic and androgen-sensitive human prostate cancer model using
intraprostatic inoculation of LNCaP cells in SCID mice. Cancer Res 57:
1584–1589.
Olfactory Receptors Promote Tumor Cell Invasion
PLOS ONE | www.plosone.org 9 January 2014 | Volume 9 | Issue 1 | e85110
38. Thalmann GN, Sikes RA, Wu TT, Degeorges A, Chang SM, et al. (2000)
LNCaP progression model of human prostate cancer: androgen-independence
and osseous metastasis. The Prostate 44: 91–103.
39. Scatena CD, Hepner MA, Oei YA, Dusich JM, Yu SF, et al. (2004) Imaging of
bioluminescent LNC aP-luc-M6 tumors: a new animal model for the study of
metastatic human prostate cancer. The Prostate 59: 292–303.
40. Wei P, Tang H, Li D (2012) Insights into pancreatic cancer etiology from
pathway analysis of genome-wide association study data. PLoS One 7: e46887.
41. Muranen TA, Greco D, Fagerholm R, Kilpivaara O, Kampjarvi K, et al. (2011)
Breast tumors from CHEK2 1100delC-mutation carriers: genomic landscape
and clinical implications. Breast Cancer Res 13: R90.
Olfactory Receptors Promote Tumor Cell Invasion
PLOS ONE | www.plosone.org 10 January 2014 | Volume 9 | Issue 1 | e85110
... ORs play essential roles in cell proliferation, apoptosis, migration, and development, but their abnormal expression is closely related to the proceeding of cancer [5,8,41]. However, its function remains largely unknown. ...
Article
Full-text available
Olfactory receptors (ORs), the largest family of G protein-coupled receptors (GPCRs), are ectopically expressed in cancer cells and are involved in cellular physiological processes, but their function as anticancer targets is still potential. OR2AT4 is expressed in leukemia cells, influencing the proliferation and apoptosis, yet the limited number of known OR2AT4 agonists makes it challenging to fully generalize the receptor's function. In this study, we aimed to identify new ligands for OR2AT4 and to investigate their functions and mechanisms in K562 leukemia cells. After producing the recombinant OR2AT4 protein, immobilizing it on a surface plasmon resonance chip, and conducting screening to confirm binding activity using 258 chemicals, five novel OR2AT4 ligands were discovered. As a result of examining changes in intracellular calcium by five ligands in OR2AT4-expressing cells and K562 cells, (−)-epigallocatechin gallate (EGCG) was identified as an OR2AT4 agonist in both cells. EGCG reduced the viability of K562 cells and induced apoptosis in K562 cells. EGCG increased the expression of cleaved caspase 3/8 and had no effect on the expression of Bax and Bcl-2, indicating that it induced apoptosis through the extrinsic pathway. Additionally, the initiation of the extrinsic apoptosis pathway in EGCG-induced K562 cells was due to the activation of OR2AT4, using an OR2AT4 antagonist. This study highlights the potential of EGCG as an anti-cancer agent against leukemia and OR2AT4 as a target, making it a new anti-cancer drug.
... Peculiar features once thought to characterize H. glaber, such as unusual longevity and cancer resistance, have since been expanded to other bathyergids such as Fukomys (Dammann et al. 2011) and even convergently evolved in Spalax species (Gorbunova et al. 2012;Manov et al. 2013). As it is now established that OR genes may be expressed outside of the olfactory epithelium and some of them were even shown to either promote or inhibit tumor formation (Sanz et al. 2014;2016;Kalbe et al. 2017), OR genes may even prove to be interesting candidates to metabolic adaptations of the underground lifestyle. ...
Article
Full-text available
Olfactory receptor (OR) genes represent the largest multigenic family in mammalian genomes and encode proteins that bind environmental odorant molecules. The OR repertoire is extremely variable among species and is subject to many gene duplications and losses, which have been linked to ecological adaptations in mammals. Although they have been studied on a broad taxonomic scale (i.e., placental), finer sampling has rarely been explored in order to better capture the mechanisms that drove the evolution of the OR repertoire. Among placental mammals, rodents are well-suited for this task, as they exhibit diverse life history traits, and genomic data are available for most major families and a diverse array of lifestyles. In this study, 53 rodent published genomes were mined for their OR subgenomes. We retrieved more than 85,000 functional and pseudogene OR sequences that were subsequently classified into phylogenetic clusters. Copy number variation among rodents is similar to that of other mammals. Using our OR counts along with comparative phylogenetic approaches, we demonstrated that ecological niches such as diet, period of activity, and a fossorial lifestyle strongly impacted the proportion of OR pseudogenes. Within the OR subgenome, phylogenetic inertia was the main factor explaining the relative variations of the 13 OR gene families. However, a striking exception was a convergent 10-fold expansion of the OR family 14 among the phylogenetically divergent subterranean mole-rat lineages belonging to Bathyergidae and Spalacidae families. This study illustrates how the diversity of the OR repertoire has evolved among rodents, both shaped by selective forces stemming from species life history traits and neutral evolution along the rodent phylogeny.
... Previous data from the literature [21,22,[67][68][69][70][71] have demonstrated that the activation of different ORs expressed in cancer cell lines may influence tumor growth, affecting cell proliferation and, in some cases, cell death. Therefore, we treated two of the analyzed AML cell lines with odorants (chemical compounds that can be detected by ORs) and evaluated if the total number of living cells was affected. ...
Article
Full-text available
Simple Summary Acute myeloid leukemia (AML) is a type of cancer that affects blood cells and is the most common type of acute leukemia in adults. Despite advances in treatment, many patients with AML still face poor outcomes, including a high risk of relapse. Therefore, new ways to treat AML are needed. In this study, we have discovered a group of genes called olfactory receptors (ORs) that are highly expressed in AML cells. ORs are normally found in the nose and are responsible for detecting smells, and are not commonly found in other tissues or parts of the body. Here, we show that 19 ORs were predominantly found in AML cells and that the expression of these ORs could predict the prognosis of AML patients. In summary, we believe that these ORs could be further investigated to be used for diagnosis and as targets for new drugs to treat AML. Abstract Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults, with a 5-year overall survival rate of approximately 30%. Despite recent advances in therapeutic options, relapse remains the leading cause of death and poor survival outcomes. New drugs benefit specific small subgroups of patients with actionable therapeutic targets. Thus, finding new targets with greater applicability should be pursued. Olfactory receptors (ORs) are seven transmembrane G-protein coupled receptors preferentially expressed in sensory neurons with a critical role in recognizing odorant molecules. Recent studies have revealed ectopic expression and putative function of ORs in nonolfactory tissues and pathologies, including AML. Here, we investigated OR expression in 151 AML samples, 6400 samples of 15 other cancer types, and 11,200 samples of 51 types of healthy tissues. First, we identified 19 ORs with a distinct and major expression pattern in AML, which were experimentally validated by RT-PCR in an independent set of 13 AML samples, 13 healthy donors, and 8 leukemia cell lines. We also identified an OR signature with prognostic potential for AML patients. Finally, we found cancer-related genes coexpressed with the ORs in the AML samples. In summary, we conducted an extensive study to identify ORs that can be used as novel biomarkers for the diagnosis of AML and as potential drug targets.
... In addition to the olfactory epithelium, ORs are expressed in a number of nonolfactory tissues and cancerous tissues including PCa (46)(47)(48)(49)(50)(51). Ectopic overexpression of ORs can promote PCa progression, metastasis, and neuroendocrine differentiation (52)(53)(54)(55). ...
Article
Full-text available
Castration-resistant prostate cancer (CRPC) poses a major clinical challenge with the androgen receptor (AR) remaining to be a critical oncogenic player. Several lines of evidence indicate that AR induces a distinct transcriptional program after androgen deprivation in CRPCs. However, the mechanism triggering AR binding to a distinct set of genomic loci in CRPC and how it promotes CRPC development remain unclear. We demonstrate here that atypical ubiquitination of AR mediated by an E3 ubiquitin ligase TRAF4 plays an important role in this process. TRAF4 is highly expressed in CRPCs and promotes CRPC development. It mediates K27-linked ubiquitination at the C-terminal tail of AR and increases its association with the pioneer factor FOXA1. Consequently, AR binds to a distinct set of genomic loci enriched with FOXA1- and HOXB13-binding motifs to drive different transcriptional programs including an olfactory transduction pathway. Through the surprising upregulation of olfactory receptor gene transcription, TRAF4 increases intracellular cAMP levels and boosts E2F transcription factor activity to promote cell proliferation under androgen deprivation conditions. Altogether, these findings reveal a posttranslational mechanism driving AR-regulated transcriptional reprogramming to provide survival advantages for prostate cancer cells under castration conditions.
... Data have documented that various olfactory receptors are expressed outside the sinonasal epithelium and may be present in hepatocellular carcinoma, breast carcinoma, prostatic adenocarcinoma, bladder carcinoma, and melanoma. [45][46][47][48][49][50][51][52][53][54][55] These receptors not only primarily activate GPCR signaling cascades but can also activate the MAPK and PI3K-AKT signaling pathways. 45,46,55 Functionally, they regulate cancer cell cycle progression, apoptosis, differentiation, and invasion and appear promising as novel therapeutic targets. ...
Article
Kaposi sarcoma (KS) is an HHV8-associated vascular proliferation that most often involves the skin. Rarely, KS shows marked nuclear atypia or pleomorphism; such examples are known as "anaplastic" KS. This poorly characterized variant often pursues an aggressive course; little is known of its genetic landscape. This study evaluated the clinicopathologic and genomic features of anaplastic KS. We identified 9 anaplastic KS cases from 7 patients and 8 conventional KS cases, including a matched conventional KS and primary-metastasis anaplastic KS pair from a single patient (anaplastic KS diagnosed 9 years after conventional KS). All patients with anaplastic KS were male, aged 51-82 years, who had locally aggressive tumors predominantly affecting the soft tissue and bone of the lower extremities (5/7). Four patients were known to be HIV-positive (all on antiretrovirals), 2 were HIV-negative, and one was of unknown HIV status. The tumors showed angiosarcoma-like or pleomorphic spindle cell sarcoma morphology. Plasma cell-rich chronic inflammation and hemosiderin deposition were commonly present. Single nucleotide polymorphism (SNP) based chromosomal microarray analysis showed the anaplastic KS cohort to demonstrate highly recurrent whole chromosome (chr) gains of chr 7, 11, 19, and 21, which primarily affected olfactory and G-protein coupled receptor (GPCR) signaling, and losses of chr6_q and chrY. Compared to conventional KS, anaplastic KS cases showed significantly more total copy number alterations (CNAs) and more frequent gains of chr7 and chr11_q13.1 (MARK2, RELA, and ESRRA; including high copy number gain in 1 case). Pathway analysis demonstrated these gains preferentially affected genes that facilitate cyclin-dependent cell signaling. Further, anaplastic KS cases were phylogenetically distinct from conventional KS cases, including the patient-matched primary-metastasis anaplastic KS pair and conventional KS. Our study is the first to demonstrate that a more complex genome and distinct CNAs distinguish anaplastic KS from conventional KS. Gains of chr7 and chr11_q13.1 appear central to biological transformation.
... The GO terms were enriched in several important molecular mechanisms, such as cotranslational protein targeting to membrane and ER, focal adhesion, and olfactory receptor activity. These pathways are considered to be closely related to tumors and metastasis (37)(38)(39). The main KEGG pathways involved included neuroactive ligand-receptor interactions, which were found to have prominent roles in adapting to the target organ environment in BM (40). ...
Article
Full-text available
Introduction Regarding whether brain magnetic resonance imaging (MRI) should be routine in patients with suspected early-stage lung cancer, guideline recommendations are inconsistent. Therefore, we performed this study to evaluate the incidence of and risk factors for brain metastasis (BM) in patients with suspected early-stage non-small-cell lung cancer (NSCLC). Methods A review of the medical charts of consecutive NSCLC patients diagnosed between January 2006 and May 2020 was performed. We identified 1,382 NSCLC patients with clinical staging of T1/2aN0M0 (excluding BM), and investigated the incidence, clinical predictors, and prognosis of BM in the cohort. We also performed RNA-sequencing differential expression analysis using transcriptome of 8 patients, using DESeq2 package (version 1.32.0) with R (version 4.1.0). Results Among 1,382 patients, nine hundred forty-nine patients (68.7%) underwent brain MRI during staging, and 34 patients (3.6%) were shown to have BM. Firth’s bias-reduced logistic regression showed that tumor size (OR 1.056; 95% CI 1.009-1.106, p=0.018) was the only predictor of BM, and pathologic type was not a predictor of BM in our cohort (p>0.05). The median overall survival for patients with brain metastasis was 5.5 years, which is better than previously reported in the literature. RNA-sequencing differential expression analysis revealed the top 10 significantly upregulated genes and top 10 significantly downregulated genes. Among the genes involved in BM, Unc-79 homolog, non-selective sodium leak channel (NALCN) channel complex subunit (UNC79) was the most highly expressed gene in the lung adenocarcinoma tissues from the BM group, and an in vitro assay using A549 cells revealed that the NALCN inhibitor suppressed lung cancer cell proliferation and migration. Conclusions Given the incidence and favorable outcome of BM in patients with suspected early-stage NSCLC, selective screening with brain MRI may be considered, especially in patients with high-risk features.
... On the basis of receptor family analysis, we hypothesized that b-ionone is an odorous ligand of OR51A7. It was previously shown that OR51E2 is expressed in cancer cells and that b-ionone promotes cancer cell migration through OR51E2, indicating that b-ionone is an OR51E2 ligand (Gelis et al., 2017;Sanz et al., 2014). In this study, the HEK293T cellÀbased shedding assay system using AP-TGFaÀ and G-proteinÀcontaining plasmids confirmed the recognition of b-ionone by OR51E2 in the presence of Gaq/il and/or Golf. ...
Article
Full-text available
Sweat maintains systemic homeostasis in humans. Although sweating disorders may cause multifaceted health problems, therapeutic options for sweat disorders have not yet been established. To gain new insight into the mechanism underlying the regulation of perspiration, we compared eccrine sweat gland transcriptomes from hidrotic and anhidrotic lesions from patients with anhidrosis and found out that olfactory receptors were expressed differentially in anhidrotic and hidrotic eccrine sweat glands. We then confirmed OR51A7 and OR51E2 expression in human eccrine sweat glands by in situ hybridization and immunohistochemistry. An alkaline phosphatase−TGFα shedding assay revealed that β-ionone activates G-proteins through OR51A7 or OR51E2. The effect of topically applied β-ionone on sweating was examined with the quantitative sudomotor axon reflex test, which showed that responses to β-ionone differed between sexes. Topical β-ionone attenuated female sweating and augmented male sweating. Taken together, this study suggests that olfactory receptors expressed in eccrine sweat glands may regulate sweating in response to odorous ligands on the basis of sex. These unexpected results indicate that olfactory receptors may modulate sweating and that olfactory receptor modulators may contribute to the management of sweat disorders.
... Olfactory receptors are expressed in and associated with many tumors [348][349][350][351][352]. OR15E2 was found in melanocytes and its stimulation with β-ionone leads to melanogenesis and dendritogenesis via calcium and cAMP signaling [243]. ...
Thesis
Immune checkpoint blockade has revolutionized immunotherapy against cancer with tremendous clinical benefits for patients. Despite these achievements, tumors utilize a plethora of suppressive mechanisms to evade immune destruction which are yet to be understood and matched by today’s immunotherapy. Our group developed a high-throughput RNAi screening to unravel the arsenal of immune checkpoints of cancer. We screened a siRNA library (around 2880 kinases and surface-associated genes) with patient-derived tumor cells and HLA-matched tumor-infiltrating lymphocytes (TILs). The library was reverse-transfected into M579-A2 melanoma cells and these were co-cultured with MART1- and gp100-specific TILs to determine TIL-mediated lysis. We identified 75 genes in tumor cells that impaired TIL-mediated cytotoxicity. Interestingly, we found that several genes and their associated pathways were found in pancreatic adenocarcinoma and multiple myeloma as well. This suggests that different cancer entities might share inhibitory modes of action. In order to distinguish between genes altering tumor susceptibility towards TIL-mediated killing and those impairing TIL activity, we established a secondary screening assaying multiple T cell activation marker, including effector cytokines. The olfactory receptor OR10H1 was one of the strongest candidates from our primary screening as its knockdown increased TIL-mediated killing in melanoma, PDAC and colorectal carcinoma. Furthermore, TILs were activated stronger after interaction with OR10H1-deficient cells as sensed by the increased secretion of type 1-associated cytokines and a reduced T cell apoptosis. We confirmed the role of OR10H1 as an immune checkpoint in vivo using a xenograft mouse model in combination with adoptive T cell transfer. We performed mode of action analyses in order to understand how OR10H1 affects T cell activity. These analyses revealed that tumor-associated OR10H1 controls cAMP-dependent signaling inside T cells. Inside TILs, cAMP activates protein kinase alpha (PKA) and PKA in turn activates C-terminal Src kinase (Csk. Csk phosphorylates an inhibitory tyrosine residue of Lck impairing its activity and shutting down TCR-associated signaling. Furthermore, PKA activates CREB and thus induces an anergy-associated gene expression profile in TILs. Our data suggest that OR10H1 alters the balance between the inhibitory (GαI) and the stimulatory/olfactory G-Protein alpha (GαS/Olf) inside tumor cells depending on the encounter of TILs. This results into increased production of cAMP in tumor cells and its subsequent transport into T cells. In summary, we established a discovery platform aiding the search for immune checkpoints in cancer. We identified OR10H1 and its associated olfactory receptor signaling as a novel pathway inhibiting TIL responses by inducing cAMP-dependent Lck inhibition.
... An activated G-protein can co-localize with regulatory factor Resistance to inhibitors of cholinesterase 8B (Ric8B) in olfactory sensory neurons. Although there is no solid information for specific functional appearance, there are some limited proof and proposed data of G-protein subunit activation in cancer cells (Sanz et al., 2014). ORs require definite cofactors to develop membrane localization of receptors. ...
Article
Full-text available
Olfactory receptors (ORs) provide the molecular basis for the detection of volatile odorant molecules by olfactory sensory neurons. The OR supergene family encodes G-protein coupled proteins that belong to the seven-transmembrane-domain receptor family. It was initially postulated that ORs are exclusively expressed in the olfactory epithelium. However, recent studies have demonstrated ectopic expression of some ORs in a variety of other tissues. In the present study, we conducted a comprehensive expression analysis of ORs using an extended panel of human tissues. This analysis made use of recent dramatic technical developments of the so-called Next Generation Sequencing (NGS) technique, which encouraged us to use open access data for the first comprehensive RNA-Seq expression analysis of ectopically expressed ORs in multiple human tissues. We analyzed mRNA-Seq data obtained by Illumina sequencing of 16 human tissues available from Illumina Body Map project 2.0 and from an additional study of OR expression in testis. At least some ORs were expressed in all the tissues analyzed. In several tissues, we could detect broadly expressed ORs such as OR2W3 and OR51E1. We also identified ORs that showed exclusive expression in one investigated tissue, such as OR4N4 in testis. For some ORs, the coding exon was found to be part of a transcript of upstream genes. In total, 111 of 400 OR genes were expressed with an FPKM (fragments per kilobase of exon per million fragments mapped) higher than 0.1 in at least one tissue. For several ORs, mRNA expression was verified by RT-PCR. Our results support the idea that ORs are broadly expressed in a variety of tissues and provide the basis for further functional studies.
Article
Full-text available
Objective: Late diagnosis hinders proper management of small intestine neuroendocrine carcinoma (SI-NEC) patients. The olfactory receptor, family 51, subfamily E, member 1 (OR51E1) has been reported as a potential novel SI-NEC marker, without protein expression recognition. Thus, we further studied whether the encoded protein may be a novel SI-NEC clinical biomarker. Design: OR51E1 coding sequence was cloned using total RNA from SI-NEC patient specimens. Quantitative real-time PCR analysis explored OR51E1 expression in laser capture microdissected SI-NEC cells and adjacent microenvironment cells. Moreover, immunohistochemistry investigated OR51E1 protein expression on operation and biopsy material from primary SI-NECs, mesentery, and liver metastases from 70 patients. Furthermore, double immunofluorescence studies explored the potential co-localization of the vesicular monoamine transporter 1 (SLC18A1, generally referred to as VMAT1) and OR51E1 in the neoplastic cells and in the intestinal mucosa adjacent to the tumor. Results: OR51E1 coding sequence analysis showed absence of mutation in SI-NEC patients at different stages of disease. OR51E1 expression was higher in microdissected SI-NEC cells than in the adjacent microenvironment cells. Furthermore, both membranous and cytoplasmic OR51E1 immunostaining patterns were detected in both primary SI-NECs and metastases. Briefly, 18/43 primary tumors, 7/28 mesentery metastases, and 6/18 liver metastases were 'positive' for OR51E1 in more than 50% of the tumor cells. In addition, co-localization studies showed that OR51E1 was expressed in >50% of the VMAT1 immunoreactive tumor cells and of the enterochromaffin cells in the intestinal mucosa adjacent to the tumor. Conclusion: OR51E1 protein is a potential novel clinical tissue biomarker for SI-NECs. Moreover, we suggest its potential therapeutic molecular target development using solid tumor radioimmunotherapy.
Article
Full-text available
Pancreatic cancer is the fourth leading cause of cancer death in the U.S. and the etiology of this highly lethal disease has not been well defined. To identify genetic susceptibility factors for pancreatic cancer, we conducted pathway analysis of genome-wide association study (GWAS) data in 3,141 pancreatic cancer patients and 3,367 controls with European ancestry. Using the gene set ridge regression in association studies (GRASS) method, we analyzed 197 pathways identified from the Kyoto Encyclopedia of Genes and Genomes database. We used the logistic kernel machine (LKM) test to identify major contributing genes to each pathway. We conducted functional enrichment analysis of the most significant genes (P<0.01) using the Database for Annotation, Visualization, and Integrated Discovery (DAVID). Two pathways were significantly associated with risk of pancreatic cancer after adjusting for multiple comparisons (P<0.00025) and in replication testing: neuroactive ligand-receptor interaction, (Ps<0.00002), and the olfactory transduction pathway (P = 0.0001). LKM test identified four genes that were significantly associated with risk of pancreatic cancer after Bonferroni correction (P<1×10(-5)): ABO, HNF1A, OR13C4, and SHH. Functional enrichment analysis using DAVID consistently found the G protein-coupled receptor signaling pathway (including both neuroactive ligand-receptor interaction and olfactory transduction pathways) to be the most significant pathway for pancreatic cancer risk in this study population. These novel findings provide new perspectives on genetic susceptibility to and molecular mechanisms of pancreatic cancer.
Article
BACKGROUND Clinically, the lethal phenotypes of human prostate cancer are characterized by their progression to androgen‐independence and their propensity to form osseous metastases. We reported previously on the establishment of androgen‐independent (AI) human prostate cancer cell lines derived from androgen‐dependent (AD) LNCaP cells, with androgen independence defined as the capability of prostate cancer cells to grow in castrated hosts. One of the sublines, C4‐2, was found to be AI, highly tumorigenic, and metastatic, having a proclivity for metastasis to the bone. METHODS We established the AI and bone metastatic cell sublines B2, B3, B4, and B5 from the parental C4‐2 subline, using a previously established coinoculating procedure. We determined the biologic behavior of the parental and derivative LNCaP sublines in vivo and in vitro, as well as their molecular and cytogenetic characteristics. RESULTS Unlike other human prostate cancer models, the LNCaP progression model shares remarkable similarities with human prostate cancer. We observed a comparable pattern of metastasis from the primary to the lymph node and to the axial skeleton, with a predominant phenotype of osteoblastic reaction; 25–37.5% of the animals developed paraplegia. Cytogenetic and biochemical characterizations of LNCaP sublines also indicate close similarities between human prostate cancer and the LNCaP progression model. Additional chromosomal changes were detected in B2–B5 sublines derived from C4‐2 bone metastases. These LNCaP sublines were found to grow faster under anchorage‐dependent but not ‐independent conditions. The in vitro invasion and in vivo metastatic potential of these LNCaP sublines surprisingly correlated with anchorage‐dependent and not ‐independent growth. The derivative LNCaP sublines when cultured in vitro produced a substantially higher (20–30‐fold) amount of basal steady‐state concentrations of PSA than that of the parental LNCaP cells. PSA production was high initially, but was markedly reduced when the derivative cell lines were inoculated and allowed to grow long‐term in vivo for the establishment of tumors and metastasis, suggesting that unknown host factors derived either from the prostate or the bone can effectively downregulate PSA expression by prostate tumor epithelium. CONCLUSIONS The LNCaP model of human prostate cancer progression will help improve our understanding of the mechanisms of androgen‐independence and osseous metastasis, and tumor‐host determinants of PSA expression. Prostate 44:91–103, 2000. © 2000 Wiley‐Liss, Inc.
Article
In the decades since the major forms of idiopathic inflammatory bowel disease were defined on the basis of clinical manifestations, investigators have been challenged to identify the fundamental pathophysiologic processes underlying these enigmatic disorders, and clinicians have struggled to provide effective therapy for the often dismaying clinical manifestations. Clinical experience has led to the generally accepted notion that Crohn's disease and ulcerative colitis are distinct, if not discrete, entities. However, whether these are fundamentally different diseases or part of a mechanistic continuum remains an unanswered question, with both conceptual and practical management implications.
Article
Somatostatin receptors (SSTRs) may be used in lung carcinoids (LCs) for diagnosis and therapy, although additional targets are clearly warranted. This study aimed to investigate whether olfactory receptor 51E1 (OR51E1) may be a potential target for LCs. OR51E1 coding sequence was analyzed in LC cell lines, NCI-H727 and NCI-H720. OR51E1 transcript expression was investigated in LC cell lines and frozen specimens by quantitative real-time PCR. OR51E1, SSTR2, SSTR3, and SSTR5 expression was evaluated by immunohistochemistry on paraffin-embedded sections of 73 typical carcinoids (TCs), 14 atypical carcinoids (ACs) and 11 regional/distant metastases, and compared to OctreoScan data. Immunohistochemistry results were rendered semiquantitatively on a scale from 0 to 3+, taking into account the cellular compartmentalization (membrane vs. cytoplasm) and the percentage of tumor cells (<50% vs. >50%). Our results showed that wild-type OR51E1 transcript was expressed in both LC cell lines. OR51E1 mRNA was expressed in 9/12 TCs and 7/9 ACs (p=NS). Immunohistochemically, OR51E1, SSTR2, SSTR3 and SSTR5 were detected in 85%, 71%, 25% and 39% of TCs, and in 86%, 79%, 43% and 36% of ACs, respectively. OR51E1 immunohistochemical scores were higher or equal compared to SSTRs in 79% of TCs and 86% of ACs. Furthermore, in the LC cases where all SSTR subtypes were lacking, membrane OR51E1 expression was detected in 10/17 TCs and 1/2 ACs. Moreover, higher OR51E1 immunohistochemical scores were detected in 5/6 OctreoScan-negative LC lesions. Therefore, the high expression of OR51E1 in LCs makes it a potential novel diagnostic target in SSTR-negative tumors.
Article
Cytokinesis, the last step during cell division, is a highly coordinated process that involves the relay of signals from both the outside and inside of the cell. We have a basic understanding of how cells regulate internal events, but how cells respond to extracellular cues is less explored. In a systematic RNAi screen of G protein-coupled receptors (GPCRs) and their effectors, we found that some GPCRs are involved in cytokinesis. RNAi knockdown of these GPCRs caused increased binucleated cell formation, and live cell imaging showed that most formed midbodies but failed at the abscission stage. OR2A4 (olfactory receptor, family 2, subfamily A, member 4) localized to cytokinetic structures in cells and its knockdown caused cytokinesis failure at an earlier stage, likely due to effects on the actin cytoskeleton. Identifying the downstream components that transmit GPCR signals during cytokinesis will be the next step and we show that GIPC1 (GIPC PDZ domain containing family, member 1), an adaptor protein for GPCRs, may play a part. RNAi knockdown of GIPC1 significantly increased binucleated cell formation. Understanding the molecular details of GPCRs and their interaction proteins in cytokinesis regulation will give us important clues about GPCRs signaling as well as how cells communicate with their environment during division. © 2012 Wiley Periodicals, Inc.
Article
Clinically, the lethal phenotypes of human prostate cancer are characterized by their progression to androgen-independence and their propensity to form osseous metastases. We reported previously on the establishment of androgen-independent (AI) human prostate cancer cell lines derived from androgen-dependent (AD) LNCaP cells, with androgen independence defined as the capability of prostate cancer cells to grow in castrated hosts. One of the sublines, C4-2, was found to be AI, highly tumorigenic, and metastatic, having a proclivity for metastasis to the bone.