ArticlePDF Available

Abstract and Figures

Cell migration is regulated by adhesion to the extracellular matrix (ECM) through integrins and activation of small RhoGTPases, such as RhoA and Rac1, resulting in changes to actomyosin organization. During invasion, epithelial-derived tumor cells switch from laminin-enriched basal membrane to collagen and fibronectin-enriched connective tissue. How this switch affects the tumor migration is still unclear. We tested the hypothesis that ECM dictates the invasiveness of Oral Squamous Cell Carcinoma (OSCC). We analyzed the migratory properties of two OSCC lines, a low invasive cell line with high e-cadherin levels (Linv/HE-cad) or a highly invasive cell line with low e-cadherin levels (Hinv/LE-cad), plated on different ECM components. Compared to laminin, fibronectin induced non-directional collective migration and decreased RhoA activity in Linv/HE-cad OSCC. For Hinv/LE-cad OSCC, fibronectin increased Rac1 activity and induced smaller adhesions, resulting in a fast single cell migration in both 2D and 3D environments. Consistent with these observations, human OSCC biopsies exhibited similar changes in cell-ECM adhesion distribution at the invasive front of the tumor, where cells encounter fibronectin. Our results indicate that ECM composition might induce a switch from collective to single cell migration according to tumor invasiveness due to changes in cell-ECM adhesion and the resulting signaling pathways that alter actomyosin organization.
Content may be subject to copyright.
RESEARCH ARTICLE
Fibronectin Modulates Cell Adhesion and
Signaling to Promote Single Cell Migration of
Highly Invasive Oral Squamous Cell
Carcinoma
Grasieli de Oliveira Ramos
1,2
, Lisiane Bernardi
1
, Isabel Lauxen
1
, Manoel SantAna Filho
1,2
,
Alan Rick Horwitz
3
, Marcelo Lazzaron Lamers
1,2,4
*
1Basic Research Center, Dentistry School, Federal University of Rio Grande of Sul, Porto Alegre, Rio
Grande do Sul, Brazil, 2Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil,
3Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America,
4Department of Morphological Sciences, Institute of Basic Health Sciences, Federal University of Rio
Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
*marcelo.lamers@ufrgs.br
Abstract
Cell migration is regulated by adhesion to the extracellular matrix (ECM) through integrins
and activation of small RhoGTPases, such as RhoA and Rac1, resulting in changes to acto-
myosin organization. During invasion, epithelial-derived tumor cells switch from laminin-
enriched basal membrane to collagen and fibronectin-enriched connective tissue. How this
switch affects the tumor migration is still unclear. We tested the hypothesis that ECM dictates
the invasiveness of Oral Squamous Cell Carcinoma (OSCC). We analyzed the migratory
properties of two OSCC lines, a low invasive cell line with high e-cadherin levels (L
inv
/H
E-cad
)
or a highly invasive cell line with low e-cadherin levels (H
inv
/L
E-cad
), plated on different ECM
components. Compared to laminin, fibronectin induced non-directional collective migration
and decreased RhoA activity in L
inv
/H
E-cad
OSCC. For H
inv
/L
E-cad
OSCC, fibronectin
increased Rac1 activity and induced smaller adhesions, resulting in a fast single cell migra-
tion in both 2D and 3D environments. Consistent with these observations, human OSCC
biopsies exhibited similar changes in cell-ECM adhesion distribution at the invasive front of
the tumor, where cells encounter fibronectin. Our results indicate that ECM composition
might induce a switch from collective to single cell migration according to tumor invasiveness
due to changes in cell-ECM adhesion and the resulting signaling pathways that alter actomy-
osin organization.
Introduction
Oral squamous cell carcinoma (OSCC) is an epithelial neoplasm found in 8090% of head and
neck cancer [1]. OSCC can occur at several sites of the oral mucosa and is originated from
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 1/18
OPEN ACCESS
Citation: Ramos GdO, Bernardi L, Lauxen I,
SantAna Filho M, Horwitz AR, Lamers ML (2016)
Fibronectin Modulates Cell Adhesion and Signaling to
Promote Single Cell Migration of Highly Invasive Oral
Squamous Cell Carcinoma. PLoS ONE 11(3):
e0151338. doi:10.1371/journal.pone.0151338
Editor: Thomas Abraham, Pennsylvania State
Hershey College of Medicine, UNITED STATES
Received: March 5, 2015
Accepted: February 27, 2016
Published: March 15, 2016
Copyright: © 2016 Ramos et al. This is an open
access article 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.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Funding: This work was supported by Coordenação
de Pessoal de Nível Superior (CAPES, Brazil, #9748-
13-0, #202188/2014-0), Conselho Nacional de
Desenvolvimento Científico e Tecnológico (CNPq,
Brazil, #479527/2012-1, #443699/2014-3) and
National Institutes of Health (NIH, GM23244). FIPE/
HCPA - Fundo de Incentivo à Pesquisa e Eventos /
Hospital de Clínicas de Porto Alegre. The funders
had no role in study design, data collection and
genetically altered keratinocytes arising from exposure to a wide range of mutagenic agents [2].
Histopathologically, OSCC lesions are characterized by the presence of different degrees of
squamous differentiation, keratin production, nuclear pleomorphisms, mitotic activity, invasive
growth and metastasis. Despite advances in treatment, the OSCC prognosis remains poor with a
5 year survival rate of around 50%. This prognosis has not improved over the past several years
due to the development of distant metastasis, local recurrences and new tumors [1,3,4].
The ability of tumor cells to invade connective tissue is essential for them to access blood
vessels and ultimately promote distant metastasis. Both events, tissue invasion and metastasis,
are highly heterogeneous processes [5], requiring tumor cell adaptation to new environments
that alter the migratory mode. Depending on the tumor origin, differentiation level, and tumor
microenvironment, cancer cells migrate either as collective or single cells [6]. Amoeboid- and
mesenchymal-like single cell migration involve the coordinated interaction of structural and
signaling molecules that results in polymerization of actin at the leading edge, adhesion to the
extracellular matrix (ECM) through integrins, contraction of the cell cortex and detachment of
adhesions at the cell rear [7,8], whereas cluster or strand like collective cell migration involves
the single cell migration steps associated with the presence of cell-cell contacts, mainly medi-
ated by cadherin family members [6,9]. Rho family GTPases orchestrates changes in actomyo-
sin organization that drive these key events in cell migration. For example, Rac1 regulates actin
filament nucleation associated with nascent adhesion formation, and RhoA controls cell con-
tractility, actin elongation and adhesion maturation [7,10]. Changes in RhoGTPase activation
levels interfere with the balance between cell-cell and cell-ECM adhesions and likely influences
collective vs single cell migration [1013].
Tumor formation is sensitive to the microenvironment, which varies by the region of the
tumor. The tumor microenvironment is characterized by intense angiogenesis, high concentra-
tions of growth factors and inflammatory cytokines, and ECM remodeling [14,15]. An abrupt
adaptation occurs during invasion of epithelial-derived tumors when they move from the basal
membrane, a laminin enriched environment, to the connective tissue region, which is rich in
collagen and fibronectin [16,17]. Oral squamous cell carcinoma biopsies exhibit decreased
laminin content and increased fibronectin, depending on the aggressiveness and the location of
the tumor [18,19]. It is likely that the characteristics of the tumor microenvironment, such as
the composition of the extracellular matrix, influence metastatic and invasive behavior due to
biochemical or physical activation of migration-related proteins and signaling pathways.
In this study, we report that the change from a laminin- to a fibronectin-rich environment
has a differential effect on the migration properties of OSCCs. In high invasive and low E-cad-
herin expressing OSCC cells (H
inv
/L
E-cad
), fibronectin induced a fast single cell migration phe-
notype that is associated with increased Rac1 activation levels and small cell-ECM adhesions;
in low invasive and high E-cadherin OSCC cells (L
inv
/H
E-cad
), fibronectin produces a collective,
non-directional migration, with high RhoA activity and altered cell-ECM adhesion. Consistent
with these results, human OSCC biopsies also demonstrated changes in cell-ECM and cell-cell
adhesion according to the tumor region. Together, these data show that the composition of the
extracellular matrix differentially affects cell-ECM adhesion, cell migration signaling pathways
and the migratory output of OSCC cells and that these effects vary according to the differentia-
tion level of the tumor.
Material and Methods
Human Biopsies and OSCC Cell Culture
The experimental design and the informed consent procedures were approved by the Ethical
Committee of Federal University of Rio Grande do SulBrazil and of Hospital de Clínicas de
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 2/18
analysis, decision to publish, or preparation of the
manuscript.
Competing Interests: The authors have declared
that no competing interests exist.
Porto AlegreBrazil (CAE#06397313.7.0000.5347) and all patients in this study provided writ-
ten informed consent. Patients (n = 10) with oral lesions were interviewed and submitted to
surgery; OSCC diagnosis was confirmed histopathologically by a pathologist and fragments
from regions corresponding to the center of the tumor and the carcinoma edge tissue, named
as tumor adjacent epithelia (TAE) were collected. OSCC cell lines were obtained from the Tis-
sue Culture Facility at School of Medicine of University of Virginia and checked for myco-
plasma by this facility. Cal27 cells (ATCC
1
CRL-2095) were cultivated in DMEM high
glucose (Gibco) supplemented with 10% Fetal Bovine Serum (FBS) (Gibco) while SCC25 cells
(ATCC
1
CRL-1628) in DMEM/F12 with 15mM HEPES and 0.5mM sodium pyruvate
(Gibco) supplemented with FBS 10% and hydrocortisone (400ng/ml, Sigma), and cells were
maintained in incubator (37°C, 5% CO
2
). Cal27 cells are considered low invasive OSCC cells
[20] with high E-cadherin levels (L
inv
/H
E-cad
), while SCC25 cells are highly invasive with low
E-cadherin levels (H
inv
/L
E-cad
). Spheroids were performed plating 5x10
4
cells in a 96 wells dish
covered with 1.5% agarose and, after 3 days, spheroids were gently collected and used for
experiments. For Total Internal Reflectance Fluorescence (TIRF) microscopy, cell lines (1x10
6
)
were nucleofected 24h before the experiment with 0.2μg Paxillin-GFP plasmid [21], using
Amaxa Nucleofection System (Lonza).
Experimental Conditions
Unless stated otherwise, all reagents were purchased from Sigma Aldrich. For 2D imaging
experiments, cells were trypsinized, washed and plated in glass-bottomed dishes covered with
fibronectin (2μg/ml), laminin (poly-l-lysine (1mg/ml) + laminin (2μg/ml)) or Matrigel
1
(50μl/
cm
2
, BD Bioscience) in the presence of CCM1 media (Hyclone, Thermo Scientific). For 3D
imaging experiments, it was used collagen (1.2mg/ml, rat tail collagen) matrices assembled
according to the manufacturer (Gibco) in the presence/absence of fibronectin (10μg/ml) or
laminin (10μg/ml). For each condition, a thin layer of the respective collagen matrix was ini-
tially plated at the surface of the glass-bottomed dishes. After polymerization, 3x10
4
cells or
spheroids were embedded in a new collagen matrix and, after 3h, imaged using CCM1 media.
To ensure that cells were in the 3D matrix, it was verified the lower and the upper focus with
detectable cells and it was always selected cells for imaging at an intermediate focus position.
Immunoblots
Antibodies were purchased from Cell Signaling (E-cadherin, N-cadherin, Integrins α4, α5, αv,
β1, β3), BD-Transduction (Paxillin, FAK) and Sigma (β-Tubulin, Vinculin). Cells (1x10
6
) were
trypsinized, washed and lysed in RIPA Buffer (25mM Tris-HCL pH 7.6, 150mM NaCl, 1% NP-
40, 1% sodium deoxycholate, 0.1% SDS) containing protease and phosphatase inhibitors cock-
tails. Cell lysates (20μg) were separated in 420% SDS Gels (Biorad) and proteins transferred
to PVDF membranes, blocked (4% BSA) and immunoassayed for E-cadherin, FAK, Paxilllin,
β-Tubulin, Vinculin or integrins (α4, α5, αv, β1, β3) using Pierce ECL Western Blotting Sub-
strate (Thermo Scientific). Densitometry of the bands was performed using ImageJ software
(http://rsb.info.nih.gov/ij), and values for each protein were normalized to the loading control.
Immunofluorescence
For tumor staining, human biopsies were fixed immediately after collection (4% formaldehyde,
4h, 4°C), cryoprotected with increasing sucrose concentrations (1030%, 4°C), embedded in
OCT compound, frozen (-20°C), cut using cryostat and seven μm-thick slices were collected in
gelatin-covered slides. For cell lines staining, L
inv
/H
E-cad
and H
inv
/L
E-cad
were plated in cover-
slips covered with fibronectin (2μg/ml) or poly-l-lysine (1mg/ml) + laminin (2μg/ml) in the
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 3/18
presence of CCM1 media. After 3h, cells were washed (PBS) and fixed (formaldehyde, 4%,
10min, RT). Fixed cells or human biopsies were permeabilized (Triton X-100 0.3%, RT
10 min), blocked (10% normal goat serum, RT, 1h), incubated with antibodies for E-cadherin,
FAK, Paxillin, Vinculin or Fibronectin (ON, 4°C), washed (PBS) and incubated (2h, RT) with
the corresponding secondary antibodies containing Alexa488 dye (Molecular Probes, Oregon,
USA). Actin filaments were stained with phalloidin toxin conjugated to rhodamine (Molecular
Probes, Oregon, USA) for 2h (RT). Samples were washed (PBS) and mounted with antifade
medium (Vectashield, VectorLab, Burlingame, CA). Images were obtained in confocal micro-
scope (Olympus Fluoview 1000, Tokyo, Japan) with a 63x objective (UPlanSApo x63, 1.20 NA,
oil immersion objective) using FV-1000 ASW Fluoview software (Olympus, Tokyo, Japan).
Alexa488 was excited with the 488nm laser line of an Argon ion laser (Melles Griot, Albuquer-
que, NM), while rhodamine with the 543nm laser line of a Helium-Neon laser (Melles Griot,
Albuquerque, NM). Z-stacks were obtained from cells (0.1μm step size) and biopsy slices
(0.5μm step size) with or without digital zoom (3x for cell lines; 5x for biopsies). In order to
analyze the whole adhesion and avoid image background, 3 confocal-obtained slices were
merged using the Z-stack/maximum projectiontool from the ImageJ software. This new
merged image corresponds to an equivalent 0.3μm or 1.5μm thick slice of the cell lines or the
biopsy samples, respectively. Besides brightness/contrast corrections, no further image editing
was performed and figures were prepared using Adobe Photoshop
1
7 software.
RhoGTPase Activity
For analysis of RhoGTPase activation, pull down assays [22] were performed. L
inv
/H
E-cad
and
H
inv
/L
E-cad
cells were plated in plastic dishes covered with fibronectin (2μg/ml) or poly-l-lysine
(1mg/ml) + laminin (2μg/ml) in the presence of CCM1 media. After 3h, cells were washed
(PBS), harvested, lysed with CRIBs buffer in the presence of protease and phosphatase inhibi-
tors and incubated in the presence of GST-PAK-CRIB (Rac1) or GST-RBD-CRIB (RhoA)
beads. After washing, samples were prepared for SDS-PAGE and submitted to immunoblotting
for Rac1 (BD Bioscience) or RhoA (Santa Cruz Biotechnologies). Densitometry of the bands
was performed using ImageJ software.
FRET Imaging and Analysis
L
inv
/H
E-cad
and H
inv
/L
E-cad
OSCC cells were nucleofected with Raichu-Rac1-WT or Raichu-
Rac1-V12 plasmids for Rac1 activity and RaichuRhoA-WT or Raichu-RhoA-Q63L plasmids
[23] for RhoA activity (0.5μg/10
6
cells) and 24h later were trypsinized and plated in plastic
dishes covered with fibronectin (2μg/ml) or poly-l-lysine (1mg/ml) + laminin (2μg/ml) in the
presence of CCM1 media. Cells were washed (PBS), fixed in formaldehyde 4% and sacarose 4%
(10 min, RT), washed (PBS) and analyzed by confocal microscopy with 2x digital zoom. Donor
probe was excited with the 458nm laser line of an Argon ion laser (Melles Griot, Albuquerque,
NM). Images were analyzed by Matlab
1
software (MathWorks, Natick, MA) using the Biosen-
sor Processing software 2.1 [24]. The mean intensity values from FRET-ratio TIFF images were
obtained on ImageJ software. Using ImageJ software, selected images were adjusted for the
same levels of brightness/contrast and a 0.5 pixel-wide Gausian filter was applied.
Migration and Adhesion Dynamics Assays
Imaging acquisition and analysis for migration assays were performed as previously described
[25]. For phase microscopy movies, images were captured at 10min intervals using a Nikon
TE300 microscope (10x 0.25 NA CFI Achro DL106 Nikon objective) with a charge coupled
device camera (Orca II, Hamamatsu Photonics) using Metamorph software (Molecular
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 4/18
Devices). For TIRF microscopy, images were taken at 3s intervals using an Olympus IX70
inverted microscope (63x 1.45 NA oil Olympus PlanAplo 660 TIRFM objective) fitted with a
Ludl modular automation controller (Ludl Electronic Products) with a charge-couple device
camera (Retiga Exi, Qimaging) and controlled by Metamorph software. GFP was excited with
the 488nm laser line of an Argon laser (Melles Griot) and a dichroic mirror (HQ485/30) and
an emission filter (HQ525/50) were used. All images and movies were analyzed using ImageJ
software and panels mounted using Adobe Photoshop
1
7 software. For analysis of migration
parameters, it was performed at least 4 independent experiments (phase contrast microscopy
movies) and the nucleus of each migratory cell was tracked using the manual trackingplug-
in on ImageJ. It was considered as migratory cell only cells that migrated for at least 6h. In case
of migratory cells that underwent mitosis, the tracking process was ended 1h before cytokine-
sis. To determine migration speed, it was performed the ratio between the total distance trav-
eled (distance) and the number of slices (time) that cell migrated. To analyze the cell trajectory
and persistence of migration, the X and Y coordinates obtained during the tracking of the
nucleus of the migratory cell in each slice were normalized to start at a virtual X = 0 and Y = 0
position and the variation on the position was plotted in a polar plot graph [25]. For analysis of
adhesion properties, it was used H
inv
/L
E-cad
cells expressing paxillin-GFP from at least 4 inde-
pendent experiments (TIRF microscopy) for each experimental group. Adhesion length and
area was determined by measuring, respectively, the long axis or the area of each adhesion that
assembled during the movie. The percentage of total adhesion area in each newly formed pro-
trusion was measured by the ratio of the sum of the area of all adhesions that assembled in the
protrusion by the total area of the protrusion. The adhesion assembly speed was measured
using the kymographplug-in on ImageJ. For each adhesion, a line (1 pixel-wide) was drawn
in the long axis of the adhesion and the X (distance) and Y (time) coordinates originated by the
kymograph were used to measure the speed of adhesion assembly. All data were calculated
using Microsoft Excel
1
(Microsoft Corporation) and SPSS 21 software (Statistical Package for
the Social Science, IBM).
Statistical Analysis
Student t test or One-way analysis of variance (ANOVA) followed by Tukeys post-test were
employed, using SPSS 21 software and differences were considered significant when p<0.05.
Results
Fibronectin Induces Fast Single Cell Migration of Highly Invasive
OSCCs Cells
Since the extracellular matrix composition can influence the migratory properties of various
cell types, L
inv
/H
E-cad
(Cal27) or H
inv
/L
E-cad
(SCC25) oral squamous cell carcinoma cell lines
[20] were plated on 2D- Matrigel
1
(50μl/cm
2
), laminin (2μg/ml) or fibronectin (2μg/ml)-
coated glass bottomed dishes and imaged for 24h. We tracked the migration velocity of individ-
ual as well as group of cells. Matrigel and laminin were used to mimic the laminin-rich base-
ment membrane that supports cells in an epithelial sheet, whereas fibronectin was used to
challenge the cells with the connective tissue matrix encountered when cells metastasize. On
Matrigel, both cell lines migrated collectively (S1 Movie), while on laminin, both cell types
exhibited collective as well as single cell migration (S2 and S3 Movies). While L
inv
/H
E-cad
cells
showed no changes in migration speed, H
inv
/L
E-cad
cells exhibited a ~40% increase in migration
speed on laminin when compared to Matrigel (Fig 1A). When cells were plated on fibronectin,
both cell types migrated faster than on laminin, and exhibited pronounced changes in
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 5/18
directionality. Both OSCC lines showed a ~40% increase in migration speed; but L
inv
/H
E-cad
cells migrated collectively in circles (S2 Movie), whereas H
inv
/L
E-cad
cells migrated as single
cells with persistent directionality (Fig 1B,S3 Movie).
Fig 1. Fibronectin induces faster migration speed in 2D and 3D substrates. (A) Effects of different 2D
substrates on migration speed (24h) of L
inv
/H
E-cad
(Cal27) or H
inv
/L
E-cad
(SCC25) OSCC cell lines (n = 3);
(B-E) Cell migration trajectory of L
inv
/H
E-cad
(B-C) or H
inv
/L
E-cad
(D-E) cells plated on laminin (B and D) or
fibronectin (C and E); (F) Effects of different 3D substrates on migration speed of L
inv
/H
E-cad
and H
inv
/L
E-cad
cell lines (n = 3). Results are expressed as mean ±SEM. (*)p<0.05 according to One-way analysis of
variance (ANOVA) followed by Tukeys post-test.
doi:10.1371/journal.pone.0151338.g001
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 6/18
To complement the observations in a 2D environment, L
inv
/H
E-cad
or H
inv
/L
E-cad
OSCC
cells were plated in a 3D matrix, containing collagen (1.2mg/ml), collagen + laminin (1.2mg/
ml+10μg/ml) or collagen + fibronectin (1.2mg/ml+10μg/ml), and imaged for 24h. When com-
pared to a 3D collagen only gel, L
inv
/H
E-cad
cells showed a ~50% increase in migration speed
when plated in 3D collagen gel containing laminin or fibronectin (Fig 1F,S4 Movie) with a
slight increase in directional persistence when plated on collagen+laminin. H
inv
/L
E-cad
tumor
cells showed no changes in migration speed when plated in a collagen+laminin 3D environ-
ment, but were able to invade the collagen gel when plated in collagen+fibronectin matrices
(Fig 1F,S5 Movie). Both cells migrated poorly when plated in a 3D matrix containing only
collagen.
Since OSCC biopsies exhibit tumor islands inside the connective tissue, we developed spher-
oids from both cell lines, plated them in a collagen (1.2mg/ml) or a collagen+fibronectin
(1.2mg/ml+2μg/ml) 3D environment, and imaged for 36h. S6 movie shows that small or big
spheroids derived from L
inv
/H
E-cad
OSCCs proliferated, but showed little migratory activity.
However, spheroids of the H
inv
/L
E-cad
OSCC cells showed cells that migrated out of the spher-
oid and invaded the surrounding tissue only when plated in a collagen+fibronectin 3D
environment.
To summarize, these results in 2D and 3D matrices show that L
inv
/H
E-cad
OSCC cells
migrate more directionally when plated using conditions similar to the epithelial and blood
vessel basal lamina; whereas H
inv
/L
E-cad
tumor cells switch from a collective to a faster single
cell migration when transitioning from a laminin to a fibronectin rich connective tissue-like
environment.
OSCCs Extracellular Matrix-Derived Migration Properties Are
Associated with Changes in RhoGTPase Signaling
A differential activation of RhoGTPase signaling is a likely mechanism for the ECM-derived
differences in cell migration observed in the L
inv
/H
E-cad
and H
inv
/L
E-cad
OSCC cell lines. To
address this, we analyzed the Rac1 and RhoA activation levels by pull down and FRET assay of
cells plated on either laminin (2μg/ml) or fibronectin (2μg/ml) coated dishes. Consistent with
the increased migration speed observed for both cell types on fibronectin, fibronectin increased
Rac1 activation levels when compared to cells plated on laminin, which was accompanied by a
FRET signal mainly at the cell borders (Fig 2A); this effect was slightly, but consistently, more
pronounced in the H
inv
/L
E-cad
tumor cells. In contrast, RhoA activity was observed mainly at
the cell body and showed a decrease in the L
inv
/H
E-cad
cells plated on fibronectin, but was unal-
tered in the H
inv
/L
E-cad
cell line (Fig 2B). This decreased RhoA activity may reflect the fact that
L
inv
/H
E-cad
cells migrate collectively on fibronectin, whereas the H
inv
/L
E-cad
cells migrate as sin-
gle cells, where RhoA is necessary for formation of the contractile cell rear underlying persis-
tent directional migration [26]. These data indicate that the effects of ECM constitution on
tumor invasion process involve a differential activation of RhoGTPases that varies according to
the aggressiveness and differentiation level of the tumor cells.
Extracellular-Matrix Composition Interferes with Tumor Cell Adhesion
Properties
Since L
inv
/H
E-cad
OSCCs migrated collectively, whereas H
inv
/L
E-cad
OSCCs migrated as single
cells, we asked whether these effects correlated with the ratio of cell-cell versus cell-ECM adhe-
sions. We hypothesized that L
inv
/H
E-cad
cell line, which exhibit collective cell migration, would
exhibit increased cell-cell adhesion markers, notably cadherin, whereas highly invasive single
cells would most likely favor cell-ECM adhesions. Consistent with this hypothesis, by western
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 7/18
blotting, we observed that both cell lines expressed integrins for fibronectin, but H
inv
/L
E-cad
OSCCs presented an increase in the expression levels of α5 and β1(Fig 3B) when compared to
L
inv
/H
E-cad
OSCCs. Also, H
inv
/L
E-cad
OSCCs show a slightly increased expression of cell-ECM
adhesion related proteins, including the nascent adhesion marker paxillin, the mechanosensing
modulator vinculin and the adhesion signaling marker focal adhesion kinase (FAK). Addition-
ally, H
inv
/L
E-cad
OSCCs gained expression of N-Cadherin, a marker of the epithelial to mesen-
chymal transition (Fig 3A). These data suggest that epithelial-derived tumor cells show a
differential expression of cell-cell and cell-ECM adhesion markers according to the differentia-
tion levels.
In order to analyze the effects of ECM on cellular distribution of adhesion markers, we
observed by immunofluorescence that L
inv
/H
E-cad
OSCCs showed a localization of E-cadherin
Fig 2. RhoGTPase activation varies according to extracellular matrix composition and tumor differentiation levels. FRET analysis and pull down
assay for Rac1 (A) and RhoA (B) of L
inv
/H
E-cad
(Cal27) or H
inv
/L
E-cad
(SCC25) OSCC plated in laminin (2μg/ml) or fibronectin (2μg/ml). Raichu-Rac1-V12 and
Raichu-RhoA-Q63L represents the constitutively activated isoform. Results are expressed as mean ±SD. (*)p<0.05, n = 4.
doi:10.1371/journal.pone.0151338.g002
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 8/18
preferentially at the cell-cell contacts on both fibronectin and laminin (Fig 4A,S1 Fig) while
the ECM adhesion markers, paxillin, vinculin and FAK, all localized primarily to large, elon-
gated adhesions, as is often observed in slow migratory cells. By contrast, H
inv
/L
E-cad
OSCC
cells displayed mainly cytoplasmic E-cadherin with only weak staining at cell-cell contacts in
both ECM environments (Fig 4B). On laminin, paxillin, vinculin and FAK localized to elon-
gated adhesions similar to L
inv
/H
E-cad
OSCCs. On fibronectin, however, these adhesion mark-
ers preferentially redistributed from large adhesions to small adhesions at the cell border,
consistent with the observed increase in Rac activity and the faster migration speed. To confirm
these effects of ECM composition on the adhesion of H
inv
/L
E-cad
OSCCs, we performed live cell
imaging (TIRF microscopy) of cells expressing the nascent adhesion marker paxillin-GFP in
order to analyze adhesions properties during protrusion (Fig 4C and S7 movie). While there
was no difference in adhesion assembly speed when H
inv
/L
E-cad
OSCCs were plated on fibro-
nectin or laminin, fibronectin decreased adhesion length by ~80% (p 0.001, n = 43 adhe-
sions, Student T test), and similarly decreased both individual adhesion area by ~50%
(p 0.05, n = 221 adhesions, Student T test) as well as total adhesion area relative to protru-
sion area by 30% (p 0.01, n = 17 protrusions, Student T test). These data indicate that the
switch from laminin to fibronectin induces smaller adhesions on H
inv
/L
E-cad
OSCCs cells,
which is consistent with the phenotype of highly migratory cells. Thus, the increased persistent
migration of H
inv
/L
E-cad
OSCCs on fibronectin at least in part reflects a preference for cell-
ECM adhesion, particularly nascent signaling adhesions, rather than cell-cell adhesions.
Fig 3. Decreased cell-cell and increased cell-ECM adhesion proteins characterize invasive OSCC. Representative western blotting images of cell-cell
(E-cadherin, N-cadherin), cell-ECM (paxillin, vinculin and FAK) and integrins (α4, α5, αv, β1 and β3) from L
inv
/H
E-cad
(Cal27) or H
inv
/L
E-cad
(SCC25) OSCC
total cell lysates. Densitometry values for each protein were normalized to the loading control (β-Tubulin).
doi:10.1371/journal.pone.0151338.g003
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 9/18
Fig 4. Fibronectin induces smaller adhesion on low E-cadherin expression OSCC cell line. L
inv
/H
E-cad
(A) or H
inv
/L
E-cad
(B) invasive OSCC were plated
on laminin or fibronectin, fixed and stained for E-cadherin and actin, paxillin, vinculin and FAK. White arrows indicate the signal of E-cadherin between cells.
Scale bar = 20μm. Data regarding adhesion properties (C) were obtained using Total Internal Reflectance Fluorescent microscopy analysis of H
inv
/L
E-cad
OSCC cells expressing paxillin-GFP and plated on laminin (light gray) or fibronectin (dark gray). The data shows the assembly speed (μm/sec), adhesion
area (μm
2
), total adhesion area (as % of total protrusion area) and adhesion length (μm). Results are expressed as mean ±SEM. (*) p = 0.05; (**)p<0.01,
according to Student Ttest.
doi:10.1371/journal.pone.0151338.g004
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 10 / 18
Human Oral Squamous Cell Carcinoma Biopsies Show Increased
Levels of Cell-ECM Adhesion Proteins
Since ECM-associated changes on OSCC invasiveness are likely driven by changes in adhesion,
we analyzed the adhesion proteins on biopsies from 10 patients. The sections were stained for
fibronectin, E-cadherin, paxillin, vinculin, and FAK and analyzed using confocal microscopy.
We compared regions from Tumor Adjacent Epithelia (TAE) (Fig 5, columns 1 and 2; S2 and
S3 Figs) with regions at the center of the tumor (Fig 5, columns 3 and 4; S2 and S3 Figs). Fibro-
nectin signal was observed in the connective tissue in both regions (S3 Fig). E-cadherin labeling
in TAE cells was strongest at cell-cell contacts and co-localized with actin; while the center of
the tumor cells showed a weak signal mainly within the cytoplasm, suggesting protein degrada-
tion and/or mislocalization from junctional regions. In TAE regions, paxillin and FAK stained
weakly in puncta through the cytoplasm of the epithelia basal layer and weakly co-localized
with actin; while vinculin was present mainly at the basal membrane. In the center of tumor,
cells that appeared to have detached from the tumor island showed an increase in the staining
of proteins related to cell-ECM adhesion, with paxillin showing increased labeling at the cell
border, close to the ECM, while vinculin and FAK were observed at regions of membrane
extensions of cells at the periphery of the tumor island, with some co-localization with actin.
Thus, human biopsies exhibit changes in the distribution of cell-ECM adhesion proteins, par-
ticularly at the invasive front of the tumor, that correspond with changes observed in cell-ECM
adhesion of H
inv
/L
E-cad
OSCC plated on fibronectin.
Discussion
Clinical failures in cancer therapies are due in part to the plasticity of tumor cells to a changing
microenvironment [14,15]. For example, tumor cells physically and biochemically alter extra-
cellular matrix organization [27], which appears to impact several aspects of the epithelial-to-
mesenchymal transition (EMT) and cell survival [28,29]. Furthermore, the migration of epi-
thelial-derived tumors can vary from collective to single cell migration, reflecting changes in
tumor cell-cell adhesion and the structure of the tissue that the cells are invading [6,30,31]. In
glioblastomas, for example, targeted depletion of fibronectin modifies collective cell migration,
making cancer cells sensitive to ionizing radiation [32].
We have demonstrated that 2D substrates resembling the epithelial basal membrane induce
collective cell migration in OSCC cells independently of the levels of the cell-cell adhesion pro-
tein E-cadherin. In contrast, fibronectin-enriched 2D or 3D environments induced single cell,
mesenchymal-like cell migration but specifically in the H
inv
/L
E-cad
OSCCs. Gaggioli et al
(2007) [17] demonstrated, in a 3D collagen environment, that squamous cell carcinoma cells
showed invasive behavior due to the fibroblast-mediated proteolytic ECM remodeling and
fibronectin deposition. Fibronectin also developmentally regulates migration during embryo-
genesis and determines cell fate [33]. Similarly, fibronectin is overexpressed at the invasive
zone of OSCC biopsies [18,19], where it likely contributes to the abnormal invasive behavior
of poorly differentiated cells [34].
Here we sought to determine how the ECM composition of different tumor regions affects
cell migration and the signaling mechanisms underlying these different migratory properties.
Cell migration is regulated mainly by RhoGTPases, where Rac1 stimulates actin polymerization
and nascent adhesion formation while RhoA controls cell contractility and adhesion maturation
[7,10]. During tumor invasion, the balance of RhoGTPase activation is disrupted. Rac1 activa-
tion results in a loss of cell junctions and polarity and increased cell motility [11,35]. Chen et al
2013 [36] demonstrated that mammary epithelial cells undergo EMT when plated on fibronec-
tin, through a mechanism that involves Rac1b activation, while laminin suppresses EMT.
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 11 / 18
Consistent with this observation, increased Rac1 activity drives mesenchymal-like, single cell
migration of other cancer cells undergoing EMT [6,12]. Yap, et al 2009 [37] demonstrated that
different OSCC cell lines show increased Rac1 activation when plated on fibronectin, indicating
that the microenvironment can influence the tumor invasive behavior through the modulation
Fig 5. Human oral squamous cell carcinoma biopsies show differential distribution of adhesion proteins between center of the tumor cells and
tumor-adjacent epithelia. Regions of biopsies corresponding to the epithelia adjacent to the tumor (A) and from the center of the tumor (B) were submitted
to immunostaining for E-cadherin, paxillin, vinculin or FAK (green) and actin staining (magenta). Inserts demonstrated in actin staining, were digitally
magnified (5x) to show intracellular localization. Representative images from different patients (n = 10), scale bar = 50μmor20μm.
doi:10.1371/journal.pone.0151338.g005
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 12 / 18
of cell migration-related signaling pathways. In this study, when compared to laminin, we
showed that fibronectin induced an increase in Rac1 activation and a rapid single cell migration
phenotype in H
inv
/L
E-cad
OSCCs. Interestingly, while L
inv
/H
E-cad
OSCCs also exhibited
increased Rac1 activation on fibronectin, they had lower RhoA activation. This decreased RhoA
activation may account for the decreased directionality of L
inv
/H
E-cad
OSCCs, which tended to
migrate collectively in circles, since RhoA-mediated myosin activation promotes persistent
directional migration [26]. Thus, differential RhoGTPase activity might contribute to migration
speed and persistence, as well as collective versus single cell migration on different substrates.
In addition to differential RhoGTPase expression, we observed changes in cell-ECM adhe-
sions on different substrates, with fibronectin favoring the formation of small nascent adhe-
sions in H
inv
/L
E-cad
OSCCs [3840]. A possible explanation for the selective effect of
fibronectin in our study is the higher expression of fibronectin-related integrins observed in
H
inv
/L
E-cad
OSCCs when compared to L
inv
/H
E-cad
cells. Integrins are a family of
Fig 6. Effects of the differential composition of extracellular matrix on cell adhesion and signaling of
Oral Squamous Cell Carcinoma. OSCC with high E-cadherin levels (blue cells) shows collective and single
cell migration in the presence of laminin and collective non-directional migration in fibronectin. This switch
correlated to an increase in Rac1 and a decrease on RhoA activation and modulation of the vinculin levels in
adhesion, induced by the fibronectin-enriched environment. For OSCCs with low E-cadherin levels (orange
cells), fibronectin induced smaller adhesions and increased Rac1 signaling, which correspond to a fast single
cell migration phenotype. This model proposes that the ECM composition can trigger the tumorinvasive
behavior according to differentiation levels of OSCC cells.
doi:10.1371/journal.pone.0151338.g006
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 13 / 18
transmembrane proteins that mediate the binding of ECM proteins with intracellular proteins,
resulting in biochemical and mechanical signaling pathways that influence several steps on
tumor progression [4144]. Consistent with our results, Chen et al 2012 [45] showed that lami-
nin induced elongated and fluxing adhesions in CHO.K1 cells, while fibronectin induced
smaller and more dynamic adhesions. Similarly, we demonstrated that both, L
inv
/H
E-cad
or
H
inv
/L
E-cad
OSCCs, when plated on laminin, showed large and elongated adhesions probably
due to the epithelial origin of the tumor. However, specifically in H
inv
/L
E-cad
tumor cells, fibro-
nectin induced smaller cell-ECM adhesions with a fast turnover, which reflected in increased
Rac1 activation [46] and faster single cell migration. Thus the ability to metastasize from a lam-
inin to a fibronectin environment might reflect a switch from cadherin-mediated cell-cell adhe-
sions to signaling integrin-ECM adhesions, which promote directional cell migration.
Several reports associate differential activation of adhesion-related proteins with a worse
patient prognosis [4749]. Also, vulvar squamous cell carcinoma tumors silenced for the fibro-
nectin binding protein, integrin β1, show a more encapsulated and less invasive profile [50]
indicating that cell-ECM interaction is an important player during tumorigenesis. Consistent
with these findings, we demonstrated that human OSCC biopsies show decreased junctional E-
cadherin levels at the center of the tumor when compared to the epithelia adjacent to the
tumor, while the cell-ECM adhesion proteins paxillin, vinculin and FAK showed a differential
distribution in cancer cells at the border of tumor islands close to regions of contact to the
fibronectin enriched ECM. Therefore, our results (Fig 6) show that the extracellular matrix
composition is able to influence the pattern of tumor invasion and metastasis according to the
differentiation level of the tumor cells, probably through modulation of cell signaling and
changes in the balance between cell-cell and cell-ECM adhesion. These data suggest that the
invasive behavior of OSCC not only relies on intrinsic factors (i.e. mutations and abnormal
expression of proteins) but also on extrinsic factors (such as the ECM composition), which
could help to understand the failure of some tumor therapies and contribute to development of
new anti-tumorigenic approaches.
Supporting Information
S1 Fig. Differential distribution of E-cadherin on two different cell lines. L
inv
/H
E-cad
(A) or
H
inv
/L
E-cad
(B) OSCC were plated on laminin or fibronectin, fixed and stained for E-cadherin
and actin. Scale bar = 20μm.
(TIF)
S2 Fig. Distribution of cell-cell and cell-ECM adhesion molecules in OSCC human biopsies.
Original images showed in Fig 5 in the manuscript. Biopsies corresponding to the epithelia
adjacent to the tumor (A) and from the center of the tumor region (B) were submitted to actin
staining (first column) and immunostaining (second column) for E-cadherin, paxillin, vinculin
or FAK. Representative images of n = 10, digital zoom 5x, scale bar = 20μm.
(TIF)
S3 Fig. Distribution of fibronectin in OSCC human biopsies. Regions of biopsies corre-
sponding to the epithelia adjacent to the tumor (A) and from the center of the tumor (B) were
submitted to immunostaining for fibronectin (green) and actin staining (magenta). Represen-
tative images from the same patient (n = 10), scale bar = 50μm.
(TIF)
S1 Movie. Migratory properties of low and highly invasive Oral Squamous Cell Carcinoma
cell lines plated on Matrigel. Time-lapse images (right column) and cell tracking (left column)
of Oral Squamous Cell Carcinoma with H
inv
/L
E-cad
(upper line) or L
inv
/H
E-cad
(lower line)
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 14 / 18
plated for 1h on Matrigel (50μl/cm
2
) and imaged for 24h with a 10min time interval.
(AVI)
S2 Movie. Migratory properties of low invasive Oral Squamous Cell Carcinoma plated in
laminin or fibronectin. Time-lapse images (left column) and cell tracking (right column) of
Oral Squamous Cell Carcinoma with L
inv
/H
E-cad
plated for 1h on laminin (2μg/ml, upper line)
or fibronectin (2μg/ml, lower line) and imaged for 24h with a 10min time interval. This movie
corresponds to Fig 1B and 1C.
(AVI)
S3 Movie. Migratory properties of highly invasive Oral Squamous Cell Carcinoma plated
in laminin or fibronectin. Time-lapse images (left column) and cell tracking (right column) of
H
inv
/L
E-cad
plated for 1h on laminin (2μg/ml, upper line) or fibronectin (2μg/ml, lower line)
and imaged for 24h with a 10min time interval. This movie corresponds to Fig 1D and 1E.
(AVI)
S4 Movie. Migratory properties of low invasive Oral Squamous Cell Carcinoma plated in a
3D matrix. Time-lapse images (left column) and cell tracking (right column) of L
inv
/H
E-cad
OSCC were plated for 1h in a 3D matrix of collagen (1.2mg/ml, upper line), collagen+laminin
(1.2mg/ml+10μg/ml, center line) or collagen+fibronectin (1.2mg/ml+10μg/ml, lower line) and
imaged for 24h with a 10min time interval. This movie corresponds to Fig 1F.
(AVI)
S5 Movie. Migratory properties of highly invasive Oral Squamous Cell Carcinoma plated
in a 3D matrix. Time-lapse images (left column) and cell tracking (right column) of H
inv
/L
E-
cad
OSCC were plated for 1h in a 3D matrix of collagen (1.2mg/ml, upper line), collagen+-
laminin (1.2mg/ml+10μg/ml, center line) or collagen+fibronectin (1.2mg/ml+10μg/ml, lower
line) and imaged for 24h with a 10min time interval. This movie corresponds to Fig 1F.
(AVI)
S6 Movie. Migratory properties of Oral Squamous Cell Carcinoma cell lines-derived spher-
oids in a 3D extracellular matrix. Time-lapse images of spheroids obtained from Oral Squa-
mous Cell Carcinoma with L
inv
/H
E-cad
(left column) plated in a 3D matrix containing
collagen+fibronectin (1.2mg/ml+10μg/ml) or H
inv
/L
E-cad
(center and right column) plated in
a 3D extracellular matrix composed by collagen only (1.2mg/ml, center column) or collagen-
+fibronectin (1.2mg/ml+10μg/ml, right column) and imaged for 36h with a 10min time
interval.
(AVI)
S7 Movie. Extracellular matrix composition affects adhesion dynamics of highly invasive
Oral Squamous Cell Carcinoma. Adhesion dynamics of Oral Squamous Cell Carcinoma with
low E-cadherin levels transfected with paxillin-GFP, plated for 20min on laminin (2μg/ml, left
column) or fibronectin (2μg/ml, right column) and imaged using Total Internal Reflectance
Fluorescent (TIRF) microscopy for 10min with a 3s time interval. The black box represents a
digital zoom of the original movie showing the details of cell adhesion dynamics in each condi-
tion.
(AVI)
Acknowledgments
The authors thank Laura de Campos Hildebrand and Ana Luisa Homem de Carvalho for help
with the human sample biopsy collection and Karen Newell-Litwa for revision of the article.
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 15 / 18
Author Contributions
Conceived and designed the experiments: MSF AH ML. Performed the experiments: GR LB IL
ML. Analyzed the data: GR LB IL ML. Contributed reagents/materials/analysis tools: MSF AH
ML. Wrote the paper: GR LB IL MSF AH ML.
References
1. Barnes L, Everson J.W., Reichart P., Sidransky D. Pathology and genetics of head and neck tumours.
World Health Organization Classification of Tumours. 2005:283327.
2. Scully C, Bagan J. Oral squamous cell carcinoma overview. Oral Oncol. 2009; 45(45):3018. Epub
2009/03/03. doi: 10.1016/j.oraloncology.2009.01.004 PMID: 19249237.
3. Hunter KD, P E, Harrison PR. Profiling early head and neck cancer. Nature Reviews. 2005; 5:12735.
PMID: 15685196
4. Leemans CR, Braakhuis BJ, Brakenhoff RH. The molecular biology of head and neck cancer. Nat Rev
Cancer. 2011; 11(1):922. Epub 2010/12/17. doi: 10.1038/nrc2982 PMID: 21160525.
5. Sethi N, Kang Y. Unravelling the complexity of metastasismolecular understanding and targeted ther-
apies. Nat Rev Cancer. 2011; 11(10):73548. Epub 2011/09/24. doi: 10.1038/nrc3125 PMID:
21941285.
6. Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;
147(5):9921009. Epub 2011/11/29. doi: 10.1016/j.cell.2011.11.016 PMID: 22118458.
7. Ridley AJ, Schwartz MA, Burridge K, Firtel RA, Ginsberg MH, Borisy G, et al. Cell migration: integrating
signals from front to back. Science. 2003; 302(5651):17049. Epub 2003/12/06. doi: 10.1126/science.
1092053 PMID: 14657486.
8. Parsons JT, Horwitz AR, Schwartz MA. Cell adhesion: integrating cytoskeletal dynamics and cellular
tension. Nat Rev Mol Cell Biol. 2010; 11(9):63343. Epub 2010/08/24. doi: 10.1038/nrm2957 PMID:
20729930; PubMed Central PMCID: PMCPmc2992881.
9. Peglion F, Llense F, Etienne-Manneville S. Adherens junction treadmilling during collective migration.
Nat Cell Biol. 2014; 16(7):63951. Epub 2014/06/16. doi: 10.1038/ncb2985 PMID: 24929360.
10. Ridley AJ. Life at the leading edge. Cell. 2011; 145(7):101222. Epub 2011/06/28. doi: 10.1016/j.cell.
2011.06.010 PMID: 21703446.
11. Sahai E, Marshall CJ. RHO-GTPases and cancer. Nat Rev Cancer. 2002; 2(2):13342. Epub 2003/03/
15. doi: 10.1038/nrc725 PMID: 12635176.
12. Sanz-Moreno V, Gadea G, Ahn J, Paterson H, Marra P, Pinner S, et al. Rac activation and inactivation
control plasticity of tumor cell movement. Cell. 2008; 135(3):51023. Epub 2008/11/06. doi: 10.1016/j.
cell.2008.09.043 PMID: 18984162.
13. Takeichi M. Dynamic contacts: rearranging adherens junctions to drive epithelial remodelling. Nat Rev
Mol Cell Biol. 2014; 15(6):397410. Epub 2014/05/16. doi: 10.1038/nrm3802 PMID: 24824068.
14. Bissell MJ, Hines WC. Why don't we get more cancer? A proposed role of the microenvironment in
restraining cancer progression. Nat Med. 2011; 17(3):3209. Epub 2011/03/09. doi: 10.1038/nm.2328
PMID: 21383745; PubMed Central PMCID: PMCPmc3569482.
15. Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvi-
ronment. Cancer Cell. 2012; 21(3):30922. Epub 2012/03/24. doi: 10.1016/j.ccr.2012.02.022 PMID:
22439926.
16. Nelson CM, Bissell MJ. Of extracellular matrix, scaffolds, and signaling: tissue architecture regulates
development, homeostasis, and cancer. Annu Rev Cell Dev Biol. 2006; 22:287309. Epub 2006/07/11.
doi: 10.1146/annurev.cellbio.22.010305.104315 PMID: 16824016; PubMed Central PMCID:
PMCPmc2933192.
17. Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF, Harrington K, et al. Fibroblast-led
collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells.
Nat Cell Biol. 2007; 9(12):1392400. Epub 2007/11/27. doi: 10.1038/ncb1658 PMID: 18037882.
18. Harada T, Shinohara M, Nakamura S, Oka M. An immunohistochemical study of the extracellular matrix
in oral squamous cell carcinoma and its association with invasive and metastatic potential. Virchows
Arch. 1994; 424(3):25766. Epub 1994/01/01. PMID: 7514477.
19. Kosmehl H, Berndt A, Strassburger S, Borsi L, Rousselle P, Mandel U, et al. Distribution of laminin and
fibronectin isoforms in oral mucosa and oral squamous cell carcinoma. Br J Cancer. 1999; 81(6):1071
9. Epub 1999/11/27. doi: 10.1038/sj.bjc.6690809 PMID: 10576667; PubMed Central PMCID:
PMCPmc2362955.
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 16 / 18
20. Colley HE, Hearnden V, Jones AV, Weinreb PH, Violette SM, Macneil S, et al. Development of tissue-
engineered models of oral dysplasia and early invasive oral squamous cell carcinoma. Br J Cancer.
2011; 105(10):158292. Epub 2011/10/13. doi: 10.1038/bjc.2011.403 PMID: 21989184; PubMed Cen-
tral PMCID: PMCPmc3242522.
21. Laukaitis CM, Webb DJ, Donais K, Horwitz AF. Differential dynamics of alpha 5 integrin, paxillin, and
alpha-actinin during formation and disassembly of adhesions in migrating cells. J Cell Biol. 2001; 153
(7):142740. Epub 2001/06/27. PMID: 11425873; PubMed Central PMCID: PMCPmc2150721.
22. Ren XD, Kiosses WB, Schwartz MA. Regulation of the small GTP-binding protein Rho by cell adhesion
and the cytoskeleton. Embo j. 1999; 18(3):57885. Epub 1999/02/02. doi: 10.1093/emboj/18.3.578
PMID: 9927417; PubMed Central PMCID: PMCPmc1171150.
23. Nakamura T, Kurokawa K, Kiyokawa E, Matsuda M. Analysis of the spatiotemporal activation of rho
GTPases using Raichu probes. Methods Enzymol. 2006; 406:31532. Epub 2006/02/14. doi: 10.1016/
s0076-6879(06)06023-x PMID: 16472667.
24. Hodgson L, Shen F, Hahn K. Biosensors for characterizing the dynamics of rho family GTPases in living
cells. Curr Protoc Cell Biol. 2010; Chapter 14:Unit 14.1.126. Epub 2010/03/18. doi: 10.1002/
0471143030.cb1411s46 PMID: 20235099; PubMed Central PMCID: PMCPmc2998069.
25. Lamers ML, Almeida ME, Vicente-Manzanares M, Horwitz AF, Santos MF. High glucose-mediated oxi-
dative stress impairs cell migration. PLoS One. 2011; 6(8):e22865. Epub 2011/08/10. doi: 10.1371/
journal.pone.0022865 PMID: 21826213; PubMed Central PMCID: PMCPmc3149607.
26. Vicente-Manzanares M, Koach MA, Whitmore L, Lamers ML, Horwitz AF. Segregation and activation of
myosin IIB creates a rear in migrating cells. J Cell Biol. 2008; 183(3):54354. Epub 2008/10/29. doi: 10.
1083/jcb.200806030 PMID: 18955554; PubMed Central PMCID: PMCPmc2575793.
27. Friedl P, Wolf K. Plasticity of cell migration: a multiscale tuning model. J Cell Biol. 2010; 188(1):119.
Epub 2009/12/03. doi: 10.1083/jcb.200909003 PMID: 19951899; PubMed Central PMCID:
PMCPmc2812848.
28. Buchheit CL, Weigel KJ, Schafer ZT. Cancer cell survival during detachment from the ECM: multiple
barriers to tumour progression. Nat Rev Cancer. 2014; 14(9):63241. Epub 2014/08/08. doi: 10.1038/
nrc3789 PMID: 25098270.
29. Thiery JP, Chua K, Sim WJ, Huang R. [Epithelial mesenchymal transition during development in fibro-
sis and in the progression of carcinoma]. Bull Cancer. 2010; 97(11):128595. Epub 2010/11/19. doi:
10.1684/bdc.2010.1206 PMID: 21084241.
30. Tozluoglu M, Tournier AL, Jenkins RP, Hooper S, Bates PA, Sahai E. Matrix geometry determines opti-
mal cancer cell migration strategy and modulates response to interventions. Nat Cell Biol. 2013; 15
(7):75162. Epub 2013/06/25. doi: 10.1038/ncb2775 PMID: 23792690.
31. Wolf K, Te Lindert M, Krause M, Alexander S, Te Riet J, Willis AL, et al. Physical limits of cell migration:
control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J Cell Biol.
2013; 201(7):106984. Epub 2013/06/27. doi: 10.1083/jcb.201210152 PMID: 23798731; PubMed Cen-
tral PMCID: PMCPmc3691458.
32. Serres E, Debarbieux F, Stanchi F, Maggiorella L, Grall D, Turchi L, et al. Fibronectin expression in glio-
blastomas promotes cell cohesion, collective invasion of basement membrane in vitro and orthotopic
tumor growth in mice. Oncogene. 2014; 33(26):345162. Epub 2013/08/06. doi: 10.1038/onc.2013.305
PMID: 23912459.
33. Schwarzbauer JE, DeSimone DW. Fibronectins, their fibrillogenesis, and in vivo functions. Cold Spring
Harb Perspect Biol. 2011; 3(7). Epub 2011/05/18. doi: 10.1101/cshperspect.a005041 PMID:
21576254; PubMed Central PMCID: PMCPmc3119908.
34. Kamarajan P, Garcia-Pardo A, D'Silva NJ, Kapila YL. The CS1 segment of fibronectin is involved in
human OSCC pathogenesis by mediating OSCC cell spreading, migration, and invasion. BMC Cancer.
2010; 10:330. Epub 2010/06/29. doi: 10.1186/1471-2407-10-330 PMID: 20579373; PubMed Central
PMCID: PMCPmc3146068.
35. Hirata E, Yukinaga H, Kamioka Y, Arakawa Y, Miyamoto S, Okada T, et al. In vivo fluorescence reso-
nance energy transfer imaging reveals differential activation of Rho-family GTPases in glioblastoma
cell invasion. J Cell Sci. 2012; 125(Pt 4):85868. Epub 2012/03/09. doi: 10.1242/jcs.089995 PMID:
22399802.
36. Chen QK, Lee K, Radisky DC, Nelson CM. Extracellular matrix proteins regulate epithelial-mesenchy-
mal transition in mammary epithelial cells. Differentiation. 2013; 86(3):12632. Epub 2013/05/11. doi:
10.1016/j.diff.2013.03.003 PMID: 23660532; PubMed Central PMCID: PMCPmc3762919.
37. Yap LF, Jenei V, Robinson CM, Moutasim K, Benn TM, Threadgold SP, et al. Upregulation of Eps8 in
oral squamous cell carcinoma promotes cell migration and invasion through integrin-dependent Rac1
activation. Oncogene. 2009; 28(27):252434. Epub 2009/05/19. doi: 10.1038/onc.2009.105 PMID:
19448673.
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 17 / 18
38. von Schlippe M, Marshall JF, Perry P, Stone M, Zhu AJ, Hart IR. Functional interaction between E-cad-
herin and alphav-containing integrins in carcinoma cells. J Cell Sci. 2000; 113 (Pt 3):42537. Epub
2000/01/20. PMID: 10639330.
39. Yano H, Mazaki Y, Kurokawa K, Hanks SK, Matsuda M, Sabe H. Roles played by a subset of integrin
signaling molecules in cadherin-based cell-cell adhesion. J Cell Biol. 2004; 166(2):28395. Epub 2004/
07/21. doi: 10.1083/jcb.200312013 PMID: 15263022; PubMed Central PMCID: PMCPmc2172299.
40. Weber GF, Bjerke MA, DeSimone DW. A mechanoresponsive cadherin-keratin complex directs polar-
ized protrusive behavior and collective cell migration. Dev Cell. 2012; 22(1):10415. Epub 2011/12/16.
doi: 10.1016/j.devcel.2011.10.013 PMID: 22169071; PubMed Central PMCID: PMCPmc3264825.
41. Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 2004; 5
(10):81626. Epub 2004/10/02. doi: 10.1038/nrm1490 PMID: 15459662.
42. Giancotti FG. Mechanisms governing metastatic dormancy and reactivation. Cell. 2013; 155(4):750
64. Epub 2013/11/12. doi: 10.1016/j.cell.2013.10.029 PMID: 24209616; PubMed Central PMCID:
PMCPMC4354734.
43. Naba A, Clauser KR, Whittaker CA, Carr SA, Tanabe KK, Hynes RO. Extracellular matrix signatures of
human primary metastatic colon cancers and their metastases to liver. BMC Cancer. 2014; 14:518.
Epub 2014/07/20. doi: 10.1186/1471-2407-14-518 PMID: 25037231; PubMed Central PMCID:
PMCPMC4223627.
44. Yao X, Labelle M, Lamb CR, Dugan JM, Williamson CA, Spencer DR, et al. Determination of 35 cell sur-
face antigen levels in malignant pleural effusions identifies CD24 as a marker of disseminated tumor
cells. Int J Cancer. 2013; 133(12):292533. Epub 2013/06/19. doi: 10.1002/ijc.28312 PMID: 23775727;
PubMed Central PMCID: PMCPMC4107365.
45. Chen L, Vicente-Manzanares M, Potvin-Trottier L, Wiseman PW, Horwitz AR. The integrin-ligand inter-
action regulates adhesion and migration through a molecular clutch. PLoS One. 2012; 7(7):e40202.
Epub 2012/07/14. doi: 10.1371/journal.pone.0040202 PMID: 22792239; PubMed Central PMCID:
PMCPmc3391238.
46. Filipenko NR, Attwell S, Roskelley C, Dedhar S. Integrin-linked kinase activity regulates Rac- and
Cdc42-mediated actin cytoskeleton reorganization via alpha-PIX. Oncogene. 2005; 24(38):583749.
Epub 2005/05/18. doi: 10.1038/sj.onc.1208737 PMID: 15897874.
47. Nagata M, Fujita H, Ida H, Hoshina H, Inoue T, Seki Y, et al. Identification of potential biomarkers of
lymph node metastasis in oral squamous cell carcinoma by cDNA microarray analysis. Int J Cancer.
2003; 106(5):6839. Epub 2003/07/17. doi: 10.1002/ijc.11283 PMID: 12866027.
48. Huang WC, Chan SH, Jang TH, Chang JW, Ko YC, Yen TC, et al. miRNA-491-5p and GIT1 serve as
modulators and biomarkers for oral squamous cell carcinoma invasion and metastasis. Cancer Res.
2014; 74(3):75164. Epub 2013/12/18. doi: 10.1158/0008-5472.can-13-1297 PMID: 24335959.
49. Sulzmaier FJ, Jean C, Schlaepfer DD. FAK in cancer: mechanistic findings and clinical applications.
Nat Rev Cancer. 2014; 14(9):598610. Epub 2014/08/08. doi: 10.1038/nrc3792 PMID: 25098269.
50. Brockbank EC, Bridges J, Marshall CJ, Sahai E. Integrin beta1 is required for the invasive behaviour
but not proliferation of squamous cell carcinoma cells in vivo. Br J Cancer. 2005; 92(1):10212. Epub
2004/12/15. doi: 10.1038/sj.bjc.6602255 PMID: 15597106; PubMed Central PMCID:
PMCPmc2361733.
Extracellular Matrix Composition Affects Tumor Invasion
PLOS ONE | DOI:10.1371/journal.pone.0151338 March 15, 2016 18 / 18
... While biochemical signals such as chemokines have long been associated with stimulating and facilitating migration, more recent studies have implicated both passive and active biophysical signals as playing equally integral roles (Gaylo et al., 2016;Fowell and Kim, 2021;Tello-Lafoz et al., 2021;Du et al., 2023). Indeed, passive features of the extracellular environment, including its stiffness (Lo et al., 2000;Bangasser et al., 2017;Janmey et al., 2020;Isomursu et al., 2022), composition (Ramos et al., 2016;Hartman et al., 2017), and architecture (Pieuchot et al., 2018;Tien et al., 2020;Nicolas-Boluda et al., 2021) can all influence cell migration (Pathak and Kumar, 2012;Gaylo et al., 2016). Active forces generated internally by the cell and surrounding cells in the tissue can also have an impact (Boyd et al., 1988;Reiss et al., 1998;Zhang et al., 2006;Chen and Zhu, 2013), such as by modulating bond strength and lifetime. ...
Article
Full-text available
Immune cells are highly dynamic and able to migrate through environments with diverse biochemical and mechanical compositions. Their migration has classically been defined as amoeboid under the assumption that it is integrin independent. Here, we show that activated primary Th1 T cells require both confinement and extracellular matrix proteins to migrate efficiently. This migration is mediated through small and dynamic focal adhesions that are composed of the same proteins associated with canonical mesenchymal cell focal adhesions, such as integrins, talin, and vinculin. These focal adhesions, furthermore, localize to sites of contractile traction stresses, enabling T cells to pull themselves through confined spaces. Finally, we show that Th1 T cells preferentially follow tracks of other T cells, suggesting that these adhesions modify the extracellular matrix to provide additional environmental guidance cues. These results demonstrate not only that the boundaries between amoeboid and mesenchymal migration modes are ambiguous, but that integrin-mediated focal adhesions play a key role in T cell motility.
... CAFs are the major source of tumor ECM and a shift from a laminin-to a collagen-and fibronectin-rich environment is known to promote cancer cell invasiveness [25][26][27]. By analyzing the content of these ECM proteins by immunofluorescence, we observed that both collagen I and fibronectin were reduced by the overexpression of EMID2 (Fig. 2I-L), while laminin was up-regulated to the level of normal muscles ( Supplementary Fig. S5A, B). ...
Article
Full-text available
Background New drugs to tackle the next pathway or mutation fueling cancer are constantly proposed, but 97% of them are doomed to fail in clinical trials, largely because they are identified by cellular or in silico screens that cannot predict their in vivo effect. Methods We screened an Adeno-Associated Vector secretome library (> 1000 clones) directly in vivo in a mouse model of cancer and validated the therapeutic effect of the first hit, EMID2, in both orthotopic and genetic models of lung and pancreatic cancer. Results EMID2 overexpression inhibited both tumor growth and metastatic dissemination, consistent with prolonged survival of patients with high levels of EMID2 expression in the most aggressive human cancers. Mechanistically, EMID2 inhibited TGFβ maturation and activation of cancer-associated fibroblasts, resulting in more elastic ECM and reduced levels of YAP in the nuclei of cancer cells. Conclusion This is the first in vivo screening, precisely designed to identify proteins able to interfere with cancer cell invasiveness. EMID2 was selected as the most potent protein, in line with the emerging relevance of the tumor extracellular matrix in controlling cancer cell invasiveness and dissemination, which kills most of cancer patients.
... Mechanically, CRP has the potential to solve the inflammatory environment by directly binding fibronectin to modulate fibronectin-mediated monocyte adhesion [37]. Fibronectin can regulate cell adhesion and signaling to promote oral cancer cell migration [38]. Similarly, it has the ability to stimulate white blood cells to produce IL-8 [39]. ...
Article
Full-text available
Background Observational epidemiological studies revealed that multiple serum biomarkers can be associated with the risk of oral and oropharyngeal cancer (OC/OPC). However, the causal relationship between them remains largely unknown. This study aimed to investigate the causal relationship between potential serum biomarkers and (OC/OPC). Methods A two-sample Mendelian randomization (MR) approach was performed to assess the causal association of 10 serum biomarkers with the risk of OC / OPC. Summary data on OC/OPC were obtained from a GWAS meta-analysis that included 2497 cases and 2928 controls. The TwoSampleMR package in R was used to perform MR analyzes. Inverse-variance weighted (IVW), Weighted median and MR-Egger methods were used to assess causal effects. Results Suggestive associations with increased risk of C-reactive protein (CRP) ( OR 1.52, 95% CI 1.14 to 2.02), using the IVW method. MR-Egger regression suggested that directional pleiotropy was unlikely to bias the result ( P = 0.19). The findings were robust to sensitivity analyzes. The risk of OC/OPC was not associated with serum 25-hydroxyvitamin D, HDL cholesterol, LDL cholesterol, total cholesterol, triglycerides, adiponectin, leptin, HbA1C and Insulin-like growth factor 1 (IGF 1). Conclusions This study supports that CRP was causally associated with an increased risk of oral and oropharyngeal cancer.
... CAFs are the major source of tumor ECM and a shift from a laminin-to a collagen-and bronectin-rich environment is known to promote cancer cell invasiveness (25)(26)(27). By analyzing the content of these ECM proteins by immuno uorescence, we observed that both collagen I and bronectin were reduced by the overexpression of EMID2 (Fig. 2I-L), while laminin was up-regulated to a level like normal muscles ( Supplementary Fig. S5A, B). ...
Preprint
Full-text available
Background. New drugs to tackle the next pathway or mutation fueling cancer are constantly proposed, but 97% of them are doomed to fail in clinical trials, largely because they are identified by cellular or in silico screens that cannot predict their in vivo effect. Methods. We screened an Adeno-Associated Vector secretome library (> 1000 clones) directly in vivo in a mouse model of cancer and validated the therapeutic effect of the first hit, EMID2, in both orthotopic and genetic models of lung and pancreatic cancer. Results. EMID2 overexpression inhibited both tumor growth and metastatic dissemination, consistent with prolonged survival of patients with high levels of EMID2 expression in the most aggressive human cancers. Mechanistically, EMID2 inhibited TGFβ maturation and activation of cancer-associated fibroblasts, resulting in more elastic ECM and reduced levels of YAP in the nuclei of cancer cells. Conclusions. This is the first in vivo screening, precisely designed to identify proteins able to interfere with cancer cell invasiveness. EMID2 was selected as the most potent protein, in line with the emerging relevance of the tumor extracellular matrix in controlling cancer cell invasiveness and dissemination, which kills most of cancer patients.
... 50 52 Therefore, the extracellular matrix with EDB-fibronectin may function as a track for migrating cancer cells or endothelial cells. [53][54][55] This model is consistent with the findings that mice lacking EDA and EDB-fibronectin demonstrated collapsed microtubes consisting of endothelial cells 19 or with expression levels proportional to the degrees of malignancy. 56-59 EDB-fibronectin is typically absent from the resting endothelium of healthy tissues; thus, targeting EDB-fibronectin for cancer treatment may be devoid of on-target off-tumor toxicities. ...
Article
Full-text available
Background: The suppression of chimeric antigen receptor (CAR) T cells by the tumor microenvironment (TME) is a crucial obstacle in the T-cell-based treatment of solid tumors. Extra domain B (EDB)-fibronectin is an oncofetal antigen expressed on the endothelium layer of the neovasculature and cancer cells. Though recognized as a T cell therapy target, engineered CAR T cells thus far have failed to demonstrate satisfactory in vivo efficacy. In this study, we report that targeting EDB-fibronectin by redirected TCR-CAR T cells (rTCR-CAR) bypasses the suppressive TME for solid tumor treatment and sufficiently suppressed tumor growth.We generated EDB-targeting CAR by fusing single-chain variable fragment to CD3ε, resulting in rTCR-CAR. Human primary T cells and Jurkat cells were used to study the EDB-targeting T cells. Differences to the traditional second-generation CAR T cell in signaling, immune synapse formation, and T cell exhaustion were characterized. Cytotoxicity of the rTCR-CAR T cells was tested in vitro, and therapeutic efficacies were demonstrated using xenograft models. Methods: RESULTS: In the xenograft models, the rTCR-CAR T cells demonstrated in vivo efficacies superior to that based on traditional CAR design. A significant reduction in tumor vessel density was observed alongside tumor growth inhibition, extending even to tumor models established with EDB-negative cancer cells. The rTCR-CAR bound to immobilized EDB, and the binding led to immune synapse structures superior to that formed by second-generation CARs. By a mechanism similar to that for the conventional TCR complex, EDB-fibronectin activated the rTCR-CAR, resulting in rTCR-CAR T cells with low basal activation levels and increased in vivo expansion. Conclusion: Our study has demonstrated the potential of rTCR-CAR T cells targeting the EDB-fibronectin as an anticancer therapeutic. Engineered to possess antiangiogenic and cytotoxic activities, the rTCR-CAR T cells showed therapeutic efficacies not impacted by the suppressive TMEs. These combined characteristics of a single therapeutic agent point to its potential to achieve sustained control of solid tumors.
... Additionally, the authors have reported that collagen substrates may be responsible for enhancing hMSCs' osteogenic ability, which may benefit bone-related clinical applications (Somaiah et al. 2015). Similarly, fibronectin has been demonstrated to play a major role in cell adhesion and migration (Ruoslahti 1984;Ramos et al. 2016;Hsiao et al. 2017). Previous studies have reported that hMSC migration is also regulated by fibronectin (Veevers-Lowe et al. 2011). ...
Article
Full-text available
Bioactive materials interact with cells and modulate their characteristics which enable the generation of cell-based products with desired specifications. However, their evaluation and impact are often overlooked when establishing a cell therapy manufacturing process. In this study, we investigated the role of different surfaces for tissue culture including, untreated polystyrene surface, uncoated Cyclic Olefin Polymer (COP) and COP coated with collagen and recombinant fibronectin. It was observed that human mesenchymal stromal cells (hMSCs) expanded on COP-coated plates with different bioactive materials resulted in improved cell growth kinetics compared to traditional polystyrene plates and non-coated COP plates. The doubling time obtained was 2.78 and 3.02 days for hMSC seeded in COP plates coated with collagen type I and recombinant fibronectin respectively, and 4.64 days for cells plated in standard polystyrene treated plates. Metabolite analysis reinforced the findings of the growth kinetic studies, specifically that cells cultured on COP plates coated with collagen I and fibronectin exhibited improved growth as evidenced by a higher lactate production rate (9.38 × 10⁵ and 9.67 × 10⁵ pmol/cell/day, respectively) compared to cells from the polystyrene group (5.86 × 10⁵ pmol/cell/day). This study demonstrated that COP is an effective alternative to polystyrene-treated plates when coated with bioactive materials such as collagen and fibronectin, however COP-treated plates without additional coatings were found not to be sufficient to support cell growth. These findings demonstrate the key role biomaterials play in the cell manufacturing process and the importance of optimising this selection.
... High fibronectin expression has been associated with inferior survival for patients with other HPV-associated cancers such as cervical cancer [25] and head/neck cancer [26]. In vitro, models of HPV-associated cancers have linked fibronectin overexpression to increased activation of the focal adhesion kinase signaling pathway, which promotes cancer cell migration [27] and the polarization of anti-inflammatory M2 macrophages [28] that may promote tumor progression [29]. Consistent with these findings for other HPV-associated malignancies, our data here provide further support for fibronectin as an unfavorable prognostic biomarker for anal cancer. ...
Article
Full-text available
Simple Summary While anti-PD1 antibodies have demonstrated efficacy in some patients with metastatic anal cancer, these agents have no proven benefit for those with localized disease treated with chemoradiation. Difficulty procuring fresh tumor tissue required for RNA and protein expression analysis has limited extensive molecular profiling for this rare cancer. Our team utilized a novel digital spatial profiling technology on pretreatment anal cancer specimens to identify biomarkers associated with recurrence after chemoradiation. We observed that recurrent tumors had higher baseline expression of immune checkpoint biomarkers, higher MAPK signaling activation and higher PI3K/Akt signaling activation. These findings provide a rationale that supports future clinical trials with immunotherapy that seek to improve survival beyond chemoradiation for patients with localized squamous cell cancer of the anus. Abstract The identification of transcriptomic and protein biomarkers prognosticating recurrence risk after chemoradiation of localized squamous cell carcinoma of the anus (SCCA) has been limited by a lack of available fresh tissue at initial presentation. We analyzed archival FFPE SCCA specimens from pretreatment biopsies prior to chemoradiation for protein and RNA biomarkers from patients with localized SCCA who recurred (N = 23) and who did not recur (N = 25). Tumor cells and the tumor microenvironment (TME) were analyzed separately to identify biomarkers with significantly different expression between the recurrent and non-recurrent groups. Recurrent patients had higher mean protein expression of FoxP3, MAPK-activation markers (BRAF, p38-MAPK) and PI3K/Akt activation (phospho-Akt) within the tumor regions. The TME was characterized by the higher protein expression of immune checkpoint biomarkers such as PD-1, OX40L and LAG3. For patients with recurrent SCCA, the higher mean protein expression of fibronectin was observed in the tumor and TME compartments. No significant differences in RNA expression were observed. The higher baseline expression of immune checkpoint biomarkers, together with markers of MAPK and PI3K/Akt signaling, are associated with recurrence following chemoradiation for patients with localized SCCA. These data provide a rationale towards the application of immune-based therapeutic strategies to improve curative-intent outcomes beyond conventional therapies for patients with SCCA.
... Among matching integrin ligands, there was decreased expression of the ECM component and critical cell adhesion and morphology regulator FN1 in fibroblasts and lymphatic endothelial cells (Figure 4H). [55][56][57] Overall staining intensity of FN1 was not decreased in these cell types after androgen therapy, but fewer FN1 + fibroblasts were identified near the epithelium ( Figures 3F, S8A, and S8B). Furthermore, DNA binding motifs for the androgenresponsive gene estrogen-related receptor gamma (ESRRG) are over-represented in chromatin peaks proximal to focal adhesion genes in these cells ( Figures 4G and 4I). ...
Article
Full-text available
Estrogen and progesterone have been extensively studied in the mammary gland, but the molecular effects of androgen remain largely unexplored. Transgender men are recorded as female at birth but identify as male and may undergo gender-affirming androgen therapy to align their physical characteristics and gender identity. Here we perform single-cell-resolution transcriptome, chromatin, and spatial profiling of breast tissues from transgender men following androgen therapy. We find canonical androgen receptor gene targets are upregulated in cells expressing the androgen receptor and that paracrine signaling likely drives sex-relevant androgenic effects in other cell types. We also observe involution of the epithelium and a spatial reconfiguration of immune, fibroblast, and vascular cells, and identify a gene regulatory network associated with androgen-induced fat loss. This work elucidates the molecular consequences of androgen activity in the human breast at single-cell resolution.
... Since the ECM regulates cell response due to its structural and reservoir function, it is assumed that its biochemical properties should be considered when examining tumor behavior, as well as therapeutic interventions (Egeblad et al., 2010;Oskarsson, 2013;Terajima et al., 2021). Furthermore, cell/ECM interaction confers cell resistance, both in tumor and normal cells (Cordes and Van Beuningen, 2003;Gkretsi and Stylianopoulos, 2018;Horton et al., 2016;Hynes, 2002;Ramos et al., 2016). ...
Article
Extracellular matrix (ECM) proteins in the mammary gland provide structure and regulate its development and homeostasis. Alterations in its structure can regulate and support pathogenesis, like breast tumors. Aiming to identify the health and tumoral canine mammary ECM scaffold protein profile by immunohistochemistry, the decellularization process was carried out to remove the cellular content. Additionally, it was verified the influence of health and tumoral ECM on the attachment of health and tumoral cells. The types I, III, IV, and V structural collagens were scarce in the mammary tumor, and ECM fibers were disorganized. Vimentin and CD44 were more common in mammary tumor stroma, suggesting a role in cell migration that results in tumor progression. Elastin, fibronectin, laminin, vitronectin, and osteopontin were similarly detected under healthy and tumor conditions, providing the attachment of normal cells in healthy ECM, while tumoral cells were able to attach in tumoral ECM. The protein pattern demonstrates ECM alteration in canine mammary tumorigenesis, presenting new knowledge on mammary tumor ECM microenvironment.
Article
Fibronectin (FN), an extracellular matrix (ECM) glycoprotein, is a well-known marker for Epithelial Mesenchymal Transition (EMT). In the ECM, FN has been shown to form long fibrils and play critical roles in regulating cellular attachment and migration during EMT associated with physiological processes such as embryonic development, wound healing as well as pathological processes such as tissue fibrosis and cancer. Subsequently, the cytokine, Transforming Growth Factor b (TGFb), an inducer of EMT, was found to induce FN expression in a c-Jun N-terminal kinase (JNK) dependent manner. Moreover, extracellular FN, by itself, was also shown to induce EMT in breast epithelial cells in serum-free condition. Collectively, all the literature published so far has shown and established the role of extracellular FN during EMT. In this report, we have shown that EMT induced entry of FN into the nucleus of mouse breast epithelial cells. To our knowledge, this is the first report showing nuclear localization of the extracellular matrix protein Fibronectin during EMT and thereby suggests a possible nuclear function for the ECM protein.
Article
Full-text available
Cell migration underlies tissue formation, maintenance, and regeneration as well as pathological conditions such as cancer invasion. Structural and molecular determinants of both tissue environment and cell behavior define whether cells migrate individually (through amoeboid or mesenchymal modes) or collectively. Using a multiparameter tuning model, we describe how dimension, density, stiffness, and orientation of the extracellular matrix together with cell determinants-including cell-cell and cell-matrix adhesion, cytoskeletal polarity and stiffness, and pericellular proteolysis-interdependently control migration mode and efficiency. Motile cells integrate variable inputs to adjust interactions among themselves and with the matrix to dictate the migration mode. The tuning model provides a matrix of parameters that control cell movement as an adaptive and interconvertible process with relevance to different physiological and pathological contexts.
Article
Full-text available
Background: Colorectal cancer is the third most frequently diagnosed cancer and the third cause of cancer deaths in the United States. Despite the fact that tumor cell-intrinsic mechanisms controlling colorectal carcinogenesis have been identified, novel prognostic and diagnostic tools as well as novel therapeutic strategies are still needed to monitor and target colon cancer progression. We and others have previously shown, using mouse models, that the extracellular matrix (ECM), a major component of the tumor microenvironment, is an important contributor to tumor progression. In order to identify candidate biomarkers, we sought to define ECM signatures of metastatic colorectal cancers and their metastases to the liver. Methods: We have used enrichment of extracellular matrix (ECM) from human patient samples and proteomics to define the ECM composition of primary colon carcinomas and their metastases to liver in comparison with normal colon and liver samples. Results: We show that robust signatures of ECM proteins characteristic of each tissue, normal and malignant, can be defined using relatively small samples from small numbers of patients. Comparisons with gene expression data from larger cohorts of patients confirm the association of subsets of the proteins identified by proteomic analysis with tumor progression and metastasis. Conclusions: The ECM protein signatures of metastatic primary colon carcinomas and metastases to liver defined in this study, offer promise for development of diagnostic and prognostic signatures of metastatic potential of colon tumors. The ECM proteins defined here represent candidate serological or tissue biomarkers and potential targets for imaging of occult metastases and residual or recurrent tumors and conceivably for therapies. Furthermore, the methods described here can be applied to other tumor types and can be used to investigate other questions such as the role of ECM in resistance to therapy.
Article
Full-text available
Collective cell migration is essential for both physiological and pathological processes. Adherens junctions (AJs) maintain the integrity of the migrating cell group and promote cell coordination while allowing cellular rearrangements. Here, we show that AJs undergo a continuous treadmilling along the lateral sides of adjacent leading cells. The treadmilling is driven by an actin-dependent rearward movement of AJs and is supported by the polarized recycling of N-cadherin. N-cadherin is mainly internalized at the cell rear and then recycled to the leading edge where it accumulates before being incorporated into forming AJs at the front of lateral cell-cell contacts. The polarized dynamics of AJs is controlled by a front-to-rear gradient of p120-catenin phosphorylation, which regulates polarized trafficking of N-cadherin. Perturbation of the GSK3-dependent phosphorylation of p120-catenin impacts on the stability of AJs, and the polarity and speed of leading cells during collective migration.
Article
Epithelial cells require attachment to the extracellular matrix (ECM) for survival. However, during tumour progression and metastasis, cancerous epithelial cells must adapt to and survive in the absence of ECM. During the past 20 years, several cellular changes, including anoikis, have been shown to regulate cell viability when cells become detached from the ECM. In this Opinion article, we review in detail how cancer cells can overcome or take advantage of these specific processes. Gaining a better understanding of how cancer cells survive during detachment from the ECM will be instrumental in designing chemotherapeutic strategies that aim to eliminate ECM-detached metastatic cells.
Article
Focal adhesion kinase (FAK) is a cytoplasmic protein tyrosine kinase that is overexpressed and activated in several advanced-stage solid cancers. FAK promotes tumour progression and metastasis through effects on cancer cells, as well as stromal cells of the tumour microenvironment. The kinase-dependent and kinase-independent functions of FAK control cell movement, invasion, survival, gene expression and cancer stem cell self-renewal. Small molecule FAK inhibitors decrease tumour growth and metastasis in several preclinical models and have initial clinical activity in patients with limited adverse events. In this Review, we discuss FAK signalling effects on both tumour and stromal cell biology that provide rationale and support for future therapeutic opportunities.
Article
Epithelial cells display dynamic behaviours, such as rearrangement, movement and shape changes, particularly during embryonic development and in equivalent processes in adults. Accumulating evidence suggests that the remodelling of cell junctions, especially adherens junctions (AJs), has major roles in controlling these behaviours. AJs comprise cadherin adhesion receptors and cytoplasmic proteins that associate with them, including catenins and actin filaments, and exhibit various forms, such as linear or punctate. Remodelling of AJs induces epithelial reshaping in various ways, including by planar-polarized apical constriction that is driven by the contraction of AJ-associated actomyosin and that occurs during neural plate bending and germband extension. RHO GTPases and their effectors regulate actin polymerization and actomyosin contraction at AJs during the epithelial reshaping processes.
Article
microRNAs offer tools to identify and treat invasive cancers. Using highly invasive isogenic oral squamous cell carcinoma (OSCC) cells established using in vitro and in vivo selection protocols from poorly invasive parental cell populations, we used microarray expression analysis to identify a relative and specific decrease in miR-491-5p in invasive cells. Lower expression of miR-491-5p correlated with poor overall survival of OSCC patients. miR-491-5p overexpression in invasive OSCC cells suppressed their migratory behavior in vitro and lung metastatic behavior in vivo. We defined the G protein-coupled receptor kinase-interacting protein 1 (GIT1) as a direct target gene for miR-491-5p control. GIT1 overexpression was sufficient to rescue miR-491-5p-mediated inhibition of migration/invasion and lung metastasis. Conversely, GIT1 silencing phenocopied the ability of miR-491-5p to inhibit migration/invasion and metastasis of OSCC cells. Mechanistic investigations indicated that miR-491-5p overexpression or GIT1 attenuation reduced focal adhesions, with a concurrent decrease in steady-state levels of paxillin, phospho-paxillin, phospho-FAK, EGF/EGFR-mediated ERK1/2 activation and MMP2/9 levels and activities. In clinical specimens of OSCC, GIT1 levels were elevated relative to paired normal tissues and were correlated with lymph node metastasis, with expression levels of miR-491-5p and GIT1 correlated inversely in OSCC where they informed tumor grade. Together, our findings identify a functional axis for OSCC invasion that suggests miR-491-5p and GIT1 as biomarkers for prognosis in this cancer.
Article
Many cancer patients suffer from metastatic relapse several years after they have undergone radical surgery. Early cancer cell dissemination followed by a protracted period of dormancy potentially explains this prevalent clinical behavior. Increasing evidence suggests that the metastasis-initiating cells are cancer stem cells or revert to this functional state upon infiltrating a target organ. Their entry into dormancy and subsequent reactivation are governed by intrinsic programs and by contextual cues, which resemble those regulating the self-renewal capability of adult stem cells. In addition, metastatic cells undergoing reactivation are nursed by specialized extracellular matrix niches, which support positive signals, such as Wnt and Notch, and attenuate negative signals, such as BMP. In spite of significant remaining uncertainties, these findings provide a framework to understand the logic of metastatic dormancy and reactivation and open new avenues for therapeutic intervention.