ArticlePDF Available

3D collagen migration patterns reveal a SMAD3-dependent and TGF-β1-independent mechanism of recruitment for tumour-associated fibroblasts in lung adenocarcinoma

Authors:

Abstract and Figures

Background The TGF-β1 transcription factor SMAD3 is epigenetically repressed in tumour-associated fibroblasts (TAFs) from lung squamous cell carcinoma (SCC) but not adenocarcinoma (ADC) patients, which elicits a compensatory increase in SMAD2 that renders SCC-TAFs less fibrotic. Here we examined the effects of altered SMAD2/3 in fibroblast migration and its impact on the desmoplastic stroma formation in lung cancer. Methods We used a microfluidic device to examine descriptors of early protrusions and subsequent migration in 3D collagen gels upon knocking down SMAD2 or SMAD3 by shRNA in control fibroblasts and TAFs. Results High SMAD3 conditions as in shSMAD2 fibroblasts and ADC-TAFs exhibited a migratory advantage in terms of protrusions (fewer and longer) and migration (faster and more directional) selectively without TGF-β1 along with Erk1/2 hyperactivation. This enhanced migration was abrogated by TGF-β1 as well as low glucose medium and the MEK inhibitor Trametinib. In contrast, high SMAD2 fibroblasts were poorly responsive to TGF-β1, high glucose and Trametinib, exhibiting impaired migration in all conditions. Conclusions The basal migration advantage of high SMAD3 fibroblasts provides a straightforward mechanism underlying the larger accumulation of TAFs previously reported in ADC compared to SCC. Moreover, our results encourage using MEK inhibitors in ADC-TAFs but not SCC-TAFs.
Mechanistic insights on the intrinsic basal migration priming of shSMAD3 fibroblasts and ADC-TAFs: role of high glucose, insulin-transferrin-selenium and Erk1/2. a-c Average migration assessed using the Boyden Transwell assay of shSMAD2 and shSMAD3 control fibroblasts (#5) in the absence (a) or presence of TGF-β1 (b), and corresponding fold values (c). The same plots including shControl fibroblasts are shown in Supplementary Fig. 5A, B. Bottom panels show representative images of the porous membrane of the Transwell insert containing the migratory fibroblasts stained with crystal violet at the end of the experimental time-window (16 h) henceforth. d, e Average Transwell migration of shControl versus shSMAD3 ADC-TAFs (#37) (d) and siControl versus siSMAD3 ADC-TAFs (#13) (e) in the absence of TGF-β1. SMAD3 mRNA levels of siControl and siSMAD3 ADC-TAFs are shown in Supplementary Fig. 5C. f, g Average Transwell migration with low or standard high glucose concentration in either shSMAD2 and shSMAD3 control fibroblasts (#5) (f) or siControl and siSMAD3 ADC-TAFs (#13) (g) in the absence of TGF-β1 (i.e. basal conditions). Low glucose conditions started 3 days before seeding cells for the migration experiment. Basal average Transwell migration with low or high glucose in the presence or absence of insulin-transferrin-selenium (ITS) are shown in Supplementary Fig. 5F, G. h, i Representative Western blot analysis of phosphorylated Erk1/2 (pErk1/2), total Erk1/2 and loading (α-tubulin) of shSMAD2 and shSMAD3 fibroblasts cultured as in f and examined 30 min after seeding. Corresponding densitometric values of pErk1/2/α-tubulin are shown at the bottom (h) thereafter. Average densitometry ratio of pErk1/2 / α-tubulin in shSMAD2 with respect to shSMAD3 are shown in i. j Western blot of pErk1/2, total Erk1/2 and loading of siControl and siSMAD3 ADC-TAFs (#13) cultured as in h. Densitometry ratio of pErk1/2/α-tubulin in siControl with respect to siSMAD3 is shown at the bottom. k-m Average Transwell migration with or without the MEK1/2 inhibitor Trametinib (100 nM) in either shSMAD2 or shSMAD3 control fibroblasts (#5) (k), shControl and shSMAD3 ADCTAFs (#37) (l) or siControl and siSMAD3 ADC-TAFs (#13) (m) in the absence of TGF-β1. Error bars represent mean ± s.e.m. # p < 0.05; ### p < 0.005 comparing shSMAD2 and shSMAD3. *p < 0.05; ***p < 0.005 with respect to shControl. + p < 0.05; +++ p < 0.005 comparing either low and high glucose or the presence and absence of Trametinib. Statistical comparisons were done using Student's t test. Mean values correspond to n ≥ 2 experiments.
… 
Content may be subject to copyright.
ARTICLE OPEN
Cellular and Molecular Biology
3D collagen migration patterns reveal a SMAD3-dependent
and TGF-β1-independent mechanism of recruitment for
tumour-associated broblasts in lung adenocarcinoma
Yago Juste-Lanas
1,2
, Natalia Díaz-Valdivia
3
, Alejandro Llorente
3,4
, Rafael Ikemori
3
, Alejandro Bernardo
3
,
Marselina Arshakyan
3
, Carlos Borau
1
, Josep Ramírez
5,6
, José Carlos Rufnelli
7,8
, Ernest Nadal
7,8
, Noemí Reguart
6,9
,
José M. García-Aznar
1
and Jordi Alcaraz
3,4,6
© The Author(s) 2022
BACKGROUND: The TGF-β1 transcription factor SMAD3 is epigenetically repressed in tumour-associated broblasts (TAFs) from
lung squamous cell carcinoma (SCC) but not adenocarcinoma (ADC) patients, which elicits a compensatory increase in SMAD2 that
renders SCC-TAFs less brotic. Here we examined the effects of altered SMAD2/3 in broblast migration and its impact on the
desmoplastic stroma formation in lung cancer.
METHODS: We used a microuidic device to examine descriptors of early protrusions and subsequent migration in 3D collagen
gels upon knocking down SMAD2 or SMAD3 by shRNA in control broblasts and TAFs.
RESULTS: High SMAD3 conditions as in shSMAD2 broblasts and ADC-TAFs exhibited a migratory advantage in terms of
protrusions (fewer and longer) and migration (faster and more directional) selectively without TGF-β1 along with Erk1/2
hyperactivation. This enhanced migration was abrogated by TGF-β1 as well as low glucose medium and the MEK inhibitor
Trametinib. In contrast, high SMAD2 broblasts were poorly responsive to TGF-β1, high glucose and Trametinib, exhibiting impaired
migration in all conditions.
CONCLUSIONS: The basal migration advantage of high SMAD3 broblasts provides a straightforward mechanism underlying the
larger accumulation of TAFs previously reported in ADC compared to SCC. Moreover, our results encourage using MEK inhibitors in
ADC-TAFs but not SCC-TAFs.
British Journal of Cancer; https://doi.org/10.1038/s41416-022-02093-x
INTRODUCTION
Lung cancer is the leading cause of cancer mortality worldwide,
with a 5-year survival rate of ~19% [1]. Histologically, non-small
cell lung cancer (NSCLC) is diagnosed in ~85% of lung cancer
patients, and is classied into adenocarcinoma (ADC; ~50%),
squamous cell carcinoma (SCC; ~40%) and other less frequent
subtypes [2]. SCC tumours are strongly associated with smoking
and are commonly located in proximal airways, whereas ADC
typically arise in distal pulmonary sites [2]. Although both ADC
and SCC are epithelial in origin, it is increasingly recognised that
the desmoplastic/brotic stroma rich in tumour-associated
broblasts (TAFs, also referred to as cancer-associated bro-
blasts or CAFs), play a key role in tumour progression and
therapy resistance [3,4]. Accordingly, there is growing interest
in understanding the aberrant behaviour of broblasts in solid
tumours [5].
Most lung TAFs exhibit an activated/myobroblast-like pheno-
type [6,7], which is roughly characterised by the intracellular
expression of α-smooth muscle actin (α-SMA) and the abundant
extracellular deposition of brillar collagens [3]. Transforming
growth factor-β1 (TGF-β1) is the most potent broblast activator
known to date and is frequently upregulated in NSCLC. Moreover,
both TGF-β1 and TAF activation markers are associated with poor
prognosis in NSCLC [7,8]. Intriguingly, we recently reported that
TAF activation and associated brosis is higher in ADC compared
to SCC, owing to the larger epigenetic repression of the important
pro-brotic transcription factor SMAD3 of the canonical TGF-β
pathway selectively in SCC-TAFs, caused by their increased
Received: 11 March 2022 Revised: 19 November 2022 Accepted: 25 November 2022
1
Department of Mechanical Engineering, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain.
2
Department of Biochemistry and
Molecular and Cellular Biology, University of Zaragoza, 50009 Zaragoza, Spain.
3
Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and
Health Sciences, Universitat de Barcelona, 08036 Barcelona, Spain.
4
Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST),
08028 Barcelona, Spain.
5
Pathology Service, Hospital Clínic de Barcelona, 08036 Barcelona, Spain.
6
Thoracic Oncology Unit, Hospital Clinic Barcelona, 08036 Barcelona, Spain.
7
Department of Medical Oncology, Catalan Institute of Oncology, LHospitalet de Llobregat (Barcelona), 08908 Barcelona, Spain.
8
Preclinical and Experimental Research in
Thoracic Tumors (PrETT) group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), LHospitalet de Llobregat (Barcelona), 08908 Barcelona, Spain.
9
Institut
dInvestigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain. email: jalcaraz@ub.edu
www.nature.com/bjc
British Journal of Cancer
1234567890();,:
exposure to cigarette smoke particles. We also showed that the
epigenetic repression of SMAD3 elicited a compensatory increase
in the expression and activity of its closely related homologue
SMAD2 in SCC-TAFs, which is weakly associated with brosis [9].
Consistently, ADC-TAFs but not SCC-TAFs exhibited a positive
response to the antibrotic drug nintedanib in culture [10],
thereby mimicking the therapeutic benets reported by ninteda-
nib in ADC but not in SCC patients in the LUME-1 clinical trial [11].
Likewise, both the lower expression of brosis markers and the
poor nintedanib response of SCC-TAFs could be reproduced in
normal broblasts upon knocking down SMAD3 with shRNA,
whereas knocking down SMAD2 had opposite effects, supporting
that shSMAD2 and shSMAD3 broblasts exhibit ADC-like and SCC-
like phenotypes, respectively [9,12].
SMADs 2 and 3 (referred to as SMAD2/3 thereafter) are direct
mediators of canonical TGF-β1 signalling and exhibit some
overlapping functions; however, they also regulate distinct
processes, as reported in knock-out mice [13]. In broblasts,
SMAD2/3 not only differentially regulate brosis and response to
antibrotic drugs [9,14] but they may also control cell migration,
which may be relevant for the formation of the brotic tumour
microenvironment (TME). However, our knowledge of how
SMAD2/3 control cell migration is very limited [15], and their
impact on the formation of the desmoplastic TME in lung cancer is
unknown. Moreover, the few available broblast-specic analyses
of migration regulation by SMAD2/3 were performed in two
dimension (2D) or transwells [15,16], whereas broblast migration
in vivo occurs within a three-dimensional (3D) microenvironment
rich in type I collagen [17]. To address this gap of knowledge, we
knocked down either SMAD2 or SMAD3 in primary human
pulmonary broblasts as surrogate models of ADC-TAFs or SCC-
TAFs, respectively. Fibroblasts were cultured in dense 3D collagen
gels in the absence or presence of TGF-β1 to mimic the
progression of the desmoplastic tumour stroma [18], and the
formation of pro-migratory protrusions and subsequent migration
was analysed. For this purpose, 3D cultures were prepared within
a microuidic device to assess a panel of biophysical descriptors of
protrusions and migration by multidimensional microscopy, and
key ndings were validated with ADC-TAFs.
METHODS
Patient-derived tissue samples and pulmonary broblasts
Primary broblasts were previously obtained from a cohort of 20 NSCLC
surgical patients [6]. Fibroblasts were derived from either tumour or
patient-matched uninvolved pulmonary tissue (referred to as control
broblasts thereafter) using protocols approved by the Ethics Committees
of the Hospital Clinic de Barcelona and the Universitat de Barcelona.
Selected patients were male, chemo-naïve, Caucasian, >55 years old and
current/former smokers. Tumour tissue samples for histological analysis
were obtained from the Hospital de Bellvitge (10 ADC, 9 SCC) with the
approval of the Ethics Committee. The study was performed in accordance
with the Declaration of Helsinki and written informed consent was
obtained from all patients. Further clinical characteristics are shown in
Supplementary Table 1.
Histologic analysis
Tumour samples were processed as described [9], counterstained with
haematoxylin and stained for either cleaved microtubule-associated
protein 1 light chain 3 (LC3A) (#Ap1805a, Abgent), which is largely
negative in broblasts [19], eosin or α-SMA as reported [6]. Fibroblast
nuclear density was assessed by image analysis of haematoxylin staining
using the QuPath software [20] under the guidance of our pathologist (JR).
Further details are provided in Supplementary Materials.
2D cell culture and broblast immortalisation
Control broblasts and ADC-TAFs from randomly selected patients (#5, #13,
#37) were immortalised with hTERT as reported [9]. Unless otherwise
indicated, all broblast experiments were performed in culture medium
containing serum-free high-glucose (4.5 g/l) DMEM supplemented with 1%
insulintransferrinselenium (ITS) and antibiotics as described [10]. In some
experiments, broblasts were stimulated with 2.5 ng/ml recombinant
human TGF-β1 (Miltenyi Biotec) at different time points as indicated, which
is similar to the average TGF-β1 concentration reported in the
bronchoalveolar lavage uid of lung cancer patients [21].
SMAD2 and SMAD3 knock down with shRNA and siRNA
SMAD2 or SMAD3 were stably knocked down in immortalised primary
control broblasts and ADC-TAFs with lentiviral vectors derived from
Sigma MISSION collection as reported [9]. A nonmammalian targeting
shRNA vector was used as control (shControl). Alternatively, SMAD3 was
transiently knocked down by siRNA as described [9]. Further details are
provided in Supplementary Materials.
qRT-PCR
RNA extraction and reverse transcription were conducted as reported
[10,22]. SMAD2/3and MMP1 mRNA levels were assessed using specic
primers, with ACTB or POLR2A as endogenous controls, respectively.
Further details are provided in Supplementary Materials.
Western blot (WB) analysis
WB analysis of SMAD2/3 and Erk1/2 was conducted as described [6,9],
using primary antibodies against total SMAD2/3 (#3102, Cell Signaling),
pSMAD2 (#3104, Cell Signaling), pSMAD3 (#07-1389, Merck Millipore), Erk1/
2, pErk1/2 (#9102 and #9101; Cell Signaling Technology), β-actin (#A1978,
Sigma-Aldrich) and α-tubulin (#2144; Cell Signaling Technology). The latter
two were used as loading controls. Additional details are provided in
Supplementary Materials.
Fabrication of the microuidic device
Microuidic devices were fabricated as described [23], using photomasks
as previously reported [24,25]. In brief, masks were used to fabricate
positive 300 µm high SU8 masters (Stanford University). Polydimethylsilox-
ane (PDMS) (Sylgard 184, Dow Corning) was mixed at a 10:1 weight ratio of
base to curing agent and poured on the SU8 master until the desired
thickness (4 mm) was obtained. The PDMS solution was cured in an oven,
cut out and removed from the wafer, perforated and autocleaved. PDMS
microdevices were plasma-bonded to 35 mm glass-bottom petri dishes
(Ibidi) and coated with 1 mg/ml poly-D-lysine (PDL) (Sigma-Aldrich) to
enhance surface-collagen gel attachment. The geometry of the micro-
device was based on [26] and included a 300 µm high central chamber to
allocate the 3D collagen culture and two parallel liquid channels located
on each side of the central chamber that were in direct contact with the
gel for hydration and transport of nutrients and other factors [25]. Further
details are provided in Supplementary Materials.
3D collagen cell culture within the microuidic device
Collagen hydrogels were prepared as reported [23,25]. Briey, type I
collagen solution (BD Bioscience) was mixed with DPBS (Thermo Fisher
Sci.) and neutralised with NaOH (Sigma-Aldrich) to pH 7.4. For 3D culture
experiments, cells suspended in culture medium were mixed with the
collagen solution to a nal concentration of 4 mg/ml and cell dilution of
2×10
5
cells/ml, which enables local matrix remodelling but not global gel
contraction [17]. The collagen and cells solution were loaded into the
central chamber of the microuidic device using the auxiliary inlet
channels and attached to it through surface tension. The device containing
the collagen and cells solution was placed into an incubator to allow
collagen polymerisation for 30 min. Next, the 3D culture was hydrated with
culture medium and kept in the incubator before experiments.
Analysis of the protrusions of single broblasts in 3D collagen
cultures
Protrusion analysis was performed by adapting our previous protocol [27,28].
Fibroblasts were kept with culture medium with or without 2.5 ng/ml TGF-β1
for 72 h in 2D culture, trypsinised and used to prepare 3D collagen cultures
within the microuidic device. Protrusions were imaged after collagen
polymerisation and up to 4 h based on previous observations [17]as
reported [27,28], using a phase contrast Nikon Eclipse Ti-E inverted
microscope (Nikon, Japan) provided with an incubator. Imaging was
conducted at least 100 µm away from the glass and PDMS surface to avoid
potential edge effects [29]. The whole gel thickness (300μm) was imaged at
Y. Juste-Lanas et al.
2
British Journal of Cancer
5μm intervals (Zaxis) every 5 min at ×200 magnication with a ×20 objective
(CFI Plan Fluor ELWD ADM, NA 0.45; Nikon), eliciting 49 time points and 2940
images/broblast. Best Zplane was chosen for each image, and both the cell
body and the protrusions of randomly selected broblasts were manually
outlined [27,28]. The aspect ratio was computed as major axis/minor axis as
reported [30] (Supplementary Fig. 1). A panel of descriptors was analysed for
each motherprotrusion that stemmed from the cell body (referred to as
protrusions thereafter) [27,31], including length, number, and protrusion and
retraction growth rates, using in-house MATLAB (MathWorks) algorithms. For
each broblast, the absolute values for the protrusion growth and retraction
###
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
✱✱
###
– TGF-
β
1 (basal)– TGF-
β
1 (basal) + TGF-
β
1
TGF-
β
1
cba
fed
ih
g
jlk
SMAD2
SMAD3
β-actin
m
TGF-β1+++
SCC-TAFs
(#22)
β-actin
ADC-TAFs
(#12)
0 min 30 min0 min 30 min
SMAD2
SMAD3
– TGF-
β
1
(0 min)
+ TGF-
β
1
(30 min)
ADC-TAFs
SCC-TAFs
ADC-TAFs
SCC-TAFs
0
1
2
3
0
1
2
3
Fold SMAD3 mRNA
Fold SMAD2 mRNAFold SMAD2 mRNA
Fold SMAD3 mRNA
Fold SMAD3 mRNA
Fold SMAD3/SMAD2 mRNAFold SMAD3/SMAD2 mRNAFold SMAD3/SMAD2
##
###
ADC-TAFs
SCC-TAFs
ADC-TAFs
SCC-TAFs
ADC-TAFs
SCC-TAFs
ADC-TAFs
SCC-TAFs
ADC-TAFs
SCC-TAFs
ADC-TAFs
SCC-TAFs
0.0
0.5
1.0
1.5
2.0
2.5
###
0
1
2
3
0
1
2
3
p = 0.1
0.0
0.5
1.0
1.5
2.0
2.5 #
#
p = 0.09
0.0
0.5
1.0
1.5
Fold SMAD3/SMAD2
0.0
0.5
1.0
1.5 p = 0.08
0.0
0.5
1.0
1.5
Fold SMAD2 mRNA
0.0
0.5
1.0
1.5
Fold SMAD3/SMAD2
0.0
0.5
1.0
1.5
Y. Juste-Lanas et al.
3
British Journal of Cancer
rates were averaged to indicate protrusion evolution rate. A total of 4
broblasts/condition were examined from 3 independent experiments,
which required analysing >8000 images in total.
Analysis of the migration of single broblasts in 3D collagen
cultures
Random broblast migration was analysed using our previous protocols
[24,25,32]. Fibroblasts were kept with culture medium with or without
2.5 ng/ml TGF-β1 for 48 h in 2D culture, trypsinised and used in 3D
collagen cultures within the microuidic device. 3D cultures were kept with
or without TGF-β1 for 24 h within the device, and subsequently imaged for
24 h with a phase contrast inverted microscope provided with an
incubator. Images were acquired every 20 minutes in a manually selected
Zplane at ×100 magnication using a ×10 air objective (CFI Plan Fluor DLL,
NA 0.30, WD 16.0 mm; Nikon) as described [24,33]. Imaging was
conducted at least 100 µm away from the glass and PDMS surface to
avoid potential artifacts. The trajectories of randomly selected broblasts
were tracked and used to compute a panel of migration descriptors with
in-house MATLAB algorithms [24,34], including average cell speed, net
displacement and cell persistence. On average, 35 broblasts were
analysed for each device, and 190 per condition.
Boyden Transwell migration assay
Fibroblasts migration was performed using the Transwell Boyden assay as
reported [35]. In brief, broblasts were maintained for 3 days in serum-free
culture medium with or without 2.5 ng/ml TGF-β1 before seeding them on
Transwell inserts. Culture medium alone or supplemented with 2.5 ng/ml
TGF-β1 was added to the lower Transwell compartment, and cells that
migrated into the lower insert membrane side after 16 h were xed,
stained with crystal violet and imaged by phase-contrast microscopy with
a ×10 objective. Migration was assessed as percentage of positively stained
image area with Image J. In some experiments, broblasts were treated
with 100 nM Trametinib (Selleckchem).
Fibroblast number density
Cell number density of TAFs in 2D cultures was assessed as reported [6]. In
brief, TAFs were cultured in serum-free medium with or without 2.5 ng/ml
TGF-β1 for 5 days and their nuclei were stained with Hoechst 33342
(Molecular Probes) and imaged with a ×10 objective. Number density in the
same 3D cultures used for 3D migration analysis were assessed by manually
counting cells imaged at the end of the experiment. For each experiment,
number density was determined as the average cell density/image.
Statistical analysis
Two-group comparisons were performed with either two-tailed Studentst
test or MannWhitney test for non-parametric data (GraphPad Prism v9.0.).
Statistical signicance was assumed at p< 0.05. All experiments were
conducted at least in triplicates (3 microuidic devices). All data shown
are mean ± s.e.m.
RESULTS
The relative differences in SMAD2/3 expression between ADC-
TAFs and SCC-TAFs can be mimicked through shRNA in
control pulmonary broblasts
To characterise the differences in SMAD2/3 in ADC-TAFs and SCC-
TAFs in more detail, we examined SMAD2/3 expression in TAFs
from randomly selected patients in the absence or presence of
TGF-β1 and without normalising by patient-matched control
broblasts as in our previous study [9]. In basal conditions (i.e.
absence of TGF-β1) SMAD3 mRNA was signicantly higher in ADC-
TAFs than SCC-TAFs (Fig. 1a), whereas SMAD2 mRNA exhibited the
opposite trend (Fig. 1b), eliciting a markedly higher SMAD3/SMAD2
mRNA ratio in ADC-TAFs (Fig. 1c). These histotype differences were
maintained in response to TGF-β1 (Fig. 1df) and are in agreement
with the larger epigenetic repression of SMAD3 in SCC-TAFs [9].
Similar histotype differences in SMAD3/SMAD2 ratio were found at
the protein level (Fig. 1gi and Supplementary Fig. 2), and
collectively reveal that ADC-TAFs exhibit high SMAD3 mRNA and
SMAD3/SMAD2 expression ratio, whereas SCC-TAFs exhibit high
SMAD2 mRNA and lower SMAD3/SMAD2 expression ratio. Con-
sistently, we previously showed that the response to TGF-β1is
dominated by the activation through phosphorylation of either
SMAD3 in ADC-TAFs or SMAD2 in SCC-TAFs, concomitantly with a
higher expression of brosis markers in ADC-TAFs compared to
SCC-TAFs [9].
To model the SMAD2/3 differences observed in TAFs, we stably
knocked down SMAD2 or SMAD3 by shRNA in control broblasts
derived from uninvolved pulmonary tissue of a randomly selected
surgical lung cancer patient (#5). In basal conditions, shSMAD2
broblasts exhibited higher SMAD3 mRNA (Fig. 1j) and protein
expression (Fig. 1m) than shSMAD3 broblasts as in ADC-TAFs.
Conversely, shSMAD3 broblasts exhibited the largest SMAD2
mRNA (Fig. 1k) and protein levels (Fig. 1m) as in SCC-TAFs, which
elicited the largest SMAD3/SMAD2 mRNA ratio in shSMAD2
broblasts (Fig. 1l). In further agreement with TAFs, the response
of shSMAD2 broblasts to exogenous TGF-β1 was dominated by
the activation through phosphorylation of SMAD3 as in ADC-TAFs,
whereas that of shSMAD3 broblasts was dominated by the
phosphorylation of SMAD2 (Supplementary Fig. 2) as in SCC-TAFs
[9]. Accordingly, and in agreement with previous studies [9,10],
shSMAD2 and shSMAD3 broblasts were used as ADC-like and
SCC-like models henceforth, respectively.
Analysis of protrusions in 3D collagen gels reveals that high
SMAD3 conditions as in ADC-TAFs primes broblasts for
migration in the absence of exogenous TGF-β1
Membrane protrusions are pointed as critical regulators of cell
migration in 3D [31,36]. We used a microdevice-based assay [27]
to monitor a panel of protrusion descriptors in single broblasts
embedded in a dense 3D collagen gel within the rst 4 h as
outlined in Fig. 2a. Protrusion analysis was limited to 4 broblasts
per condition owing to the large number of images involved in
each broblast analysis. All broblasts initially exhibited a round
morphology with a dendritic network of protusions (Fig. 2b, h and
Supplementary Fig. 3A, B). In basal conditions, the average
number of protrusions uctuated around 47, with a modest
increase within the rst 1 h followed by a slow decline in shSMAD2
(ADC-like) and shControl broblasts down to 4, whereas they
remained stable around 5 in shSMAD3 (SCC-like) broblasts
(Fig. 2c, e). In contrast, protrusion length increased with time
Fig. 1 Genetic models to mimic SMAD2/3 alterations in patient-derived ADC-TAFs and SCC-TAFs. acFold SMAD3 (a) and SMAD2 (b) mRNA
and corresponding ratio (c) in primary lung TAFs cultured in 2D for 3 days in basal conditions (i.e. without exogenous TGF-β1) (3 ADC, 3 SCC).
dfFold SMAD3 (d) and SMAD2 (e) mRNA and corresponding ratio (f) in primary lung TAFs cultured in 2D for 3 days in the presence of 2.5 ng/
ml TGF-β1 (7 ADC, 5 SCC). gRepresentative Western blot for total SMAD2, SMAD3 and β-actin of ADC-TAFs and SCC-TAFs from randomly
selected patients at 0 min or 30 min after stimulation with TGF-β1. h,iDensitometry analysis of total SMAD3/SMAD2 ratio in TAFs from
randomly selected patients (3 ADC, 3 SCC) at 0 min (h) or 30 min (i) after stimulation with TGF-β1. jlFold SMAD3 (j), SMAD2 (k) and SMAD3/
SMAD2 mRNA ratio (l) of shControl, shSMAD2 and shSMAD3 control broblasts from patient #5 cultured in 2D for 3 days in basal conditions.
mRepresentative Western blot for total SMAD2, SMAD3 and β-actin of shControl, shSMAD2 and shSMAD3 control broblasts (#5) in basal
conditions. Error bars represent mean ± s.e.m. Each dot corresponds to a different patient (ai).
#
p< 0.05;
##
p< 0.01;
###
p< 0.005 comparing
either ADC-TAFs and SCC-TAFs or shSMAD2 and shSMAD3. **p< 0.01; ***p< 0.005 with respect to shControl. Statistical comparisons were
done using Studentsttest.
Y. Juste-Lanas et al.
4
British Journal of Cancer
selectively in shSMAD2 broblasts up to 50 μm, whereas it
reached a plateau in the last 12 h that was the lowest in
shSMAD3 broblasts (Fig. 2d). Accordingly, protrusion number and
length were averaged within the last hour (34 h) henceforth and
were found consistently higher in shSMAD2 compared to
shSMAD3 broblasts in terms of length (Fig. 2f) and growth/
retraction evolution rate (Fig. 2g), with average values of ~40 μm
and 0.015 μm/s, respectively. Conversely, protrusion number was
0
2
4
6
Aspect ratio
0
2
4
6
Aspect ratio
p = 0.09
0
2
4
6
8
10
p = 0.1
0
2
4
6
8
10
Avg
basal
Avg
basal
Avg
basal
a
bc d
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
– TGF-
β
1 (basal)+ TGF-
β
1
efg
hji
klm
npo
– TGF-
β
1 + TGF-
β
1
– TGF-
β
1 + TGF-
β
1
4h
±TGFβ1±TGFβ1
72h [ 2D ] 4h [ 3D ]
Imaging
20 mm Culture medium
Best-focused z-plane
Culture medium
Collagen & cell
input channels
4h
50 μm
40 μmprot./ret
rates
6
73
5
4
1
2
Protrusion
length
50 μm
0
10
20
30
40
50
60
Protrusions length (
μ
m)
Protrusions length (
μ
m)Protrusions length (
μ
m)
Protrusions length (
μ
m)
#
0.000
0.005
0.010
0.015
0.020 #
0
10
20
30
40
50
60
p = 0.08
0.000
0.005
0.010
0.015
0.020
Protrusions evolution
rate (
μ
m/s)
Protrusions evolution
rate (
μ
m/s)
#
#
01234
0
10
20
30
40
50
60
Time (h)
01234
0
2
4
6
8
10
Time (h)
Number of protrusionNumber of protrusionsNumber of protrusionsNumber of protrusions
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
01234
0
2
4
6
8
10
Time (h)
01234
0
10
20
30
40
50
60
Time (h)
shSMAD2
(ADC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
50 μm
Y. Juste-Lanas et al.
5
British Journal of Cancer
lower in shSMAD2 than shSMAD3 broblasts with marginal
signicance (Fig. 2e).
Unlike basal conditions, all TGF-β1-preactivated broblasts
exhibited a low initial number of protrusions (1-3) that increased
within the rst 1 h up to 7, although at different rates, in which
shSMAD3 broblasts were the fastest (Fig. 2i). In contrast,
protrusion length increased over time at a much lower rate than
in basal conditions, particularly in shSMAD2 broblasts, which
attained a protrusion length of ~10 μm (Fig. 2j). Globally, TGF-β1
elicited an average 50% increase of protrusion number
compared to basal conditions (Fig. 2k and Supplementary Fig. 3C),
whereas it reduced both protrusion length and evolution rate by
>50% in shSMAD2 conditions, and these descriptors remained
stable in shSMAD3 broblasts (Fig. 2l, m and Supplementary
Fig. 3D, E), suggesting that shSMAD3 broblasts are poorly
responsive to TGF-β1. Since longer and fewer protrusions have
been previously associated with enhanced migration [31], these
results suggest that shSMAD2 (ADC-like) broblasts may be
primed for migration selectively in basal conditions. Consistently,
the aspect ratio at 4 h, which is indicative of polarisation along a
major protrusion (Supplementary Fig. 1) and has been positively
associated with migration [36], was the highest in shSMAD2
broblasts in basal conditions (Fig. 2n, o), whereas it was the
lowest upon TGF-β1 preactivation (Fig. 2p).
Analysis of migration in 3D collagen gels conrms that high
SMAD3 as in ADC-TAFs enhances migration selectively in
basal conditions
Next, we adapted the microuidic device assay to analyse single
broblast migration within 2448 h in 3D culture as outlined in
Fig. 3a. All broblasts exhibited the archetypical elongated
morphology found in histologic sections [6,17] (Fig. 3b). In
agreement with protrusion analysis, shSMAD2 (ADC-like) bro-
blasts exhibited signicantly enhanced migration in basal condi-
tions, as illustrated by their larger trajectories (Fig. 3c) and
signicantly higher values of all migration descriptors compared
to shSMAD3 and shControl broblasts (Fig. 3df), with an average
speed of ~0.10 μm/min (Fig. 3d), total net displacement of ~50 μm
(Fig. 3e) and directional persistence of ~0.4 (Fig. 3f) that were
~30% (speed), ~90% (displacement) and ~40% (persistence) larger
than the corresponding values of shSMAD3 broblasts. To validate
our observations, we analysed ADC-TAFs from patient #37 upon
knocking down SMAD3 with shRNA (Fig. 3g) in the absence of
exogenous TGF-β1. Although migration descriptors in ADC-TAFs
(#37) (Fig. 3hl) were lower than those found in control broblasts
(Fig. 3df), shControl ADC-TAFs exhibited in average a larger
effective speed (Fig. 3j), net displacement (Fig. 3k) and persistence
(Fig. 3l) than shSMAD3 ADC-TAFs (#37).
The basal migratory advantage of shSMAD2 broblasts was
abrogated upon TGF-β1 stimulation, and broblasts in all
conditions exhibited similar trajectories (Fig. 4a, b), speed (Fig. 4c),
net displacement (Fig. 4d) and persistence (Fig. 4e). Moreover,
even though TGF-β1 barely reduced the average cell speed in
shSMAD2 with respect to basal conditions (Fig. 4f), it elicited a
marked reduction in the average persistence and associated net
displacement (Fig. 4g, h), revealing that TGF-β1 promotes the
spatial connement of ADC-like broblasts. In contrast, shSMAD3
broblasts were poorly responsive to TGF-β1, exhibiting a small
increase in speed and displacement compared to basal settings
(Fig. 4f, g) in further agreement with protrusion analysis. A similar
trend was observed in shControl compared to shSMAD3 ADC-TAFs
(#37) in terms of speed and persistence (Fig. 4km); however,
these differences did not attain statistical signicance due to the
large variability associated with their globally low speed and
directionality. These results suggest that the spatial connement
of ADC-TAFs is evident even in the absence of exogenous TGF-β1.
The strong qualitative agreement between early protrusions
(04 h) and subsequent migration data (2448 h) encouraged us
to conduct a correlation analysis between descriptors of both
processes. Both migration persistence and net displacement were
strongly and positively correlated (R
2
> 0.7) with all protrusion
descriptors but protrusion number (Supplementary Fig. 4EL),
with the highest correlations consistently observed with aspect
ratio (Supplementary Fig. 4K, L). In contrast, we observed a poor
correlation between migration speed and all protrusions descrip-
tors (Supplementary Fig. 4A, D, G, J). These results further support
the notion that those conditions that elicit fewer and longer
protrusions may help polarise the cell along a major protrusion
and facilitate directed movements, whereas increased number of
protrusions is associated with reduced migration and subsequent
spatial connement [31,36].
The enhanced migration of high SMAD3 broblasts in basal
conditions is independent of collagen degradation
Because our migration analysis was conducted in broblasts
embedded in a dense collagen matrix with a expected range of
pore sizes comparable or even lower than the typical width of
elongated broblasts [37], it is possible that the enhanced basal
migration of high SMAD3 broblasts is driven by increased
collagen degradation rather than intrinsic migratory priming. To
address this question, we analysed migration in the complete
absence of exogenous extracellular matrix (ECM) degradation
using the Boyden Transwell assay without any ECM coating in the
porous Transwell insert membrane. In agreement with our 3D
migration data, the percentage of cells that migrated through the
insert membrane was markedly higher in shSMAD2 broblasts
Fig. 2 Impact of altered SMAD2/3 in 3D collagen protrusions of lung broblasts and ADC-TAFs with or without exogenous TGF-β1.
aOutline of the microdevice-based analysis of protrusions (04 h) of single broblasts cultured in dense 3D collagen gels. bRepresentative
phase contrast images of single control broblasts (#5) for each group (shControl, shSMAD2, shSMAD3) cultured in 3D collagen gels within
the microdevice for 4 h in basal conditions (TGF-β1). Scale bar, 50 μm. Representative images at other time points are shown in
Supplementary Fig. 3A, B. c,dTime-course of the average number of protrusions (c) and protrusion length (d)of4broblasts for each group
cultured in 3D in basal conditions. egNumber of protrusions (e), protrusion length (f) and evolution rate (g) averaged for 4 broblasts per
group within the last 1 h (34 h) of the experimental time-window (e,f) or the full experiment (04h) (g) in basal conditions (shown as bars).
Each dot indicates the average of a single broblast henceforth. hRepresentative phase contrast images of single control broblasts (#5) for
each group (shControl, shSMAD2, shSMAD3) cultured in 3D collagen gels within the microdevice for 4 h in the presence of TGF-β1. Scale bar,
50 μm. Representative images at other time points are shown in Supplementary Fig. 3A, B. i,jTime-course of the average number of
protrusions (i) and protrusion length (j)of4broblasts for each group cultured in 3D in the presence of TGF-β1. kmNumber of protrusions
(k), protrusion length (l) and evolution rate (m) averaged for 4 broblasts per group as in egin the presence of TGF-β1. nRepresentative
phase contrast images of the aspect ratio analysis of single shSMAD2 broblasts cultured in 3D collagen gels for 4 h without or with TGF-β1.
Scale bar, 50 μm. Further details on the assessment of the aspect ratio shown in Supplementary Fig. 1. o,pAspect ratio averaged for 4
broblasts per group at the end of the experimental time-window (4 h) in basal conditions (o) or in the presence of TGF-β1(p). Error bars
represent mean ± s.e.m. Each dot corresponds to the average of a different broblast (eg,km,o,p) examined within 3 independent
microdevices.
#
p< 0.05 comparing shSMAD2 and shSMAD3. Other p-values comparing to shControl. Statistical comparisons were done using
Studentsttest.
Y. Juste-Lanas et al.
6
British Journal of Cancer
compared to shSMAD3 in basal settings (Fig. 5a and Supplemen-
tary Fig. 5A). Likewise, the migration advantage of shSMAD2
broblasts was attenuated by TGF-β1, whereas shSMAD3 bro-
blasts were poorly responsive (Fig. 5b, c and Supplementary
Fig. 5B). Consistent differences were observed in ADC-TAFs from
randomly selected patients (#13, #37) in which SMAD3 had been
knocked down by shRNA (#37) (Fig. 3g) or siRNA (#13)
(Supplementary Fig. 5C) compared to parental cells in Transwells
0.00
0.05
0.10
0.15
0.20
0.25
Cell speed (μ
μ
m/min)
###
a
b
c
f
de
– TGF-
E
1 (basal) – TGF-
E
1 (basal)
– TGF-
E
1 (basal)
– TGF-
E
1 (basal)
shControl shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
24 h
36 h
48 h
0
50
100
150
200
250
300
Net displacement (
μ
m)
###
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1.0
Persistence ratio
###
ADC-TAF (#37)
shControl
ADC-TAF (#37)
shSMAD3
h
24 h 36 h 48 h
50 μm
50 μm
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1.0
Persistence ratio
0
50
100
150
200
250
300
Net displacement (
μ
m)
p = 0.1
0.00
0.05
0.10
0.15
0.20
0.25
Cell speed (
μ
m/min)
0.00
0.25
0.50
0.75
1.00
1.25
1.50
Fold SMAD3 mRNA
ADC-TAFs (#37)
shSMAD3
ADC-TAFs (#37)
shControl
ADC-TAFs (#37)
shSMAD3
ADC-TAFs (#37)
shControl
ADC-TAFs (#37)
shSMAD3
ADC-TAFs (#37)
shControl
ADC-TAFs (#37)
shSMAD3
ADC-TAFs (#37)
shControl
g
jkl
i
– TGF-
E
1 (basal)
250
–250
–250 250
250
–250
–250 250
250
–250
–250 250
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
100 μm
100–100
–100
100 μm
–100
100–100
±TGFβ1 ±TGFβ1 ±TGFβ1
48h [ 2D ] 24h [ 2D ] 24h [ 3D ]
20 mm
Collagen & cell
input channels
Culture medium
Trajectory = rd
rd
nd
Net displacement = nd
Persistence = nd / rd
Cell speed = rd / time
Culture medium
Imaging
Y. Juste-Lanas et al.
7
British Journal of Cancer
in the absence of TGF-β1 (Fig. 5d, e). Likewise, migration of ADC-
TAFs in Transwells was globally lower than control broblasts as in
3D. In contrast, we did not observe signicant differences in the
mRNA levels of the major collagenase MMP1 between either ADC-
TAFs and SCC-TAFs from randomly selected patients (Supplemen-
tary Fig. 5D) or between shSMAD2 and shSMAD3 broblasts
(Supplementary Fig. 5E). These ndings reveal that the enhanced
basal migration of high SMAD3 broblasts is largely independent
of collagen degradation but rather due to an intrinsic priming of
their migratory properties.
The larger migration of high SMAD3 broblasts in basal
conditions requires high glucose-dependent Erk1/2
hyperactivation
Prompted by the consistent migratory advantage observed in
high SMAD3 conditions in both 3D collagen gels and Transwell
assays selectively in the absence of TGF-β1, we began to explore
the underlying mechanisms. A common exogenous factor in both
assays was the presence of high glucose in the culture medium,
which are standard conditions for the cell culture of broblasts
and TAFs [38,39]. Moreover, Erk1/2 can be activated by both high
glucose and 3D collagen [40,41], have been strongly implicated in
the regulation of migration in numerous cell types including
broblasts [42], and may interact with SMAD3 in the absence of
TGF-β1[43]. To examine the potential involvement of high
glucose and/or Erk1/2 activation, we rst analysed basal Transwell
migration with either standard high glucose (4.5 g/l) or low
glucose (1 g/l) medium and found a signicant increased
migration with high glucose in shSMAD2 but not in shSMAD3
conditions. In contrast, low glucose medium abrogated the
migration differences between shSMAD2 and shSMAD3 broblasts
(Fig. 5f). Likewise, Transwell migration was signicantly increased
in siControl ADC-TAFs (#13) in high versus low glucose conditions,
whereas no differences were observed in siSMAD3 ADC-TAFs
(Fig. 5g). Another supplemented soluble factor present in all
assays was insulin-transferrin-selenium (ITS). Because a major
function of insulin in cell culture is to stimulate glucose entry [44],
we also analysed Transwell migration in the presence or absence
of ITS and found a signicant migration increase (~35%) with ITS
in low glucose conditions selectively in shSMAD2 broblasts,
whereas such migration increase was attenuated in high glucose
conditions (~12%) (Supplementary Fig. 5F, G), further under-
scoring the requirement of high glucose in the basal migration
priming of high SMAD3 broblasts. In contrast, shSMAD3
broblasts were consistently poorly responsive to both high
glucose and ITS in terms of migration (Fig. 5f and Supplementary
Fig. 5F, G).
Regarding Erk1/2 activation, we found that phosphorylated
Erk1/2 (pErk1/2) levels normalised by α-tubulin were increased by
~70% in standard high glucose in shSMAD2 compared to
shSMAD3 broblasts, whereas such increase was reduced to
~20% in low glucose conditions (Fig. 5h, i and Supplementary
Fig. 5H, I). Likewise, normalised pErk1/2 levels were ~35% higher
in siControl compared to siSMAD3 ADC-TAFs (#13) in our standard
high glucose medium (Fig. 5j); however, this difference was
smaller than that found between shSMAD2 and shSMAD3
broblasts, possibly due to the high endogenous expression of
TGF-β1 in ADC-TAFs [9], which may alter pErk1/2 [45] and
therefore bias the response to high glucose. In further qualitative
agreement with migration data, normalised pErk1/2 levels were
higher in shSMAD2 compared to shSMAD3 broblasts even in the
absence of ITS (Supplementary Fig. 5J). On the other hand, total
Erk1/2 levels remained fairly stable in all conditions (Fig. 5h, j and
Supplementary Fig. 5I). These results unveil a SMAD3-dependent
hyperactivation of Erk1/2 in the presence of high glucose.
To assess whether pErk1/2 hyperactivation is required for the
basal migration priming of high SMAD3 broblasts, we analysed
basal Transwell migration in the presence of 100 nM Trametinib, a
clinically approved inhibitor of MEK1/2 MAP kinases in NSCLC and
melanoma that acts right upstream of Erk1/2 [46]. Of note,
Trametinib signicantly downregulated basal migration in stan-
dard high glucose medium in shSMAD2 but not shSMAD3
broblasts (Fig. 5k). Consistently, Trametinib elicited a drop in
basal Transwell migration in ADC-TAFs (#37, #13) in control
conditions but not upon knocking down SMAD3 in the presence
of standard high glucose (Fig. 5l, m). Yet, we noticed that the
migration reduction elicited by Trametinib varied between ~20-
80% depending on the cell model, even though Erk1/2 activation
was strongly abrogated in all cases (Supplementary Fig. 5K, L),
suggesting that additional molecular events other than pErk1/2
hyperactivation contribute to the migratory differences between
high SMAD3 and high SMAD2 broblasts in high glucose
conditions. Collectively, these results implicate high glucose-
dependent Erk1/2 hyperactivation in the migratory advantage of
high SMAD3 broblasts in the absence of TGF-β1, and reveal that
high SMAD2 broblasts are poorly responsive to both high
glucose/ITS and MEK1/2 inhibition.
The enhanced basal migration of high SMAD3 broblasts is
consistent with the larger accumulation of TAFs observed in
ADC compared to SCC at early stages
Finally, we examined the potential contribution of our observed
relationship between altered SMAD2/3 expression and migration
to the excessive accumulation of TAFs in lung cancer, since we
previously reported a larger TAF density in histologic samples in
ADC compared to SCC patients [6] in a small patient cohort (5
ADC, 5 SCC) (re-analysed as number density/image eld in
Supplementary Fig. 6a). We conrmed this observation by
Fig. 3 Impact of altered SMAD2/3 in 3D collagen migration in lung broblasts and ADC-TAFs in basal conditions (no exogenous TGF-β1).
aOutline of the microdevice-based analysis of migration (2448 h) of single broblasts cultured in dense 3D collagen gels. bRepresentative
phase contrast images of single control broblasts (#5) for each group (shControl, shSMAD2, shSMAD3) cultured in 3D collagen gels within
the microdevice in basal conditions during the experimental time-window (2448 h). Yellow, green and red dots indicate the position of the
broblast centre at 24, 36 and 48 h, respectively. Scale bar, 50 μm. cTrajectory maps corresponding to the tracking of the centre of broblasts
from each group in basal conditions throughout the experimental time-window. The starting of each broblast was shifted to the origin of
coordinates for clarity here and thereafter. dfAverage cell speed (d), net displacement (e) and persistence ratio (f) in basal conditions of
single control broblasts (#5) for each group (shControl, shSMAD2, shSMAD3) gathered from 3 independent microdevices per condition
(shown as bars). Each dot indicates the average of a single broblast henceforth. Note that persistence values range between 0 and 1, where 1
indicates migration without changing direction [30]. gFold SMAD3 mRNA of shControl and shSMAD3 ADC-TAFs (#37) cultured as in Fig. 1j.
hRepresentative phase contrast images of single shControl or shSMAD3 ADC-TAFs (#37) cultured as in b. Yellow, green and red dots indicate
the position of the broblast centre at 24, 36 and 48 h, respectively. Scale bar, 50 μm. iTrajectory maps corresponding to the tracking of the
centre of ADC-TAFs from each group in basal conditions throughout the experimental time-window as in c.jlAverage cell speed (j), net
displacement (k) and persistence ratio (l) of single ADC-TAFs (#37) for each group (shown as bars) cultured as in b. Error bars represent
mean ± s.e.m.
###
p< 0.005 comparing shSMAD2 and shSMAD3. *p< 0.05; ***p< 0.005 with respect to shControl or comparing shControl and
shSMAD3 ADC-TAFs. Statistical comparisons were done using Studentsttest or MannWhitney (df). Mean values correspond to three
independent microdevices.
Y. Juste-Lanas et al.
8
British Journal of Cancer
assessing the number density of TAFs in an independent patient
cohort (Hospital de Bellvitge; 10 ADC, 9 SCC), identied by their
elongated nuclei (Fig. 6a top) and conrmed by their α-SMA
expression (Fig. 6a bottom), and found consistent results (Fig. 6b).
TAF accumulation is thought to arise largely from the
recruitment and/or proliferation of local resident broblasts
[5,47]. From the recruitment perspective, our observed migratory
advantage without TGF-β1 in high SMAD3 broblasts is consistent
0.0
0.5
1.0
1.5
Fold cell speed
(+TGF-E
E
1/–TGF-
E
1)
###
0.0
0.5
1.0
1.5
0.0
0.5
1.0
1.5
Fold net displacement
(+TGF-
E
1/–TGF-
E
1)
###
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1.0
Persistence ratio
a
b
e
cd
+ TGF-
β
1 + TGF-
β
1
+ TGF-
β
1
+ TGF-
β
1
+ TGF-
β
1
shControl shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
24 h
36 h
48 h
0.00
0.05
0.10
0.15
0.20
0.25
Cell speed (
μ
m/min)
0
50
100
150
200
250
300
Net displacement (
μ
m)
fgh
ADC-TAFs (#37)
shControl
ADC-TAFs (#37)
shSMAD3
ADC-TAFs (#37)
shControl
ADC-TAFs (#37)
shSMAD3
ADC-TAFs (#37)
shControl
ADC-TAFs (#37)
shSMAD3
kl m
ADC-TAF (#37)
shControl
ADC-TAF (#37)
shSMAD3
i
24 h 36 h 48 h
Fold persistence ratio
(+TGF-
E
1/–TGF-
E
1)
0.0
0.5
1.0
1.5
Fold cell speed
(+TGF-
E
1/–TGF-
E
1)
0.0
0.5
1.0
1.5
0.0
0.5
1.0
1.5
Fold net displacement
(+TGF-
E
1/–TGF-
E
1)
Fold persistence ratio
(+TGF-
E
1/ –TGF-
E
1)
###
50 μm
50 μm
j
250 μm
–250
–250 250
250 μm
–250
–250 250
250 μm
–250
–250 250
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
100 μm
100–100
–100
100 μm
100–100
–100
Y. Juste-Lanas et al.
9
British Journal of Cancer
with a larger recruitment of TAFs in ADC patients selectively at
early stages, when active TGF-β1 is expected to be low [18]. To
assess the potential contribution of differential proliferation, we
examined cell number densitywhich is a common growth
metric [5]in different culture settings. Unlike histologic sections,
ADC-TAFs exhibited a signicantly lower number density in basal
conditions compared to SCC-TAFs from randomly selected
patients in 2D cultures (Fig. 6c). Likewise, basal number density
was lower in shSMAD2 (ADC-like) broblasts compared to
shSMAD3 both in 2D (Supplementary Fig. 6B) and in the same
3D cultures used in our migration studies (Fig. 6e). In contrast, all
these differences were abrogated in the presence of TGF-β1
(Fig. 6d, f and Supplementary Fig. 6C). Moreover TGF-β1
consistently elicited a 10-20% increase in number density in
ADC-TAFs (Fig. 6d) and shSMAD2 broblasts (Fig. 6f) compared to
basal conditions, whereas such increase was not observed in SCC-
TAFs, in agreement with our previously observed poor response of
SCC-TAFs to soluble mitogenic cues [6]. Since we previously
reported that differences in number density between ADC-TAFs
and SCC-TAFs in 2D cultures were largely attributed to prolifera-
tion changes [6], our results support that altered SMAD2/3
expression elicits a proliferation advantage in terms of number
density selectively in high SMAD2 broblasts as in SCC-TAFs in
basal conditions that is not consistent with the larger TAF
accumulation observed in ADC. Collectively these ndings
strongly support that the larger histologic TAF accumulation in
ADC is driven, at least in part, by the enhanced migration of ADC-
TAFs caused by their high SMAD3 expression at early stages, when
active TGF-β1 is expected to be low [18], as summarised in Fig. 6g.
DISCUSSION
SMAD2/3 are important transcription factors of the canonical TGF-
β1 pathway that are expressed in virtually all cell types [13,45]. In
broblasts, the TGF-β1/SMAD3 pathway has been extensively
documented as a positive regulator of brosis in the lung and
other organs [9,14,48]. In contrast, previous studies on the role of
SMAD2/3 in broblast migration are scarce and limited to 2D
cultures or transwells [15,16], which do not capture the
physiologic complexity of 3D microenvironments. To address
these limitations, we used a microuidic device to examine for the
rst time a panel of protrusions and migration descriptors of
single broblasts in dense 3D collagen gels. Our gel density was
>50% higher than that commonly used in other studies (1.52 mg/
ml) [17,29,31,49] to mimic the high collagen content reported in
lung cancer patients [7]. In addition, we used a technical
improvement in the protrusion analysis by checking different Z
planes instead of a single Zplane as commonly reported
[29,36,49].
We found that SMAD2/3 have a markedly distinct impact on
migration depending on the TGF-β1 context. Specically, our
results revealed for the rst time that high SMAD3 conditions (as
in shSMAD2 broblasts and ADC-TAFs) provide a migratory
advantage in terms of protrusions (fewer and longer) and
subsequent migration (faster and more directional) compared to
high SMAD2 conditions (as in shSMAD3 broblasts and SCC-TAFs)
selectively in the absence of exogenous TGF-β1, whereas TGF-β1
markedly abrogated this migratory advantage, promoting the
spatial connement of broblasts. In contrast, high SMAD2
conditions were poorly responsive to TGF-β1 and consistently
exhibited the largest number of protrusions concomitantly with
the shortest protrusion length with and without TGF-β1, which
were subsequently associated with impaired migration through
less directional movement and a shorter net displacement.
Consistently, downregulating SMAD3 in ADC-TAFs was sufcient
to reduce basal migration in 3D and in Transwells. In agreement
with our ndings, TGF-β1 was associated with reduced migration
in keratinocytes [50] and cardiac broblasts in Transwells [16].
Likewise, SMAD3 null cardiac broblasts exhibited a lower
migration than wild-type cells upon 1% FBS stimulation in
Transwells [16], in agreement with our high SMAD2 observations.
In contrast, unlike our 3D ndings, no migratory effects were
observed in cardiac broblasts stimulated with 1% FBS upon
knocking down either SMAD2 or SMAD3 with siRNA using
Transwells [51], supporting that our multiparametric
microdevice-based 3D migration analysis may be more sensitive
in detecting migration changes in response to altered SMAD2/3
expression.
High SMAD3 broblasts were also the most sensitive to the
presence of exogenous TGF-β1, exhibiting more and shorter
protrusions that elicited a less directional movement and
subsequently a shorter net displacement, thereby increasing their
spatial connement. Indeed, because TGF-β1 increased the
number of broblast protrusions in all SMAD2/3 settings and it is
known to increase traction forces in TAFs [52], it is likely that TGF-
β1 elicits more simultaneous traction in different (random)
directions, yielding an ineffective (non-persistent) movement
despite holding or increasing cell speed. In line with this
interpretation, ADC-TAFs exhibited lower basal migration than
control broblasts, which is consistent with the higher basal
secretion of TGF-β1 and expression of the contractility marker α-
SMA [9,53] in ADC-TAFs. In qualitative agreement with our
observations, TGF-β1 did not promote migration in cardiac
broblasts in Transwells [16], or even impaired it in keratinocytes
[50]. In contrast, our results are not consistent with the TGF-β1
stimulation of in vitro wound healing reported in human vocal
cord broblasts and keratinocytes or with its downregulation upon
SMAD3 inhibition using a scratch assay [15,54], since we observed
either no migratory changes in 3D or even a moderate increase in
Transwells in shSMAD3 broblasts in the presence of TGF-β1.
However, the marked differences between the scratch assay and
our 3D/Transwell assays may account for this discrepancy.
Fig. 4 Impact of altered SMAD2/3 in 3D collagen migration in lung broblasts and ADC-TAFs stimulated with TGF-β1. a Representative
phase contrast images of single control broblasts (#5) for each group (shControl, shSMAD2, shSMAD3) cultured in 3D collagen gels within
the microdevice with TGF-β1 during the experimental time-window (2448 h). Yellow, green and red dots indicate the position of the
broblast centre at 24 h, 36 h and 48 h, respectively. Scale bar, 50 μm. bTrajectory maps corresponding to the tracking of the centre of
broblasts from each group in basal conditions throughout the experimental time-window. ceAverage cell speed (c), net displacement (d)
and persistence ratio (e) with TGF-β1 of single control broblasts (#5) for each group (shControl, shSMAD2, shSMAD3) gathered from three
independent microdevices per condition (shown as bars). Each dot indicates the average of a single broblast. fhFold average cell speed (f),
net displacement (g) and persistence ratio (h) assessed in the presence and absence of TGF-β1. Error bars were computed using error
propagation [67]. iRepresentative phase contrast images of single shControl or shSMAD3 ADC-TAFs (#37) cultured as in a. Yellow, green and
red dots indicate the position of the broblast centre at 24, 36 and 48 h, respectively. Scale bar, 50 μm. jTrajectory maps corresponding to the
tracking of the centre of ADC-TAFs from each group with TGF-β1 throughout the experimental time-window as in b.kmFold average cell
speed (k), net displacement (l) and persistence ratio (m) of ADC-TAFs (#37) for each group assessed in the presence and absence of TGF-β1.
###
p< 0.005 comparing shSMAD2 and shSMAD3. **p< 0.01; ***p< 0.005 with respect to shControl or comparing shControl and shSMAD3
ADC-TAFs. Statistical comparisons were done using Studentsttest or MannWhitney (ce). Mean values correspond to three independent
microdevices.
Y. Juste-Lanas et al.
10
British Journal of Cancer
0
20
40
60
0
20
40
60
Migration (% area)
0
20
40
60
Migration (% area)
0
20
40
60
Migration (% area)
0
20
40
60
Migration (% area)
###
Low glucose High glucose
dbac
Fold migration
(+TGf
β
/-TGF
β
)
#
0.0
0.5
1.0
1.5 #
– TGF-
β
1+ TGF-
β
1– TGF-
β
1– TGF-
β
1
f
Migration (% area)
0
20
40
60
Migration (% area)
Glucose
concentration:
1 g/L 4.5 g/L 1 g/L 4.5 g/L
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
ADC-TAFs (#13)
siControl
ADC-TAFs (#13)
siSmad3
0
20
40
60
2.0
1.5
1.0
0.5
0.0
+
1 g/L 4.5 g/L
#
#
+
+
+++
1 g/L 4.5 g/L
hg
klm
ADC-TAFs (#37)
shSmad3
ADC-TAFs (#37)
shControl
-+ -+
i
25
μ
m
25
μ
m25
μ
m
25
μ
m
+
ADC-TAFs (#13)
siSmad3
ADC-TAFs (#13)
siControl
-+ -+
25
μ
m
pErk1/2
shSMAD3
shSMAD2
shSMAD3
shSMAD2
Erk1/2
D-Tubulin
0.86 0.53
0.92 0.74
pErk1/2
DTubulin :
1.24 1.62
ratio shSMAD2
shSMAD3:
– TGF-
β
1 (basal)
e
25
μ
m
j
pErk1/2
Erk1/2
D-Tubulin
Low glucose High glucose
ADC-TAFs (#13)
1.98 1.47
0.37 0.59
0.62 1.35
pErk1/2
DTubulin :
ratio
siControl
siSMAD3:
pErk1/2 /
α
-tubulin
shSMAD2 / shSMAD3
Migration (% area)
0
20
40
60
Migration (% area)
0
20
40
60
Migration (% area)
shSMAD3
(SCC-like)
MEK inhibitor:
(Trametinib) -+ -+
shSMAD2
(ADC-like)
25
μ
m
Glucose
concentration: 1 g/L 4.5 g/L
siSMAD3
siControl
siSMAD3
siControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
ADC-TAFs (#37)
shSMAD3
ADC-TAFs (#37)
shControl
ADC-TAFs (#37)
shSMAD3
ADC-TAFs (#37)
shControl
25
μ
m25
μ
m
Fig. 5 Mechanistic insights on the intrinsic basal migration priming of shSMAD3 broblasts and ADC-TAFs: role of high
glucose, insulintransferrinselenium and Erk1/2. acAverage migration assessed using the Boyden Transwell assay of shSMAD2 and
shSMAD3 control broblasts (#5) in the absence (a) or presence of TGF-β1(b), and corresponding fold values (c). The same plots including
shControl broblasts are shown in Supplementary Fig. 5A, B. Bottom panels show representative images of the porous membrane of the
Transwell insert containing the migratory broblasts stained with crystal violet at the end of the experimental time-window (16 h) henceforth.
d,eAverage Transwell migration of shControl versus shSMAD3 ADC-TAFs (#37) (d) and siControl versus siSMAD3 ADC-TAFs (#13) (e) in the
absence of TGF-β1. SMAD3 mRNA levels of siControl and siSMAD3 ADC-TAFs are shown in Supplementary Fig. 5C. f,gAverage Transwell
migration with low or standard high glucose concentration in either shSMAD2 and shSMAD3 control broblasts (#5) (f) or siControl and
siSMAD3 ADC-TAFs (#13) (g) in the absence of TGF-β1 (i.e. basal conditions). Low glucose conditions started 3 days before seeding cells for the
migration experiment. Basal average Transwell migration with low or high glucose in the presence or absence of insulin-transferrin-selenium
(ITS) are shown in Supplementary Fig. 5F, G. h,iRepresentative Western blot analysis of phosphorylated Erk1/2 (pErk1/2), total Erk1/2 and
loading (α-tubulin) of shSMAD2 and shSMAD3 broblasts cultured as in fand examined 30 min after seeding. Corresponding densitometric
values of pErk1/2/α-tubulin are shown at the bottom (h) thereafter. Average densitometry ratio of pErk1/2 / α-tubulin in shSMAD2 with respect
to shSMAD3 are shown in i.jWestern blot of pErk1/2, total Erk1/2 and loading of siControl and siSMAD3 ADC-TAFs (#13) cultured as in h.
Densitometry ratio of pErk1/2/α-tubulin in siControl with respect to siSMAD3 is shown at the bottom. kmAverage Transwell migration with
or without the MEK1/2 inhibitor Trametinib (100 nM) in either shSMAD2 or shSMAD3 control broblasts (#5) (k), shControl and shSMAD3 ADC-
TAFs (#37) (l) or siControl and siSMAD3 ADC-TAFs (#13) (m) in the absence of TGF-β1. Error bars represent mean ± s.e.m.
#
p< 0.05;
###
p< 0.005
comparing shSMAD2 and shSMAD3. *p< 0.05; ***p< 0.005 with respect to shControl.
+
p< 0.05;
+++
p< 0.005 comparing either low and high
glucose or the presence and absence of Trametinib. Statistical comparisons were done using Studentsttest. Mean values correspond to n2
experiments.
Y. Juste-Lanas et al.
11
British Journal of Cancer
Collectively, our results clarify the migration regulation of SMAD2/3
in broblasts in 3D and how it depends on the TGF-β1 context.
TGF-β1 inhibits growth through SMAD3 in numerous cell types
including epithelial cells and keratinocytes [5557]. In contrast, we
observed cytostatic effects in high SMAD3 conditions in the
absence of TGF-β1 only, whereas TGF-β1 elicited a moderate
but consistent increase in number density particularly in high
SMAD3 broblasts and ADC-TAFs. Likewise, a TGF-β1-dependent
a
ADC
SCC
0.0
0.5
1.0
1.5
2.0 #
#
bH. Bellvitge cohort
Cell density
e
-TGF-
β
1
3D
p = 0.06 p = 0.09
p = 0.09
f
c
d
2D
360
45
30
15
0
60
45
30
15
0
2
1
0
3
2
1
0
+ TGF-
β
1
ADC-TAFs
SCC-TAFs
Cell density
g
α-SMA H&E
ADC
20 μm
SCC
20 μm
EARLY STAGE
( low active TGF-β1)
↑↑ Migration
Fast
recruitment
High SMAD3 fibroblasts
(as in ADC-TAFs)
Early cancer
cell
dissemination?
LATE STAGE
(high active TGF-β1)
Spatial
confinement
Cytostasis
Tumour evolution
↓↓ Migration Number
density
Mild TAF
expansion
Long-term TAF-cancer cell
crosstalk
MEK inh
Cell density Cell density
Fold fibroblasts density
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
ADC-TAFs
SCC-TAFs
shControl
shSMAD2
(ADC-like)
shSMAD3
(SCC-like)
Fig. 6 Differential TAF number density in ADC and SCC and potential contribution of SMAD2/3-regulated migration versus proliferation.
aIllustrative haematoxylineosin (H&E) (top) and α-SMA (bottom) staining of ADC and SCC patients. Arrow heads point to scattered and
spindle-shaped nuclei, which are histologic hallmarks of TAFs. bTAF number density assessed by morphometric analysis of haematoxylin
images from the Hospital de Bellvitge patient cohort (10 ADC, 9 SCC). Independent validation with a smaller cohort is shown in Supplementary
Fig. 6A. c,dFibroblast number density of TAFs randomly selected from our cohort (5 ADC, 5 SCC) cultured in 2D in the absence (c) or presence
(d) of TGF-β1. e,fFibroblast number density of the control broblasts (#5) from all groups (shControl, shSMAD2, shSMAD3) cultured within the
3D collagen gels used to analyse migration in the absence (e) or presence (f) of TGF-β1. Corresponding values of broblasts cultured in 2D as
in care shown in Supplementary Fig. 6B, C. gEmerging model for the impact of the interplay between SMAD3, migration and proliferation in
ADC-TAFs in early and late stages. Error bars represent mean ± s.e.m. Each dot corresponds to a different patient (bd).
#
p< 0.05; comparing
either ADC-TAFs and SCC-TAFs or shSMAD2 and shSMAD3. Other p-values with respect to shControl. Statistical comparisons were done using
Studentsttest. Mean values correspond to n2 experiments.
Y. Juste-Lanas et al.
12
British Journal of Cancer
proliferation increase was reported in dermal broblasts [58],
further supporting that TGF-β1 is not an effective cytostatic
cytokine in broblasts. Although our mechanistic understanding
of these TGF-β1-independent pro-migratory and cytostatic func-
tions in high SMAD3 broblasts is limited, our results implicated
high glucose-dependent Erk1/2 hyperactivation in the migratory
advantage. Erk1/2 are important regulators of migration in
numerous cell types [42], and we found that standard high
glucose culture medium markedly increased both migration and
pErk1/2 selectively in high SMAD3 broblasts compared to low
glucose conditions. Conversely, the MEK inhibitor Trametinib
consistently attenuated basal migration in high SMAD3 but not
high SMAD2 broblasts. Our results are in agreement with
previous work reporting high glucose-dependent Erk1/2 hyper-
activation in kidney cells [41], and implicate for the rst time
SMAD3 in this hyperactivation and a subsequent migration
enhancement in broblasts.
The molecular underpinnings of these new SMAD3-specic
broblast functions remain to be determined. Yet, it is unlikely
that they involve the C-terminal residues of SMAD3 that become
phosphorylated in response to TGF-β1 as part of the canonical
TGF-β1/SMAD3 pathway [59], since we previously showed that
these residues remain unphosphorylated in the basal conditions
(i.e. absence of TGF-β1) [9] in which the migration enhancement
was observed. Alternatively, the linker region of SMAD3 could be
implicated, since this region contains several phosphorylation
sites that can be regulated by Erk1/2 and other kinases in the
absence of TGF-β1[43,45], and there is growing evidence that the
linker regions may modulate different cell functions in SMAD2 and
SMAD3 independently of TGF-β1[43,60]. Moreover, it has been
shown that high glucose enhanced the sensitivity to exogenous
TGF-β1 in mouse embryonic broblasts and kidney epithelial cells
[41,61], and the SMAD3 linker was involved in this enhancement
[41], in agreement with our observed strongest response to TGF-
β1 selectively in high SMAD3 broblasts. On the other hand, it is
worth noting that a larger glucose uptake was recently reported in
ADC-TAFs compared to SCC-TAFs [62], which could also contribute
to our observed high glucose-dependent Erk1/2 activation in high
SMAD3 conditions as in ADC-TAFs. However, our current knowl-
edge of the Erk1/2-SMAD2/3 crosstalk in the absence of TGF-β1is
still very scarce and warrants further investigations. Likewise, it
would be interesting to assess how our observed high SMAD2-
driven poor response to MEK inhibition in broblasts may
contribute to the resistance to MEK inhibitors in cancer [63].
Our ndings provide new insights on how TAFs may contribute
to tumour progression in lung cancer depending on the TGF-β1
context and the histologic subtype. Under low active TGF-β1
conditions as in early tumour stages [18], the enhanced migration
of high SMAD3 broblasts may be a major contributor to the
larger TAF recruitment observed in ADC compared to SCC. In
addition, because TAFs can promote cancer cell dissemination by
leading collective cancer cell migration [64], it is conceivable that
the basal migration priming of high SMAD3 broblasts may
contribute to the early dissemination of cancer cells in ADC, which
is a common clinical observation whose underlying mechanisms
remain unknown [65]. However, testing this hypothesis awaits
future investigations. Moreover, our results support that the
tumour-promoting effects associated with the enhanced migra-
tion of high SMAD3 broblasts could be prevented with
Trametinib or other MEK inhibitors in ADC but not SCC, since
high SMAD2 broblasts as in SCC-TAFs were largely non-
responsive to MEK inhibition. In contrast, at later stages where
active TGF-β1 is expected to be abundant at doses within the
same range used in our experimental settings [18,21], the
impaired migration observed in all SMAD2/3 conditions con-
comitantly with the expected α-SMA-dependent increased con-
tractility may collectively increase the spatial connement of TAFs
and ultimately facilitate the long-term interactions with adjacent
cancer cells to promote their epigenetic reprogramming [52,66]
(Fig. 6g).
In summary, we unveil how altered SMAD2/3 expression
provides migration and proliferation advantages only in the
absence of TGF-β1, although in opposite directions, since
enhanced migration (but not proliferation) was observed selec-
tively in high SMAD3 conditions, strongly supporting that the
larger TAF accumulation in ADC occurs at early stages (under low
active TGF-β1). Moreover, our results provide a rationale for the
implication of ADC-TAFs in the early ADC cancer cell dissemination
observed in clinical settings and for the therapeutic use of MEK
inhibitors against the enhanced migratory phenotype of ADC-
TAFs (Fig. 6g).
DATA AVAILABILITY
The data sets generated and analysed in the study are available from the
corresponding author on reasonable request.
REFERENCES
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin.
2019;69:734.
2. Chen Z, Fillmore CM, Hammerman PS, Kim CF, Wong K-K. Non-small-cell lung
cancers: a heterogeneous set of diseases. Nat Rev Cancer. 2014;14:53546.
3. Kalluri R. The biology and function of broblasts in cancer. Nat Rev Cancer.
2016;16:58298.
4. Ohlund D, Elyada E, Tuveson D. Fibroblast heterogeneity in the cancer wound. J
Exp Med. 2014;211:150323.
5. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. A
framework for advancing our understanding of cancer-associated broblasts. Nat
Rev Cancer. 2020;20:17486.
6. Puig M, Lugo R, Gabasa M, Gimenez A, Velasquez A, Galgoczy R, et al. Matrix
stiffening and beta(1) integrin drive subtype-specicbroblast accumulation in
lung cancer. Mol Cancer Res. 2015;13:16173.
7. Alcaraz J, Carrasco JL, Millares L, Luis I-C, Fernández-Porras FJ, Martínez-Romero
A, et al. Stromal markers of activated tumor associated broblasts predict poor
survival and are associated with necrosis in non-small cell lung cancer. Lung
Cancer. 2019;135:15160.
8. Hasegawa Y, Takanashi S, Kanehira Y, Tsushima T, Imai T, Okumura K. Trans-
forming growth factor-beta 1 level correlates with angiogenesis, tumor pro-
gression, and prognosis in patients with nonsmall cell lung carcinoma. Cancer.
2001;91:96471.
9. Ikemori R, Gabasa M, Duch P, Vizoso M, Bragado P, Arshakyan M, et al. Epigenetic
SMAD3 repression in tumor-associated broblasts impairs brosis and response
to the antibrotic drug nintedanib in lung squamous cell carcinoma. Cancer Res.
2020;80:27690.
10. Gabasa M, Ikemori R, Hilberg F, Reguart N, Alcaraz J. Nintedanib selectively
inhibits the activation and tumor-promoting effects of broblasts from lung
adenocarcinoma patients. Br J Cancer. 2017;117:112838.
11. Reck M, Kaiser R, Mellemgaard A, Douillard J-Y, Orlov S, Krzakowski M, et al.
Docetaxel plus nintedanib versus docetaxel plus placebo in patients with pre-
viously treated non-small-cell lung cancer (LUME-Lung 1): a phase 3, double-
blind, randomised controlled trial. Lancet Oncol. 2014;15:14355.
12. Duch P, Díaz-Valdivia N, Ikemori R, Gabasa M, Radisky ES, Arshakyan M, et al.
Aberrant TIMP-1 overexpression in tumor-associated broblasts drives tumor
progression through CD63 in lung adenocarcinoma. Matrix Biol. 2022;111:20725.
13. Brown KA, Pietenpol JA, Moses HL. A tale of two proteins: differential roles and
regulation of Smad2 and Smad3 in TGF-beta signaling. J Cell Biochem.
2007;101:933.
14. Huang S, Chen B, Humeres C, Alex L, Hanna A, Frangogiannis NG. The role of
Smad2 and Smad3 in regulating homeostatic functions of broblasts in vitro and
in adult mice. Biochimt Biophys Acta Mol Cell Res. 2020;1867:118703.
15. Branski RC, Bing R, Kraja I, Amin MR. The role of Smad3 in the brotic phenotype
in human vocal fold broblasts. Laryngoscope. 2016;126:11516.
16. Dobaczewski M, Bujak M, Li N, Gonzalez-Quesada C, Mendoza LH, Wang X-F, et al.
Smad3 signaling critically regulates broblast phenotype and function in healing
myocardial infarction. Circ Res. 2010;107:41828.
17. Grinnell F, Ho CH, Tamariz E, Lee DJ, Skuta G. Dendritic broblasts in three-
dimensional collagen matrices. Mol Biol Cell. 2003;14:38495.
18. Prudhomme GJ. Pathobiology of transforming growth factor beta in cancer,
brosis and immunologic disease, and therapeutic considerations. Lab Investig.
2007;87:107791.
Y. Juste-Lanas et al.
13
British Journal of Cancer
19. Giatromanolaki A, Koukourakis MI, Harris AL, Polychronidis A, Gatter KC, Sivridis E.
Prognostic relevance of light chain 3 (LC3A) autophagy patterns in colorectal
adenocarcinomas. J Clin Pathol. 2010;63:86772.
20. Bankhead P, Loughrey MB, Fernández JA, Dombrowski Y, McArt DG, Dunne PD,
et al. QuPath: Open source software for digital pathology image analysis. Sci Rep.
2017;7:16878.
21. Domagała-Kulawik J, Hoser G, Saanowska A, Grubek-Jaworska H, Chazan R.
Elevated TGF-beta1 concentration in bronchoalveolar lavage uid from patients
with primary lung cancer. Arch Immunol Ther Exp. 2006;54:1437.
22. Gabasa M, Radisky ES, Ikemori R, Bertolini G, Arshakyan M, Hockla A, et al. MMP1
drives tumor progression in large cell carcinoma of the lung through broblast
senescence. Cancer Lett. 2021;507:112.
23. Shin Y, Han S, Jeon JS, Yamamoto K, Zervantonakis IK, Sudo R, et al. Microuidic
assay for simultaneous culture of multiple cell types on surfaces or within
hydrogels. Nat Protoc. 2012;7:124759.
24. Plou J, Juste-Lanas Y, Olivares V, del Amo C, Borau C, García-Aznar JM. From
individual to collective 3D cancer dissemination: roles of collagen concentration
and TGF-β. Sci Rep. 2018;8:12723.
25. Movilla N, Borau C, Valero C, García-Aznar JM. Degradation of extracellular matrix
regulates osteoblast migration: a microuidic-based study. Bone. 2018;107:1017.
26. Farahat WA, Wood LB, Zervantonakis IK, Schor A, Ong S, Neal D, et al. Ensemble
analysis of angiogenic growth in three-dimensional microuidic cell cultures.
PLoS ONE. 2012;7:e37333.
27. Movilla N, Valero C, Borau C, García-A znar JM. Matrix degradation regulates
osteoblast protrusion dynamics and individual migration. Integr Biol.
2019;11:40413.
28. Merino-Casallo F, Gomez-Benito MJ, Juste-Lanas Y, Martinez-Cantin R, Garcia-
Aznar JM. Integration of in vitro and in silico models using Bayesian optimization
with an application to stochastic modeling of mesenchymal 3D cell migration.
Front Physiol. 2018;9:1246.
29. Giri A, Bajpai S, Trenton N, Jayatilaka H, Longmore GD, Wirtz D. The Arp2/3
complex mediates multigeneration dendritic protrusions for efcient
3-dimensional cancer cell migration. FASEB J. 2013;27:408999.
30. Rahman-Zaman A, Shan S, Reinhart-King CA. Cell migration in microfabricated 3D
collagen microtracks is mediated through the prometastatic protein Girdin. Cell
Mol Bioeng. 2018;11:110.
31. Fraley SI, Feng Y, Krishnamur thy R, Kim D-H, Celedon A, Longmore GD, et al. A
distinctive role for focal adhesion proteins in three-dimensional cell motility. Nat
Cell Biol. 2010;12:598604.
32. Juste-Lanas Y, Guerrero PE, Camacho-Gómez D, Hervás-Raluy S, García-Aznar JM,
Gomez-Benito MJ. Conned cell migration and asymmetric hydraulic environ-
ments to evaluate the metastatic potential of cancer cells. J Biomech Eng.
2022;144:074502.
33. Moreno-Arotzena O, Borau C, Movilla N, Vicente-Manzanares M, García-Aznar JM.
Fibroblast migration in 3D is controlled by haptotaxis in a non-muscle myosin II-
dependent manner. Ann Biomed Eng. 2015;43:302539.
34. Del Amo C, Olivares V, Cóndor M, Blanco A, Santolaria J, Asín J, et al. Matrix
architecture plays a pivotal role in 3D osteoblast migration: the effect of inter-
stitial uid ow. J Mech Behav Biomed Mater. 2018;83:5262.
35. Gabasa M, Arshakyan M, Llorente A, Chuliá-Peris L, Pavelescu I, Xaubet A, et al.
Interleukin-1βmodulation of the mechanobiology of primary human pulmonary
broblasts: potential implications in lung repair. Int J Mol Sci. 2020;21:8417.
36. Carey SP, Goldblatt ZE, Martin KE, Romero B, Williams RM, Reinhart-King CA. Local
extracellular matrix alignment directs cellular protrusion dynamics and migration
through Rac1 and FAK. Integr Biol. 2016;8:82135.
37. Galgoczy R, Pastor I, Colom A, Giménez A, Mas F, Alcaraz J. A spectrophotometer-
based diffusivity assay reveals that diffusion hindrance of small molecules in
extracellular matrix gels used in 3D cultures is dominated by viscous effects.
Colloid Surf B Biointerfaces. 2014;120:2007.
38. Huang W, Zhang L, Yang M, Wu X, Wang X, Huang W, et al. Cancer-associated
broblasts promote the survival of irradiated nasopharyngeal carcinoma cells via
the NF-κB pathway. J Exp Clin Cancer Res. 2021;40:87.
39. Franco-Barraza J, Raghavan KS, Luong T, Cukierman E. Engineering clinically-
relevant human broblastic cell-derived extracellular matrices. Methods Cell Biol.
2020;156:10960.
40. Cukierman E, Pankov R, Stevens DR, Yamada KM. Taking cell-matrix adhesions to
the third dimension. Science. 2001;294:170812.
41. Pan X, Phanish MK, Baines DL, Dockrell MEC. High glucose-induced Smad3 linker
phosphorylation and CCN2 expression are inhibited by dapagliozin in a diabetic
tubule epithelial cell model. Biosci Rep. 2021;41:BSR20203947.
42. Huang C, Jacobson K, Schaller MD. MAP kinases and cell migration. J Cell Sci.
2004;117:461928.
43. Ooshima A, Park J, Kim SJ. Phosphorylation status at Smad3 linker region mod-
ulates transforming growth factor-β-induced epithelial-mesenchymal transition
and cancer progression. Cancer Sci. 2019;110:4818.
44. Howard BV, Mott DM, Fields RM, Bennett PH. Insulin stimulation of glucose entry
in cultured human broblasts. J Cell Physiol. 1979;101:12938.
45. Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-
beta family signalling. Nature. 2003;425:57784.
46. Han J, Liu Y, Yang S, Wu X, Li H, Wang Q. MEK inhibitors for the treatment of non-
small cell lung cancer. J Hematol Oncol. 2021;14:1.
47. Alexander J, Cukierman E. Cancer associated broblast: mediators of tumor-
igenesis. Matrix Biol. 2020;91-92:1934.
48. Meng XM, Huang XR, Chung ACK, Qin W, Shao X, Igarashi P, et al. Smad2 protects
against TGF-beta/Smad3-mediated renal brosis. J Am Soc Nephrol.
2010;21:147787.
49. Fraley SI, Feng Y, Giri A, Longmore GD, Wirtz D. Dimensional and temporal
controls of three-dimensional cell migration by zyxin and binding partners. Nat
Commun. 2012;3:719.
50. Hosokawa R, Urata MM, Ito Y, Bringas P Jr, Chai Y. Functional signicance of
Smad2 in regulating basal keratinocyte migration during wound healing. J Invest
Dermatol. 2005;125:13029.
51. Huang S, Chen B, Su Y, Alex L, Humeres C, Shinde AV, et al. Distinct roles of
myobroblast-specic Smad2 and Smad3 signaling in repair and remodeling of
the infarcted heart. J Mol Cell Cardiol. 2019;132:8497.
52. Vizoso M, Puig M, Carmona FJ, Maqueda M, Velásquez A, Gómez A, et al. Aberrant
DNA methylation in non small cell lung cancer associated broblasts. Carcino-
genesis. 2015;36:145363.
53. Lugo R, Gabasa M, Andriani F, Puig M, Facchinetti F, Ramírez J, et al. Heterotypic
paracrine signaling drives broblast senescence and tumor progression of large
cell carcinoma of the lung. Oncotarget. 2016;7:8232437.
54. Kocic J, Bugarski D, Santibanez JF. SMAD3 is essential for transforming growth
factor-β1-induced urokinase type plasminogen activator expression and migra-
tion in transformed keratinocytes. Eur J Cancer. 2012;48:15507.
55. Uemura M, Swenson ES, Gaça MDA, Giordano FJ, Reiss M, Wells RG. Smad2 and
smad3 play different roles in rat hepatic stellate cell function and alpha-smooth
muscle actin organization. Mol Biol Cell. 2005;16:421424.
56. Kim SG, Kim H-A, Jong H-S, Park J-H, Kim NK, Hong SH, et al. The endogenous
ratio of Smad2 and Smad3 inuences the cytostatic function of Smad3. Mol Biol
Cell. 2005;16:467283.
57. Ashcroft GS, Yang X, Glick AB, Weinstein M, Letterio JL, Mizel DE, et al. Mice
lacking Smad3 show accelerated wound healing and an impaired local inam-
matory response. Nat Cell Biol. 1999;1:2606.
58. Sapudom J, Müller CD, Nguyen K-T, Martin S, Anderegg U, Pompe T. Matrix
remodeling and hyaluronan production by myobroblasts and cancer-associated
broblasts in 3D collagen matrices. Gels. 2020;6:33.
59. Massagué J. How cells read TGF-beta signals. Nat Rev Mol Cell Biol.
2000;1:16978.
60. Kamato D, Do BH, Osman N, Ross BP, Mohamed R, Xu S, et al. Smad linker region
phosphorylation is a signalling pathway in its own right and not only a modulator
of canonical TGF-βsignalling. Cell Mol Life Sci. 2020;77:24351.
61. Wu L, Derynck R. Essential role of TGF-beta signaling in glucose-induced cell
hypertrophy. Dev Cell. 2009;17:3548.
62. Zhang W, Bouchard G, Yu A, Shaq M, Jamali M, Shrager JB, et al. GFPT2-
expressing cancer-associated broblasts mediate metabolic reprogramming in
human lung adenocarcinoma. Cancer Res. 2018;78:344557.
63. Kun E, Tsang YTM, Ng CW, Gershenson DM, Wong KK. MEK inhibitor resistance
mechanisms and recent developments in combination trials. Cancer Treat Rev.
2021;92:102137.
64. Labernadie A, Kato T, Brugués A, Serra-Picamal X, Derzsi S, Arwert E, et al. A
mechanically active heterotypic E-cadherin/N-cadherin adhesion enables bro-
blasts to drive cancer cell invasion. Nat Cell Biol. 2017;19:22438.
65. Hoffman PC, Mauer AM, Vokes EE. Lung cancer. Lancet. 2000;355:47985.
66. Alcaraz J, Ikemori R, Llorente A, Díaz-Valdivia N, Reguart N, Vizoso M. Epigenetic
reprogramming of tumor-associated broblasts in lung cancer: therapeutic
opportunities. Cancers. 2021;13:3782.
67. Taylor JR. An introduction to error analysis. Sausalito: University Science Books;
1997.
ACKNOWLEDGEMENTS
We thank Raimon Sunyer, Pere Roca-Cusachs, Patricia Fernández, Kate Neal (UB),
Zanetta Zoi and Joan Montero (IBEC) for technical support, and Ramon Farré (UB) for
support.
AUTHOR CONTRIBUTIONS
Study concept, design and supervision: JMG-A, JA; acquisition of data: YJ-L, ND-V, AL,
RI, AB, MA, CB, JCR; analysis and interpretation of data: YJ-L, ND-V, AL, JA;
Y. Juste-Lanas et al.
14
British Journal of Cancer
development of the methodology: YJ-L, RI, CB, AL, JMG-A, JA; drafting of the
manuscript: YJ-L, ND-V, AL, JA; provided patient samples and clinical information: JCR,
EN, NR, JR; critical revision of the manuscript: AB, NR, JMG-A; obtained funding: JMG-
A, EN, JA; technical or material support: CB, JR.
FUNDING
This work was supported by grants from the Agencia Estatal de Investigación (AEI/
FEDER) (PI13/02368, SAF2016-79527-R and PID2019-110944RB-I00 to JA, PID2021-
122409OB-C21, RTI2018-094494-B-C21 to JMG-A), European Research Council (Adg-
101018587 to JMG-A), Instituto de Salud Carlos III (PI14/01109, PI18/00920 to EN) (co-
funded by European Regional Development Fund. ERDF, a way to build Europe),
Fundació Privada Cellex (to JA), Generalitat de Catalunya (AGAUR SGR 661 and CERCA
Programme to JA), Junta Provincial de Barcelona de lAssociació Espanyola Contra el
Càncer (AECC B16-917 to JA), Sociedad Española de Neumología y Cirugía Torácica
SEPAR (SEPAR 951/2019 to JA), Spanish Society of Medical Oncology grant for
emerging research groups (to EN), and by fellowships from Spanish Ministry of
Science and Innovation (FPU17/03867 to YJ-L), Ciência sem Fronteiras CNPq (to RI),
CONICYT (to NDV) and IBEC (to ALL).
COMPETING INTERESTS
The authors declare no competing interests.
ETHICS APPROVAL AND CONSENT TO PARTICIPATE
This study was approved by the Institutional Review Board of the Hospital Clínic,
Hospital de Bellvitge and Universitat de Barcelona. Informed consent was obtained
from all participants involved in the study, and all experiments were conducted in
line with the principles of the Declaration of Helsinki.
CONSENT FOR PUBLICATION
There are no individual persons data from all participants involved in the study.
ADDITIONAL INFORMATION
Supplementary information The online version contains supplementary material
available at https://doi.org/10.1038/s41416-022-02093-x.
Correspondence and requests for materials should be addressed to Jordi Alcaraz.
Reprints and permission information is available at http://www.nature.com/
reprints
Publishers note Springer Nature remains neutral with regard to jurisdictional claims
in published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
articles Creative Commons license and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly
from the copyright holder. To view a copy of this license, visit http://
creativecommons.org/licenses/by/4.0/.
© The Author(s) 2022
Y. Juste-Lanas et al.
15
British Journal of Cancer
... 38,39 We found that this regulation holds in ADC-TAFs, 20 compared to SCC-TAFs were attributable to the more extensive epigenetic repression of the SMAD3 promoter in SCC-TAFs. 15,40 The heightened SMAD3 production and activation in ADC-TAFs enhances the secretion of bioactive TGF-β1, creating a positive feedback loop that amplifies the profibrotic activity of the TGF-β1/SMAD3 pathway. 15 Because we showed that nintedanib attenuates the TGF-β1/SMAD3 pathway through the partial inhibition of the TGF-β1 type I receptor ALK5, 15 our current findings strongly support that the hyperactive TGF-β1/SMAD3 pathway in ADC-TAFs is responsible for their excessive production of TIMP-1 and subsequent selective positive response to nintedanib; conversely, the low SMAD3 expression and limited TIMP-1 production in SCC-TAFs may confer intrinsic resistance to this drug. ...
Article
Full-text available
The fibrotic tumor microenvironment is a pivotal therapeutic target. Nintedanib, a clinically approved multikinase antifibrotic inhibitor, is effective against lung adenocarcinoma (ADC) but not squamous cell carcinoma (SCC). Previous studies have implicated the secretome of tumor‐associated fibroblasts (TAFs) in the selective effects of nintedanib in ADC, but the driving factor(s) remained unidentified. Here we examined the role of tissue inhibitor of metalloproteinase‐1 (TIMP‐1), a tumor‐promoting cytokine overproduced in ADC‐TAFs. To this aim, we combined genetic approaches with in vitro and in vivo preclinical models based on patient‐derived TAFs. Nintedanib reduced TIMP‐1 production more efficiently in ADC‐TAFs than SCC‐TAFs through a SMAD3‐dependent mechanism. Cell culture experiments indicated that silencing TIMP1 in ADC‐TAFs abolished the therapeutic effects of nintedanib on cancer cell growth and invasion, which were otherwise enhanced by the TAF secretome. Consistently, co‐injecting ADC cells with TIMP1‐knockdown ADC‐TAFs into immunocompromised mice elicited a less effective reduction of tumor growth and invasion under nintedanib treatment compared to tumors bearing unmodified fibroblasts. Our results unveil a key mechanism underlying the selective mode of action of nintedanib in ADC based on the excessive production of TIMP‐1 in ADC‐TAFs. We further pinpoint reduced SMAD3 expression and consequent limited TIMP‐1 production in SCC‐TAFs as key for the resistance of SCC to nintedanib. These observations strongly support the emerging role of TIMP‐1 as a critical regulator of therapy response in solid tumors.
Article
Full-text available
Metastasis, a hallmark of cancer development, is also the leading reason for most cancer-related deaths. Furthermore, cancer cells are highly adaptable to microenvironments and can migrate along pre-existing channel-like tracks of anatomical structures. However, more representative three-dimensional models are required to reproduce the heterogeneity of metastatic cell migration in vivo to further understand the metastasis mechanism and develop novel therapeutic strategies against it. Here, we designed and fabricated different microfluidic-based devices that recreate confined migration and diverse environments with asymmetric hydraulic resistances. Our results show different migratory potential between metastatic and nonmetastatic cancer cells in confined environments. Moreover, although nonmetastatic cells have not been tested against barotaxis due to their low migration capacity, metastatic cells present an enhanced preference to migrate through the lowest resistance path, being sensitive to barotaxis. This device, approaching the study of metastasis capability based on confined cell migration and barotactic cell decisions, may pave the way for the implementation of such technology to determine and screen the metastatic potential of certain cancer cells.
Article
Full-text available
Lung cancer is the leading cause of cancer-related death worldwide. The desmoplastic stroma of lung cancer and other solid tumors is rich in tumor-associated fibroblasts (TAFs) exhibiting an activated/myofibroblast-like phenotype. There is growing awareness that TAFs support key steps of tumor progression and are epigenetically reprogrammed compared to healthy fibroblasts. Although the mechanisms underlying such epigenetic reprogramming are incompletely understood, there is increasing evidence that they involve interactions with either cancer cells, pro-fibrotic cytokines such as TGF-β, the stiffening of the surrounding extracellular matrix, smoking cigarette particles and other environmental cues. These aberrant interactions elicit a global DNA hypomethylation and a selective transcriptional repression through hypermethylation of the TGF-β transcription factor SMAD3 in lung TAFs. Likewise, similar DNA methylation changes have been reported in TAFs from other cancer types, as well as histone core modifications and altered microRNA expression. In this review we summarize the evidence of the epigenetic reprogramming of TAFs, how this reprogramming contributes to the acquisition and maintenance of a tumor-promoting phenotype, and how it provides novel venues for therapeutic intervention, with a special focus on lung TAFs.
Article
Full-text available
Background: In the kidney glucose is freely filtered by the glomerulus and, mainly, reabsorbed by sodium glucose cotransporter 2 (SGLT2) expressed in the early proximal tubule. Human proximal tubule epithelial cells (PTECs) undergo pathological and fibrotic changes seen in diabetic kidney disease (DKD) in response to elevated glucose. We developed a specific in vitro model of DKD using primary human PTECs with exposure to high D-glucose and TGF-β1 and propose a role for SGLT2 inhibition in regulating fibrosis. Methods: Western blotting was performed to detect cellular and secreted proteins as well as phosphorylated intracellular signalling proteins. qPCR was used to detect CCN2 RNA. Gamma glutamyl transferase (GT) activity staining was performed to confirm PTEC phenotype. SGLT2 and ERK inhibition on high D-glucose, 25 mM, and TGF-β1, 0.75 ng/ml, treated cells was explored using dapagliflozin and U0126, respectively. Results: Only the combination of high D-glucose and TGF-β1 treatment significantly up-regulated CCN2 RNA and protein expression. This increase was significantly ameliorated by dapagliflozin. High D-glucose treatment raised phospho ERK which was also inhibited by dapagliflozin. TGF-β1 increased cellular phospho SSXS Smad3 serine 423 and 425, with and without high D-glucose. Glucose alone had no effect. Smad3 serine 204 phosphorylation was significantly raised by a combination of high D-glucose+TGF-β1; this rise was significantly reduced by both SGLT2 and MEK inhibition. Conclusions: We show that high D-glucose and TGF-β1 are both required for CCN2 expression. This treatment also caused Smad3 linker region phosphorylation. Both outcomes were inhibited by dapagliflozin. We have identified a novel SGLT2 -ERK mediated promotion of TGF-β1/Smad3 signalling inducing a pro-fibrotic growth factor secretion. Our data evince support for substantial renoprotective benefits of SGLT2 inhibition in the diabetic kidney.
Article
Full-text available
Large cell carcinoma (LCC) is a rare and aggressive lung cancer subtype with poor prognosis and no targeted therapies. Tumor-associated fibroblasts (TAFs) derived from LCC tumors exhibit premature senescence, and coculture of pulmonary fibroblasts with LCC cell lines selectively induces fibroblast senescence, which in turn drives LCC cell growth and invasion. Here we identify MMP1 as overexpressed specifically in LCC cell lines, and we show that expression of MMP1 by LCC cells is necessary for induction of fibroblast senescence and consequent tumor promotion in both cell culture and mouse models. We also show that MMP1, in combination with TGF-β1, is sufficient to induce fibroblast senescence and consequent LCC promotion. Furthermore, we implicate PAR-1 and oxidative stress in MMP1/TGF-β1-induced TAF senescence. Our results establish an entirely new role for MMP1 in cancer, and support a novel therapeutic strategy in LCC based on targeting senescent TAFs.
Article
Full-text available
Background: Irradiation has emerged as a valid tool for nasopharyngeal carcinoma (NPC) in situ treatment; however, NPC derived from tissues treated with irradiation is a main cause cancer-related death. The purpose of this study is to uncover the underlying mechanism regarding tumor growth after irradiation and provided potential therapeutic strategy. Methods: Fibroblasts were extracted from fresh NPC tissue and normal nasopharyngeal mucosa. Immunohistochemistry was conducted to measure the expression of α-SMA and FAP. Cytokines were detected by protein array chip and identified by real-time PCR. CCK-8 assay was used to detect cell proliferation. Radiation-resistant (IRR) 5-8F cell line was established and colony assay was performed to evaluate tumor cell growth after irradiation. Signaling pathways were acquired via gene set enrichment analysis (GSEA). Comet assay and γ-H2AX foci assay were used to measure DNA damage level. Protein expression was detected by western blot assay. In vivo experiment was performed subcutaneously. Results: We found that radiation-resistant NPC tissues were constantly infiltrated with a greater number of cancer-associated fibroblasts (CAFs) compared to radiosensitive NPC tissues. Further research revealed that CAFs induced the formation of radioresistance and promoted NPC cell survival following irradiation via the IL-8/NF-κB pathway to reduce irradiation-induced DNA damage. Treatment with Tranilast, a CAF inhibitor, restricted the survival of CAF-induced NPC cells and attenuated the of radioresistance properties. Conclusions: Together, these data demonstrate that CAFs can promote the survival of irradiated NPC cells via the NF-κB pathway and induce radioresistance that can be interrupted by Tranilast, suggesting the potential value of Tranilast in sensitizing NPC cells to irradiation.
Article
Full-text available
BRAF and KRAS are two key oncogenes in the RAS/RAF/MEK/MAPK signaling pathway. Concomitant mutations in both KRAS and BRAF genes have been identified in non-small cell lung cancer (NSCLC). They lead to the proliferation, differentiation, and apoptosis of tumor cells by activating the RAS/RAF/MEK/ERK signaling pathway. To date, agents that target RAS/RAF/MEK/ERK signaling pathway have been investigated in NSCLC patients harboring BRAF mutations. BRAF and MEK inhibitors have gained approval for the treatment of patients with NSCLC. According to the reported findings, the combination of MEK inhibitors with chemotherapy, immune checkpoint inhibitors, epidermal growth factor receptor-tyrosine kinase inhibitors or BRAF inhibitors is highly significant for improving clinical efficacy and causing delay in the occurrence of drug resistance. This review summarized the existing experimental results and presented ongoing clinical studies as well. However, further researches need to be conducted to indicate how we can combine other drugs with MEK inhibitors to significantly increase therapeutic effects on patients with lung cancer.
Article
Full-text available
The mitogen-activated protein kinase (MAPK) pathway plays a vital role in cellular processes such as gene expression, cell proliferation, cell survival, and apoptosis. Also known as the RAS-RAF-MEK-ERK pathway, the MAPK pathway has been implicated in approximately one-third of all cancers. Mutations in RAS or RAF genes such as KRAS and BRAF are common, and these mutations typically promote malignancies by over-activating MEK and ERK downstream, which drives sustained cell proliferation and uninhibited cell growth. Development of drugs targeting this pathway has been a research area of great interest, especially drugs targeting the inhibition of MEK. In vitro and clinical studies have shown promise for certain MEK inhibitors (MEKi) , and MEKi have become the first treatment option for certain cancers. Despite promising results, not all patients have a response to MEKi, and mechanisms of resistance typically arise in patients who do have a positive initial response. This paper summarizes recent developments regarding MEKi, the mechanisms of adaptive resistance to MEKi, and the potential solutions to the issue of adaptive MEKi resistance.
Article
Full-text available
Pro-inflammatory cytokines like interleukin-1β (IL-1β) are upregulated during early responses to tissue damage and are expected to transiently compromise the mechanical microenvironment. Fibroblasts are key regulators of tissue mechanics in the lungs and other organs. However, the effects of IL-1β on fibroblast mechanics and functions remain unclear. Here we treated human pulmonary fibroblasts from control donors with IL-1β and used Atomic Force Microscopy to unveil that IL-1β significantly reduces the stiffness of fibroblasts concomitantly with a downregulation of filamentous actin (F-actin) and alpha-smooth muscle (α-SMA). Likewise, COL1A1 mRNA was reduced, whereas that of collagenases MMP1 and MMP2 were upregulated, favoring a reduction of type-I collagen. These mechanobiology changes were functionally associated with reduced proliferation and enhanced migration upon IL-1β stimulation, which could facilitate lung repair by drawing fibroblasts to sites of tissue damage. Our observations reveal that IL-1β may reduce local tissue rigidity by acting both intracellularly and extracellularly through the downregulation of fibroblast contractility and type I collagen deposition, respectively. These IL-1β-dependent mechanical effects may enhance lung repair further by locally increasing pulmonary tissue compliance to preserve normal lung distension and function. Moreover, our results support that IL-1β provides innate anti-fibrotic protection that may be relevant during the early stages of lung repair.
Article
Full-text available
The tumor microenvironment is a key modulator in cancer progression and has become a novel target in cancer therapy. An increase in hyaluronan (HA) accumulation and metabolism can be found in advancing tumor progression and are often associated with aggressive malignancy, drug resistance and poor prognosis. Wound-healing related myofibroblasts or activated cancer-associated fibroblasts (CAF) are assumed to be the major sources of HA. Both cell types are capable to synthesize new matrix components as well as reorganize the extracellular matrix. However, to which extent myofibroblasts and CAF perform these actions are still unclear. In this work, we investigated the matrix remodeling and HA production potential in normal human dermal fibroblasts (NHFB) and CAF in the absence and presence of transforming growth factor beta-1 (TGF-β1), with TGF-β1 being a major factor of regulating fibroblast differentiation. Three-dimensional (3D) collagen matrix was utilized to mimic the extracellular matrix of the tumor microenvironment. We found that CAF appeared to response insensitively towards TGF-β1 in terms of cell proliferation and matrix remodeling when compared to NHFB. In regards of HA production, we found that both cell types were capable to produce matrix bound HA, rather than a soluble counterpart, in response to TGF-β1. However, activated CAF demonstrated higher HA production when compared to myofibroblasts. The average molecular weight of produced HA was found in the range of 480 kDa for both cells. By analyzing gene expression of HA metabolizing enzymes, namely hyaluronan synthase (HAS1-3) and hyaluronidase (HYAL1-3) isoforms, we found expression of specific isoforms in dependence of TGF-β1 present in both cells. In addition, HAS2 and HYAL1 are highly expressed in CAF, which might contribute to a higher production and degradation of HA in CAF matrix. Overall, our results suggested a distinct behavior of NHFB and CAF in 3D collagen matrices in the presence of TGF-β1 in terms of matrix remodeling and HA production pointing to a specific impact on tumor modulation.
Article
Tissue inhibitor of metalloproteinase-1 (TIMP-1) is an important regulator of extracellular matrix turnover that has been traditionally regarded as a potential tumor suppressor owing to its inhibitory effects of matrix metalloproteinases. Intriguingly, this interpretation has been challenged by the consistent observation that increased expression of TIMP-1 is associated with poor prognosis in virtually all cancer types including lung cancer, supporting a tumor-promoting function. However, how TIMP-1 is dysregulated within the tumor microenvironment and how it drives tumor progression in lung cancer is poorly understood. We analyzed the expression of TIMP-1 and its cell surface receptor CD63 in two major lung cancer subtypes: lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC), and defined the tumor-promoting effects of their interaction. We found that TIMP-1 is aberrantly overexpressed in tumor-associated fibroblasts (TAFs) in ADC compared to SCC. Mechanistically, TIMP-1 overexpression was mediated by the selective hyperactivity of the pro-fibrotic TGF-β1/SMAD3 pathway in ADC-TAFs. Likewise, CD63 was upregulated in ADC compared to SCC cells. Genetic analyses revealed that TIMP-1 secreted by TGF-β1-activated ADC-TAFs is both necessary and sufficient to enhance growth and invasion of ADC cancer cells in culture, and that tumor cell expression of CD63 was required for these effects. Consistently, in vivo analyses revealed that ADC cells co-injected with fibroblasts with reduced SMAD3 or TIMP-1 expression into immunocompromised mice attenuated tumor aggressiveness compared to tumors bearing parental fibroblasts. We also found that high TIMP1 and CD63 mRNA levels combined define a stronger prognostic biomarker than TIMP1 alone. Our results identify an excessive stromal TIMP-1 within the tumor microenvironment selectively in lung ADC, and implicate it in a novel tumor-promoting TAF-carcinoma crosstalk, thereby pointing to TIMP-1/CD63 interaction as a novel therapeutic target in lung cancer.