SlideShare a Scribd company logo
1 of 9
Download to read offline
RESEARCH ARTICLE
Specific upregulation of RHOA and RAC1 in cancer-associated
fibroblasts found at primary tumor and lymph node metastatic
sites in breast cancer
Patricia Bortman Rozenchan1,2,3
& Fatima Solange Pasini1
& Rosimeire A. Roela1
&
Maria Lúcia Hirata Katayama1
& Fiorita Gonzáles Lopes Mundim4
&
Helena Brentani5
& Eduardo C. Lyra6
& Maria Mitzi Brentani1
Received: 2 April 2015 /Accepted: 28 June 2015
# International Society of Oncology and BioMarkers (ISOBM) 2015
Abstract The importance of tumor–stromal cell interactions
in breast tumor progression and invasion is well established.
Here, an evaluation of differential genomic profiles of
carcinoma-associated fibroblasts (CAFs) compared to fibro-
blasts derived from tissues adjacent to fibroadenomas (NAFs)
revealed altered focal adhesion pathways. These data were
validated through confocal assays. To verify the possible role
of fibroblasts in lymph node invasion, we constructed a tissue
microarray consisting of primary breast cancer samples and
corresponding lymph node metastasis and compared the ex-
pression of adhesion markers RhoA and Rac1 in fibroblasts
located at these different locations. Two distinct tissue micro-
arrays were constructed from the stromal component of 43
primary tumors and matched lymph node samples, respective-
ly. Fibroblasts were characterized for their expression of α-
smooth muscle actin (α-SMA) and vimentin. Moreover, we
verified the level of these proteins in the stromal compartment
from normal adjacent tissue and in non-compromised lymph
nodes. Our immunohistochemistry revealed that 59 % of fi-
broblasts associated with primary tumors and 41 % of the
respective metastatic lymph nodes (p=0.271) displayed posi-
tive staining for RhoA. In line with this, 57.1 % of fibroblasts
associated with primary tumors presented Rac1-positive stain-
ing, and the frequency of co-positivity within the lymph nodes
was 42.9 % (p=0.16). Expression of RhoA and Rac1 was
absent in fibroblasts of adjacent normal tissue and in compro-
mised lymph nodes. Based on our findings that no significant
changes were observed between primary and metastatic
lymph nodes, we suggest that fibroblasts are active partici-
pants in the invasion of cancer cells to lymph nodes and sup-
port the hypothesis that metastatic tumor cells continue to
depend on their microenvironment.
Keywords Breast cancer . Tumor microenvironment .
Carcinoma-associated fibroblasts . Rho GTPases . Metastasis
Introduction
Stromal cells, including fibroblasts, endothelial cells, and im-
mune cells, play a critical role in supporting breast cancer
(BC) growth, survival, and invasion. Fibroblasts represent
the major cell type of the stromal compartment and play an
important role in coordinating interactions between stromal
and tumor cells by modulating the composition and function
of the extracellular matrix. Carcinoma-associated fibroblasts
(CAFs) present different characteristics from those of fibro-
blasts found in normal breast tissue (NAFs). CAFs form a
heterogeneous population [1], express alpha-smooth muscle
actin (α-SMA) upon activation [2], and impact BC biological
behaviors [3].
* Patricia Bortman Rozenchan
pbrozenchan@hotmail.com
1
Radiology and Oncology Department, School of Medicine of São
Paulo University, Av. Dr. Arnaldo, 455, sala 4112, São
Paulo, SP CEP 01246-903, Brazil
2
Colsan—Blood Bank Beneficent Association, São Paulo, SP, Brazil
3
Gynecological Department, School of Medicine of São Paulo Federal
University, São Paulo, SP, Brazil
4
Pathology Department, Vale do Sapucaí University, Pouso
Alegre, MG, Brazil
5
Psychiatric Department, School of Medicine of São Paulo University
(USP), Rua Ovideo Pires de Campos S/N, São Paulo, SP, Brazil
6
Brazilian Institute of Cancer Control, Av. Alcântara Machado 2576,
São Paulo, SP CEP 03102-002, Brazil
Tumor Biol.
DOI 10.1007/s13277-015-3727-1
The importance of interactions between tumor cells and the
surrounding stromal cells [4] has been well established. In
particular, the migratory/invasive behavior of tumor cells has
been reported to be influenced by bidirectional signals be-
tween cancer cells and tumor-associated stroma fibroblasts
and affects human breast cancer cell adhesion, migration
speed, and direction [5, 6]. During cell invasion, fibroblasts
use contractile force and proteolytic activity to reorganize col-
lagen into linear fibers to generate tracks for migration of
cancer cells [7]. Notably, family members of the Rho-small
guanosine triphosphatases (GTPases), RhoA, RAC1, and
RAC2 binding proteins, have been implicated in CAF-
mediated remodeling of the tumor microenvironment in a
manner that enhances cancer cell invasion [8]. Indeed, RhoA,
Rac1, and Rac2 induce stress fiber formation when
overexpressed in fibroblasts [9].
In the present study, we analyzed genes that were differen-
tially expressed in primary breast CAFs compared to fibro-
blasts that originated from adjacent tissue of benign breast
diseases. This analysis revealed numerous differences in
genes involved in major functional pathways, including focal
adhesion and regulation of actin cytoskeleton and tight junc-
tions. Specifically, the GTPase family members RhoA,
RAC1, and RAC2, as well as collagens and integrins, which
are known to mediate migration and invasiveness, were
shown to be differentially regulated in the context of CAFs.
To date, increased expression of these proteins, particularly
within the epithelial compartment, has been shown to be im-
portant for cellular motility, loss of adhesion, invasion, and
metastasis [10–13].
Nodal status represents one of the most powerful inde-
pendent prognostic indicators of breast carcinoma [14].
Histologically, the presence of fibroblasts in the BC micro-
environment of metastatic lymph nodes further reinforces
the role that CAFs play in tumor growth and dissemination
[15]. Le Bedis et al. [16] suggested that lymph node stroma
plays an active part in the process of lymph node metasta-
sis by creating a dynamic microenvironment that mimics
the environmental conditions present at the primary tumor
site. Previous studies by Garcia et al. [17] reported a sim-
ilar expression of metalloproteases within primary tumors
and the respective lymph nodes. Moreover, tumor–stroma
cross-talk seems to influence the metastatic lymph node
microenvironment, affecting proliferation and migration
of cancer cells [16, 18].
Because the genomic profiles of fibroblasts BC primary
and lymph node metastasis have been reported to be similar
[19], we hypothesized that similar levels of RhoA and Rac1
expression in the tumor microenvironment at the primary site
and at the lymph nodes may represent an advantage to tumor
cell behavior. In this study, we determined whether the expres-
sion of RhoA and Rac1 in stromal fibroblasts of primary tu-
mors was similar to that found in lymph node fibroblasts.
Using a tissue microarray (TMA) consisting of primary BC
samples and the corresponding lymph node metastasis, we
compared the expression level of these markers in fibroblasts
residing at the two locations, as well as in adjacent normal
tissue of primary tumors and in non-compromised lymph
nodes.
Materials and methods
Primary cell culture tissue samples
Malignant and benign breast tissue specimens were obtained
from consenting patients undergoing surgery for breast dis-
ease. Carcinoma samples were obtained from four patients
clinically staged as IIa, and benign samples were obtained
from four patients diagnosed as fibroadenoma. All tissue do-
nors were patients at Instituto Brasileiro de Controle do Cân-
cer, São Paulo, Brazil, a reference center for cancer treatment.
This study was approved by the Ethical Institutional Commit-
tee, and written explicit informed consent was obtained from
all participants. Invasive breast cancer was confirmed
histopathologically.
Primary cell culture
Fibroblasts were obtained from normal adjacent tissue sam-
ples from patients with benign breast diseases (NAF) or
with primary invasive breast cancer tumors (CAF). H&E-
stained, frozen histological sections were prepared from
each tissue sample to confirm benignity or malignancy.
After adipose tissue removal, tissue was minced in
phosphate-buffered saline (PBS), washed twice in PBS
(Na2HPO4 10 mM, NaCl 1.37 mM, KCl 27 mM, KH2PO4
2 mM, Thermo Fisher Scientific Inc., MA, USA) and in
culture medium, and then chopped into small 1- to 4-mm3
pieces under sterile conditions. A total of 15–30 fragments
were transferred to a T25 culture flask (Thermo Fisher Sci-
entific Inc.) and covered with Dulbecco’s modified Eagle’s
medium (DMEM, Thermo Fisher Scientific Inc.) supple-
mented with 20 % FBS (Thermo Fisher Scientific Inc.),
100 μg/mL ampicillin, 100 μg/mL streptomycin, and
2.5 μg/mL fungizone and maintained at 37 °C in a humid-
ified atmosphere containing 5 % CO2. Outgrowth of cells
was recorded after 10 to 20 days, and the medium was
renewed once or twice a week thereafter. After sufficient
outgrowth, the tissue fragments were removed and the cells
were passaged by mild trypsinization with trypsin 0.5 %
(Thermo Fisher Scientific Inc.).
Early passages (passage 3) of all fibroblasts were
subjected to immunocytochemical evaluation. The prolif-
eration rates and description of patients were described
before [20].
Tumor Biol.
cDNA microarray assembly, hybridization, and analysis
Total RNA from four NAFs and four CAFs were extracted by
TRIzol reagent (Thermo Fisher Scientific Inc.) and purified
with RNeasy minicolumns and reagents (Qiagen, Hilden, Ger-
many). RNA microarray analysis was performed with 10 μg
of biotin-labelled cRNA target prepared by a linear amplifica-
tion method from a pool of samples. The poly (A)+
RNA
(mRNA) subpopulation within the total RNA population was
primed for reverse transcription by a DNA oligonucleotide
containing the T7 RNA polymerase promoter 5′ to a d (T)24
sequence. After second-strand cDNA synthesis, the cDNA
served as the template for an in vitro transcription (IVT) reac-
tion to produce the target cRNA. This cRNA was hybridized
using CodeLinkTM
Human Whole Genome 55K Bioarray (GE
Healthcare, Buckinghamshire, UK), and the hybridization sig-
nals were normalized using the CodeLinkTM
System Software
Analysis (GE Healthcare) and subjected to a t test with 10,000
permutations. Genes with twofold differential expression
levels in the CAF versus NAF comparison were considered
differentially expressed. The gene ontology (GO) analysis was
performed using the GO Tree Machine tool (GOTM) [21],
which identifies hyper-represented categories in our gene lists
as well as the Kyoto Encyclopedia of Genes and Genomes
(KEGG) pathways. For both GO and KEGG, we used a
hyper-geometric distribution with a p value≤0.05 [Onto-tolls].
Fluorescence microscopy
Cells were cultured on glass coverslips in 24-well plates. After
24 h, cells were fixed with 3.5 % paraformaldehyde in PBS at
room temperature, permeabilized for 5 min with 0.2 % Triton
X-100 in PBS, and blocked with 5 % bovine serum albumin
(BSA, Thermo Fisher Scientific Inc.) in PBS for 1 h. The
primary chicken polyoclonal RhoA antibody (ab23687,
Abcam Antibodies, Cambridge, UK) and mouse monoclonal
Rac1 antibody (ab33186, Abcam Antibodies) were incubated
overnight at 4 °C. After extensive washes and secondary an-
tibody staining (goat anti-mouse IgG, anti-chicken IgG, and
anti-rabbit IgG; Sigma-Aldrich, MO, USA), nuclei were
stained with 4′-6′-diamidino-2-phenylindole (DAPI, Sigma-
Aldrich). Cells were analyzed using a Zeiss 510 META con-
focal laser scanning microscope using a 488-nm argon and a
543-nm HeNe laser. Images were acquired using a Plan
NeoFluoar 40×/1 lens (Microimaging Inc., Lane Cove, New
Zealand).
Immunohistochemistry tissue samples
This study included 43 breast cancer samples that involved
axillary nodes from patients undergoing breast surgery at Hos-
pital Samuel Libânio, in Pouso Alegre, MG, Brazil from 1997
to 2005. This study was approved by the Institutional Ethics
Committee of Hospital Samuel Libânio (no. 1119/09). The
median age of patients was 58 years (range 38–88 years). All
patients were diagnosed with invasive ductal carcinomas at
clinical stages II and III in 37.2 or 62.8 % of cases, respectively
(two unknown). Statuses of the estrogen receptor (ER), pro-
gesterone receptor (PR), HER-2, and histological grade (HG)
are listed in Table 1. HER-2 was assessed by the HercepTestTM
(Dako, Glostrup, Denmark A/S) system. Membrane-based
staining cases with scores 0–1 or chromogenic in situ hybrid-
ization (CISH) negative were categorized as negative, while
cases with score ≥3 were considered as positive. The cases
with an immunohistochemistry score of 2 were further con-
firmed by CISH analysis (Ventana Medical Systems, Inc., a
member of the Roche Group, Tucson, AZ) [22].
Additionally, we included 10 female BC cases (aged be-
tween 39–84 years) in spite of analyzing the stromal compart-
ment of adjacent normal tissue of primary tumors and not
compromised lymph nodes. All samples were subjected to a
pathology analysis, where tumor margins were considered to
assure consistency within the tissues. The Pathology
Reporting of Breast Disease was used as guidelines [23].
Construction of TMA
Two distinct TMAs were constructed from the stromal com-
ponent of primary tumors and lymph node samples,
Table 1 Clinical and pathological parameters of 43 breast cancer
patients
Characteristic No. of patients (%)
Age median (range) 58 (38–88)
Clinical stage
I 1 (2.3)
II 16 (37.2)
III 26 (60.5)
Histologic grade
Grade I 9 (20.9)
Grade II 15 (34.9)
Grade III 19 (44.2)
ER—primary tumor
Negative 18 (41.5)
Positive 24 (55.8)
Missing 1 (2.3)
PR—primary tumor
Negative 17 (39.5)
Positive 21 (48.8)
Missing 5 (11.6)
HER-2
Negative 33 (76.7)
Positive 9 (20.9)
Missing 1 (2.3)
Tumor Biol.
respectively. Representative areas of each component were
surrounded by a marking pen at the donor blocks and collect-
ed using the Manual Tissue Arrayer I (Beecher Instruments
Inc., Sun Prairie, USA). Samples were orderly arranged in a
grid, and the first core was represented by a fragment or nor-
mal liver used as a reference in both TMAs. The first TMA
was built with samples of stromal component of the tumors in
order to enable the assessment of fibroblast cells within the
desmoplastic contingent of the carcinomas.
The second TMA consisted of 43 majorly compromised
lymph node samples from cases with axillary lymph node
metastasis. Because the width of the metastatic element at
the lymph nodes was frequently limited to 0.5 to 1.0 cm, it
was seldom possible to separate distinct areas of stromal and
epithelial elements. The chosen areas at the donor block in-
volved stromal and epithelial elements, which were further
evaluated separately.
From each of the two TMAs, 3-μm-thick slices were ob-
tained and collected on slides with special adhesives
(Instrumedics Inc., NJ, USA). Two sets of triplets, separated
by a gap of 40 cuts, were submitted for immunohistochemical
analyses in order to represent two levels of the same sample.
Immunohistochemistry
The immunohistochemical reactions were performed using
the complex streptavidin-biotin peroxidase (StreptABC, Dako
Corporation, Glostrup, Denmark). After deparaffinization of
tissue sections, antigen retrieval was performed using a pres-
sure cooker in citrate buffer pH 6.0, followed by blocking
endogenous peroxidase with hydrogen peroxide solution
(3 %). The sections were incubated with primary antibodies,
Rac1 (1:1,000; ab33186, Abcam) and RhoA (1:175; ab23687,
Abcam), vimentin (1:2,000; clone v9, Dako Corporation), and
α-SMA (clone: HHF-35; Cell Marque, CA, USA). After in-
cubation with primary antibody and primary blocking, a poly-
mer–peroxidase (Novolink, Leica, Wetzlar, Germany) ampli-
fication step was performed. Antigen detection was carried
out in a solution containing 3,3-diaminobenzidine (Sigma-
Aldrich) and 6 % H2O2. Counterstaining was performed with
Harris hematoxylin (Merck, NJ, USA). Carcinoma ductal in-
vasive of breast was used as positive control for RhoA, and
large intestine adenocarcimona was the positive control used
for Rac1. Negative controls were performed removing first
antibodies.
Evaluation of immunohistochemical essays
All reactions were assessed by two independent blinded ob-
servers. Disparities between the two pathologists were
reevaluated by consensus. Results from the epithelial compo-
nent and stromal component were reported separately either to
carcinomas or to lymph node samples.
The presence of RhoA and Rac1 staining was assessed in
the stromal population and separately in primary tumors or in
lymph node metastases. In relation to the immunohistochem-
ical result evaluation, RhoA was scored according to the
Allred system [24]. Samples with a score above 4 were con-
sidered positive. For Rac1 evaluation, the percentage of pos-
itive cells was assessed and classified into three groups: (1)
0 %, (2) 1−33 %, and (3) ≥34 %. Samples with greater than
10 % of cells that were positive for α-SMA and vimentin were
classified as positive. For all cases, cells presented in 10 fields
at a magnification of ×400 were counted.
Statistical methods
Correlations between categorical antigen expression and other
clinicopathological parameters were studied with the Fisher’s
exact test or chi-square test, where appropriate. Spearman’s
rank correlation coefficient was calculated to assess the rela-
tionships involving categorical antigen expression. All statis-
tical tests were two-sided, with significance defined as
p<0.05. Analyses were performed using the software SPSS
version 10.0 for Windows (SPSS Inc., IL, USA).
Results
First, we evaluated the gene expression profile of stromal fi-
broblasts derived from four benign (NAF) and four malignant
(CAF) breast tissues using the CodeLinkTM
Human Whole
Genome 55K Bioarray. Using a twofold cutoff, we found 1,
111 genes differentially expressed in CAFs compared to
NAFs. To further examine the biological functions of these
genes, an analysis of the KEGG database revealed differential
expression of several pathways, including Boxidative phos-
phorylation,^ “focal adhesion,” BMAPK signaling pathway,^
Bleukocyte transendothelial migration,^ Bregulation of actin
cytoskeleton,^ and Btight junctions^ (Fig. 1). Interestingly,
all of these pathways are involved in important processes like
cell/cell communication, migration, and invasiveness. Focal
adhesion was ranked the second top significant pathway, and
in microarray analysis, we found that both RhoA and Rac1
were upregulated in CAFs compared to NAFs.
Using confocal assays, we confirmed that protein levels of
RhoA and Rac1 were elevated in carcinoma-associated fibro-
blasts (Fig. 2). Strong RhoA staining was localized to the
cytoplasm, while Rac1 staining was associated with the plas-
ma membrane.
To further examine correlations between altered genes
found in the microarray data with patterns of protein expres-
sion, we analyzed the expression of RhoA and Rac1 in the
stromal component of 43 samples of primary breast carcinoma
and metastatic ipsilateral axillary lymph nodes by immunohis-
tochemistry. In a few metastatic lesions cases, it was not
Tumor Biol.
Fig. 1 Distribution of
differentially expressed genes in
CAFs. Representative pathways
were identified for genes
differentially expressed (black
bars) between CAFs and NAFs in
our cDNA microarray platform
(gray bars). The gene ontology
(GO) analysis was performed
using the GO Tree Machine tool
(GOTM), which identifies hyper-
represented categories in our gene
lists as well as the Kyoto
Encyclopedia of Genes and
Genomes (KEGG) pathways. For
both GO and KEGG, we used a
hyper-geometric distribution with
a p value≤0.05 [Onto-tolls]
Fig. 2 CAF-specific staining of
RhoA and Rac1. CAF (a, c) and
NAF (b, d) confocal
photomicrographs of typical
fields are shown. Merged images
of a cytoplasmatic staining of
RhoA (a, b) and cytoplasmatic
membrane staining of Rac1 (c, d)
validated the gene expression
analysis; Rac1 and RhoA were
upregulated in CAFs
Tumor Biol.
possible to count the preferred number of cells. These proteins
were also analyzed in fibroblasts of adjacent normal tissue of
primary tumors and in non-compromised lymph nodes of 10
breast cancer cases.
To distinguish fibroblasts (CAFs) from tumoral cells, we
evaluated two fibroblast cell markers: vimentin and α-SMA.
Vimentin staining was observed in 37/43 (86.0 %) of fibro-
blasts of primary tumors and in 35/41 (85.4 %) of lymph nodal
fibroblasts. α-SMA staining was observed in 51.2 % of pri-
mary lesions and in 41 % of nodal metastatic lesions (Fig. 3).
Positive RhoA staining in stromal cells was seen in 36/43
cases of primary tumors (59 %) and in 25/35 (41 %) of respec-
tive metastatic lymph nodes (p=0.271). Similarly, Rac1 stain-
ing in stromal cells was observed in 28/39 cases of primary
tumors (57.1 %), whereas the frequency of Rac1 staining in
stromal cells found in the lymph nodes was 21/38 (42.9 %)
(Table 2). In summary, the status of RhoA and Rac1 was
similar between primaries and lymph nodes (Fig. 4), and all
markers were detected in the cytoplasm. Importantly, RhoA or
Rac1 expression was not detected in fibroblasts of adjacent
normal tissue or in non-compromised lymph nodes (Fig. 5),
suggesting that the presence of RhoA or Rac1 in CAFs may
help facilitate tumorigenesis.
Discussion
A large amount of data have shown that the microenvironment
is important to breast carcinoma epithelial cells [25–29]. Ad-
ditionally, we have shown that reciprocal changes in gene
expression profiles of important cellular function pathways
occur when breast epithelial cells were co-cultured with
primary fibroblasts, reinforcing the idea that interactions be-
tween these two cell types impact cell signaling and behavior
[20]. In line with these works, we compared gene expression
data generated in CAFs and NAFs to show that a large number
of genes present in the focal adhesion pathways, including the
GTPAses RhoA and Rac1, were differentially expressed in
CAFs.
Cell motility and invasiveness require cytoskeleton reorga-
nization, which involves formation of the filopodia/
lamellipodia and changes in focal adhesion complexes. Rho
proteins are involved in stress fiber formation and focal adhe-
sion, while Rac proteins stimulate lamellipodia and
membrane-ruffle formation [30–33]. Increasing evidence sug-
gests that the RhoA and Rac1 proteins play an important role
in cell migration, loss of adhesion, invasion, and metastasis in
tumors [10, 34–37]. Building on these studies, we used im-
munohistochemistry to investigate whether the RhoA and
Rac1 expression patterns were similar in fibroblasts of prima-
ry BC tumors and matched lymph node metastases.
Fig. 3 CAF-specific α-SMA and
vimentin staining at primary at
metastatic tumor sites. α-SMA (a,
b) and vimentin (c, d) staining in
CAFs in the primary tumors (a, c)
and in their counterpart lymph
node metastasis (b, d). Original
magnification ×400
Table 2 Correlation among the proportion of biological marker
expression in stromal tissue between the primary tumors and lymph
node metastasis of 43 breast cancer patients
Variable Primary tumor Lymph node p
RhoA
Negative 7 (41.2) 10 (58.8) 0.27
Positive 36 (59.0) 25 (41.0)
Rac1
Negative 11 (39.3) 17 (60.7) 0.16
Positive 28 (57.1) 21 (42.9)
Stroma tissue: only fibroblast cells were analyzed; Values of p (two-sided)
less than 0.05 were considered significant
Tumor Biol.
Early studies noted that RhoA overexpression result-
ed in mouse fibroblast transformation [38] and stress
fiber formation [39]. It was previously shown that high-
ly metastatic mesenchymal sarcoma cells primarily use
an ameboid mode of cell invasion that depends on the
activity of the Rho family of GTPases [40]. Some au-
thors have described the role of Rho GTPases in fibro-
blasts [41–43]. Verghese et al. [41] reported that cyto-
skeletal regulation by Rho GTPases in breast fibroblasts
enhanced migration and invasion in consequence of mir-
26b dysregulation.
In agreement with Halon et al. [10], we show that a high
proportion of cells in the BC tumor–stromal compartment
were positive for RhoA and Rac1. In addition, we have shown
that activated fibroblasts, as identified by α-SMA, were pres-
ent in the majority of metastatic lymph node cases, whereas
non-involved lymph nodes were devoid of myofibroblasts.
The epithelial-to-mesenchymal transition (EMT) repre-
sents one possible mechanism by which a cancer cell metas-
tasizes and may facilitate the re-localization of fibroblasts to
the lymph node. Past work has emphasized RhoA as a medi-
ator of this process via integrin β1/TGF β activation [44].
Fig. 4 RhoA and Rac1 staining
in primaries and in lymph node
metastasis. Representative cases
depicting RhoA and Rac1 (a, c) in
CAFs in the primary tumors and
in the respective lymph node
metastasis (b, d). Original
magnification ×400
Fig. 5 Lack of RhoA and Rac1
staining in fibroblasts of adjacent
normal tissue and in non-
compromised lymph nodes.
Negative expression of RhoA and
Rac1 in fibroblasts of adjacent
normal tissue (a, c) and non-
compromised lymph nodes (b, d).
Original magnification ×400
Tumor Biol.
Moreover, EMT is a recognized source of CAFs, as it pro-
duces myofibroblast cells with enhanced migratory capacity,
invasiveness, and increased expression of ECM proteins [45].
In line with these data, we found that 41 % of fibroblasts of
lymph nodes origin were positive for RhoA and α-SMA.
CAFs may also arrive at lymph nodes in co-migration with
tumor cells, referred to as the collective pathway of invasion
[46]. Reorganization of collagen fibers by mammary fibro-
blasts can create avenues for invasion [5], and the Rho-
family of GTP-binding proteins may regulate fibroblast-
mediated collagen reorganization [8]. Therefore, RhoA and
Rac1 may be involved in both pathways.
Our study brings some clarification regarding the role of
the microenvironment in lymph node metastasis in BC. How-
ever, we acknowledge that our study is limited by the number
of samples used in immunohistochemical assays, as well as by
the presence of non-distinct histological types, as different
subtypes represent different fibroblasts populations [47].
In conclusion, we demonstrate positive expression of
RhoA and Rac1 in CAFs in paired primary breast cancer
and in the respective lymph node metastasis, suggesting that
a similar microenvironment may be present at both sites. Im-
portantly, as we were unable to detect positive expression of
these proteins in fibroblasts of normal adjacent tissue or in
non-committed lymph nodes, our findings may highlight the
stroma as an active participant in the metastatic process and
suggest that metastatic tumor cells may continue to be depen-
dent on their supportive microenvironment.
Acknowledgments The authors are grateful to Ana Lúcia Garippo for
her technical assistance in confocal microscopy. This research was sup-
ported by Fundação de Amparo à Pesquisa no Estado de São Paulo
(FAPESP) 01/13513-1, 05/51593-5, 04/04607-8, 05/60333-7, 2014/
03090-3 and 09/10088-7 and Conselho Nacional de Desenvolvimento
Científico e Tecnológico (CNPq).
Conflicts of interest None
Ethical approval All procedures performed in studies involving hu-
man participants were in accordance with the ethical standards of the
institutional and/or national research committee and with the 1964 Hel-
sinki declaration and its later amendments or comparable ethical
standards.
References
1. Sugimoto H, Mundel TM, Kieran MW, Kalluri R. Identification of
fibroblast heterogeneity in the tumor microenvironment. Cancer
Biol Ther. 2006;5(12):1640–6.
2. Xouri G, Christian S. Origin and function of tumor stroma fibro-
blasts. Semin Cell Dev Biol. 2010;21(1):40–6.
3. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer.
2006;6(5):392–401.
4. Polyak K, Kalluri R. The role of the microenvironment in mamma-
ry gland development and cancer. Cold Spring Harb Perspect Biol.
2010;2(11):a003244.
5. Dang TT, Prechtl AM, Pearson GW. Breast cancer subtype-specific
interactions with the microenvironment dictate mechanisms of in-
vasion. Cancer Res. 2011;71(21):6857–66.
6. Angelucci C, Maulucci G, Lama G, Proietti G, Colabianchi A, Papi
M, et al. Epithelial-stromal interactions in human breast cancer:
effects on adhesion, plasma membrane fluidity and migration speed
and directness. PLoS One. 2012;7(12):e50804. doi:10.1371/
journal.pone.0050804.
7. Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW,
Provenzano PP, et al. Aligned collagen is a prognostic signature for
survival in human breast carcinoma. Am J Pathol. 2011;178(3):
1221–32.
8. Rhee S, Grinnell F. Fibroblast mechanics in 3D collagen matrices.
Adv Drug Deliv Rev. 2007;59(13):1299–305.
9. Wheeler AP, Ridley AJ. Why three Rho proteins? RhoA, RhoB,
RhoC, and cell motility. Exp Cell Res. 2004;301(1):43–9.
10. Halon A, Donizy P, Surowiak P, Matkowski R. ERM/Rho protein
expression in ductal breast cancer: a 15 year follow-up. Cell Oncol.
2013;36(3):181–90.
11. Wu YJ, Tang Y, Li ZF, Li Z, Zhao Y, Wu ZJ, Su Q. Expression and
significance of Rac1, Pak1 and Rock1 in gastric carcinoma. Asia
Pac J Clin Oncol. 2013; doi: 10.1111/ajco.12052
12. Ridley AJ. Rho proteins and cancer. Breast Cancer Res Treat.
2004;84(1):13–9.
13. Sahai E, Marshall CJ. Differing modes of tumour cell invasion have
distinct requirements for Rho/ROCK signalling and extracellular
proteolysis. Nat Cell Biol. 2003;5(8):711–9.
14. Yenidunya S, Bayrak R, Haltas H. Predictive value of pathological
and immunohistochemical parameters for axillary lymph node me-
tastasis in breast carcinoma. Diagn Pathol. 2011;6:18. doi:10.1186/
1746-1596-6-18.
15. Cao Y, Paner GP, Rajan PB. Sentinel node status and tumor char-
acteristics: a study of 234 invasive breast carcinomas. Arch Pathol
Lab Med. 2005;129(1):82–4.
16. LeBedis C, Chen K, Fallavollita L, Boutros T, Brodt P. Peripheral
lymph node stromal cells can promote growth and tumorigenicity
of breast carcinoma cells through the release of IGF-I and EGF. Int J
Cancer. 2002;100(1):2–8.
17. García MF, González-Reyes S, González LO, Junquera S, Berdize
N, Del Casar JM, et al. Comparative study of the expression of
metalloproteases and their inhibitors in different localizations with-
in primary tumours and in metastatic lymph nodes of breast cancer.
Int J Exp Pathol. 2010;91(4):324–34.
18. Montel V, Mose ES, Tarin D. Tumor-stromal interactions recipro-
cally modulate gene expression patterns during carcinogenesis and
metastasis. Int J Cancer. 2006;119(2):251–63.
19. Del Valle PR, Milani C, Brentani MM, Katayama ML, de Lyra EC,
Carraro DM, et al. Transcriptional profile of fibroblasts obtained
from the primary site, lymph node and bone marrow of breast can-
cer patients. Genet Mol Biol. 2014;37(3):480–9.
20. Rozenchan PB, Carraro DM, Brentani H, et al. Reciprocal changes
in gene expression profiles of cocultured breast epithelial cells and
primary fibroblasts. Int J Cancer. 2009;125(12):2767–77.
21. Onto-Tools data base [http://vortex.cs.wayne.edu/projects.htm].
22. Wolff AC, Hammond ME, Hicks DG, et al. Recommendations for
human epidermal growth factor receptor 2 testing in breast cancer:
American Society of Clinical Oncology/College of American
Pathologists clinical practice guideline update. Arch Pathol Lab
Med. 2014;138:241–56.
23. Pathology Reporting of Breast Disease. A Joint Document
Incorporating the Third Edition of the NHS Breast Screening
Programme’s Guidelines for Pathology Reporting in Breast
Cancer Screening and the Second Edition of The Royal College
of Pathologists’ Minimum Dataset for Breast Cancer
Histopathology. NHSBSP Publication No 58. 2005.
Tumor Biol.
24. Allred DC, Harvey JM, Berardo M, et al. Prognostic and predictive
factors in breast cancer by immunohistochemical analysis. Mod
Pathol. 1998;11:155–68.
25. Casbas-Hernandez P, Fleming JM, Troester MA. Gene expression
analysis of in vitro cocultures to study interactions between breast
epithelium and stroma. J Biomed Biotechnol. 2011; 520987. doi:
10.1155/2011/520987
26. Hawsawi NM, Ghebeh H, Hendrayani SF, et al. Breast carcinoma-
associated fibroblasts and their counterparts display neoplastic-
specific changes. Cancer Res. 2008;68(8):2717–25.
27. Casey T, Bond J, Tighe S, et al. Molecular signatures suggest a
major role for stromal cells in development of invasive breast can-
cer. Breast Cancer Res Treat. 2009;114:47–62.
28. Singer CF, Gschwantler-Kaulich D, Fink-Retter A, et al.
Differential gene expression profile in breast cancer-derived stro-
mal fibroblasts. Breast Cancer Res Treat. 2008;110(2):273–81.
29. Buess M, Nuyten DS, Hastie T, Nielsen T, Pesich R, Brown PO.
Characterization of heterotypic interaction effects in vitro to
deconvolute global gene expression profiles in cancer. Genome
Biol. 2007;8(9):R191.
30. Dummler B, Ohshiro K, Kumar R, Field J. Pak protein kinases and
their role in cancer. Cancer Metastasis Rev. 2009;28(1–2):51–63.
31. Arias-Romero LE, Villamar-Cruz O, Pacheco A, Kosoff R, Huang
M, Muthuswamy SK, et al. A Rac-Pak signaling pathway is essen-
tial for ErbB2-mediated transformation of human breast epithelial
cancer cells. Oncogene. 2010;29(43):5839–49.
32. Rider L, Oladimeji P, Diakonova M. PAK1 regulates breast cancer
cell invasion through secretion of matrix metalloproteinases in re-
sponse to prolactin and three-dimensional collagen IV. Mol
Endocrinol. 2013;27(7):1048–64.
33. Shin YJ, Kim EH, Roy A, Kim JH. Evidence for a novel mecha-
nism of the PAK1 interaction with the Rho-GTPases Cdc42 and
Rac. PLoS One. 2013;8(8):e71495. doi:10.1371/journal.pone.
0071495.
34. Chan CH, Lee SW, Li CF, Wang J, Yang WL, Wu CY, et al.
Deciphering the transcriptional complex critical for RhoA gene
expression and cancer metastasis. Nat Cell Biol. 2010;12(5):457–
67.
35. Fritz G, Brachetti C, Bahlmann F, Schmidt M, Kaina B. Rho
GTPases in human breast tumours: expression and mutation anal-
yses and correlation with clinical parameters. Br J Cancer.
2002;87(6):635–44.
36. Chang YW, Marlin JW, Chance TW, et al. RhoA mediates
cyclooxygenase-2 signaling to disrupt the formation of adherens
junctions and increase cell motility. Cancer Res. 2006;66(24):
11700–8.
37. Spiering D, Hodgson L. Dynamics of the Rho-family small
GTPases in actin regulation and motility. Cell Adhes Migr.
2011;5(2):170–80.
38. Khosravi-Far R, Solski PA, Clark GJ, Kinch MS, Der CJ.
Activation of Rac1, RhoA, and mitogen-activated protein kinases
is required for Ras transformation. Mol Cell Biol. 1995;15(11):
6443–53.
39. Heasman SJ, Ridley AJ. Mammalian Rho GTPases: new insights
into their functions from in vivo studies. Nat Rev Mol Cell Biol.
2008;9(9):690–701.
40. Rösel D, Brábek J, Tolde O, Mierke CT, Zitterbart DP, Raupach C,
et al. Up-regulation of Rho/ROCK signaling in sarcoma cells drives
invasion and increased generation of protrusive forces. Mol Cancer
Res. 2008;6(9):1410–20.
41. Verghese ET, Drury R, Green CA, Holliday DL, Lu X, Nash C,
et al. MiR-26b is down-regulated in carcinoma-associated fibro-
blasts from ER-positive breast cancers leading to enhanced cell
migration and invasion. J Pathol. 2013;231(3):388–99.
42. Berenjeno IM, Bustelo XR. Identification of the Rock-dependent
transcriptome in rodent fibroblasts. Clin Transl Oncol. 2008;10(11):
726–38.
43. Sells MA, Pfaff A, Chernoff J. Temporal and spatial distribution of
activated Pak1 in fibroblasts. J Cell Biol. 2000;151(7):1449–58.
44. Tse JC, Kalluri R. Mechanisms of metastasis: epithelial-to-
mesenchymal transition and contribution of tumor microenviron-
ment. J Cell Biochem. 2007;101(4):816–29.
45. Aboussekhra A. Role of cancer-associated fibroblasts in breast can-
cer development and prognosis. Int J Dev Biol. 2011;55(7–9):841–
9.
46. Gaggioli C, Hooper S, Hidalgo-Carcedo C, et al. Fibroblast-led
collective invasion of carcinoma cells with differing roles for
RhoGTPases in leading and following cells. Nat Cell Biol.
2007;9(12):1392–400.
47. Tchou J, Kossenkov AV, Chang L, Satija C, Herlyn M, Showe LC,
et al. Human breast cancer associated fibroblasts exhibit subtype
specific gene expression profiles. BMC Med Genomics. 2012;5:39.
Tumor Biol.

More Related Content

What's hot

Pancreatic cancer
Pancreatic cancerPancreatic cancer
Pancreatic cancer
Springer
 
Compartment specific micro rna expression profiles (poster) poster
Compartment specific micro rna expression profiles (poster) posterCompartment specific micro rna expression profiles (poster) poster
Compartment specific micro rna expression profiles (poster) poster
Jackie Lau
 
3478-43782-3-PB
3478-43782-3-PB3478-43782-3-PB
3478-43782-3-PB
东妮 郑
 
2013 CD8 Lymphocytes and apoptosis in MBC-1
2013 CD8  Lymphocytes and apoptosis in MBC-12013 CD8  Lymphocytes and apoptosis in MBC-1
2013 CD8 Lymphocytes and apoptosis in MBC-1
IKA Nurlaila
 
Immuotherapy 2
Immuotherapy 2Immuotherapy 2
Immuotherapy 2
drmcbansal
 
EmergingUnderstandingofMultiscaleTumorHeterogeneityDecember2014
EmergingUnderstandingofMultiscaleTumorHeterogeneityDecember2014EmergingUnderstandingofMultiscaleTumorHeterogeneityDecember2014
EmergingUnderstandingofMultiscaleTumorHeterogeneityDecember2014
Rafael Casiano
 
Kurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLSKurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLS
Swati Jalgaonkar
 
biochem of cancer modified dialysis treatment
biochem of cancer modified dialysis treatmentbiochem of cancer modified dialysis treatment
biochem of cancer modified dialysis treatment
Thomas Brinkman
 
Breast Cancer Stem Cells
Breast Cancer Stem CellsBreast Cancer Stem Cells
Breast Cancer Stem Cells
Amber Rigdon
 
Molecular mechanism of tumor invasiveness
Molecular mechanism of tumor invasivenessMolecular mechanism of tumor invasiveness
Molecular mechanism of tumor invasiveness
tj1985
 
Disseminated Breast Cancer Cells Acquire a Highly Malignant and Aggressive Me...
Disseminated Breast Cancer Cells Acquire a Highly Malignant and Aggressive Me...Disseminated Breast Cancer Cells Acquire a Highly Malignant and Aggressive Me...
Disseminated Breast Cancer Cells Acquire a Highly Malignant and Aggressive Me...
Carolyn Marsden
 
JTM-Functional characterization of human Cd33+ And Cd11b+ myeloid-derived sup...
JTM-Functional characterization of human Cd33+ And Cd11b+ myeloid-derived sup...JTM-Functional characterization of human Cd33+ And Cd11b+ myeloid-derived sup...
JTM-Functional characterization of human Cd33+ And Cd11b+ myeloid-derived sup...
Karolina Megiel
 

What's hot (20)

Epithelial and mesenchymal transition in invasion and metastasis
Epithelial and mesenchymal transition in invasion and metastasisEpithelial and mesenchymal transition in invasion and metastasis
Epithelial and mesenchymal transition in invasion and metastasis
 
Pancreatic cancer
Pancreatic cancerPancreatic cancer
Pancreatic cancer
 
Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...
Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...
Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...
 
Compartment specific micro rna expression profiles (poster) poster
Compartment specific micro rna expression profiles (poster) posterCompartment specific micro rna expression profiles (poster) poster
Compartment specific micro rna expression profiles (poster) poster
 
A physical sciences network characterization of non-tumorigenic and metastati...
A physical sciences network characterization of non-tumorigenic and metastati...A physical sciences network characterization of non-tumorigenic and metastati...
A physical sciences network characterization of non-tumorigenic and metastati...
 
3478-43782-3-PB
3478-43782-3-PB3478-43782-3-PB
3478-43782-3-PB
 
2013 CD8 Lymphocytes and apoptosis in MBC-1
2013 CD8  Lymphocytes and apoptosis in MBC-12013 CD8  Lymphocytes and apoptosis in MBC-1
2013 CD8 Lymphocytes and apoptosis in MBC-1
 
Histopathological Patterns of Germ Cell Tumours of Ovary in a Tertiary Level ...
Histopathological Patterns of Germ Cell Tumours of Ovary in a Tertiary Level ...Histopathological Patterns of Germ Cell Tumours of Ovary in a Tertiary Level ...
Histopathological Patterns of Germ Cell Tumours of Ovary in a Tertiary Level ...
 
Immuotherapy 2
Immuotherapy 2Immuotherapy 2
Immuotherapy 2
 
Research issues in gastric cancer
Research issues in gastric cancerResearch issues in gastric cancer
Research issues in gastric cancer
 
Gastic Cancer
Gastic CancerGastic Cancer
Gastic Cancer
 
EmergingUnderstandingofMultiscaleTumorHeterogeneityDecember2014
EmergingUnderstandingofMultiscaleTumorHeterogeneityDecember2014EmergingUnderstandingofMultiscaleTumorHeterogeneityDecember2014
EmergingUnderstandingofMultiscaleTumorHeterogeneityDecember2014
 
Kurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLSKurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLS
 
biochem of cancer modified dialysis treatment
biochem of cancer modified dialysis treatmentbiochem of cancer modified dialysis treatment
biochem of cancer modified dialysis treatment
 
Breast Cancer Stem Cells
Breast Cancer Stem CellsBreast Cancer Stem Cells
Breast Cancer Stem Cells
 
Molecular mechanism of tumor invasiveness
Molecular mechanism of tumor invasivenessMolecular mechanism of tumor invasiveness
Molecular mechanism of tumor invasiveness
 
Growth Kinetics of 2- and 3-D Cell Models as Influenced by the Microenvironment
Growth Kinetics of 2- and 3-D Cell Models as Influenced by the MicroenvironmentGrowth Kinetics of 2- and 3-D Cell Models as Influenced by the Microenvironment
Growth Kinetics of 2- and 3-D Cell Models as Influenced by the Microenvironment
 
Disseminated Breast Cancer Cells Acquire a Highly Malignant and Aggressive Me...
Disseminated Breast Cancer Cells Acquire a Highly Malignant and Aggressive Me...Disseminated Breast Cancer Cells Acquire a Highly Malignant and Aggressive Me...
Disseminated Breast Cancer Cells Acquire a Highly Malignant and Aggressive Me...
 
JTM-Functional characterization of human Cd33+ And Cd11b+ myeloid-derived sup...
JTM-Functional characterization of human Cd33+ And Cd11b+ myeloid-derived sup...JTM-Functional characterization of human Cd33+ And Cd11b+ myeloid-derived sup...
JTM-Functional characterization of human Cd33+ And Cd11b+ myeloid-derived sup...
 
Breast Cancer Genetic Characteristics in North Africa: A Genome Wide Haplotyp...
Breast Cancer Genetic Characteristics in North Africa: A Genome Wide Haplotyp...Breast Cancer Genetic Characteristics in North Africa: A Genome Wide Haplotyp...
Breast Cancer Genetic Characteristics in North Africa: A Genome Wide Haplotyp...
 

Similar to PaperRozenchan 2015

Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael Araya
 
Rozenchan_et_al-2009-International_Journal_of_Cancer
Rozenchan_et_al-2009-International_Journal_of_CancerRozenchan_et_al-2009-International_Journal_of_Cancer
Rozenchan_et_al-2009-International_Journal_of_Cancer
Patricia Rozenchan
 
Rozenchan_et_al-2009-International_Journal_of_Cancer
Rozenchan_et_al-2009-International_Journal_of_CancerRozenchan_et_al-2009-International_Journal_of_Cancer
Rozenchan_et_al-2009-International_Journal_of_Cancer
Patricia Rozenchan
 
Majumder_B_et_al_Nature_Communications_2015
Majumder_B_et_al_Nature_Communications_2015Majumder_B_et_al_Nature_Communications_2015
Majumder_B_et_al_Nature_Communications_2015
Michelle Stevens
 

Similar to PaperRozenchan 2015 (20)

Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW edits
 
Rozenchan_et_al-2009-International_Journal_of_Cancer
Rozenchan_et_al-2009-International_Journal_of_CancerRozenchan_et_al-2009-International_Journal_of_Cancer
Rozenchan_et_al-2009-International_Journal_of_Cancer
 
Rozenchan_et_al-2009-International_Journal_of_Cancer
Rozenchan_et_al-2009-International_Journal_of_CancerRozenchan_et_al-2009-International_Journal_of_Cancer
Rozenchan_et_al-2009-International_Journal_of_Cancer
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
Functional Disparity of Carcinoma Associated Fibroblasts in Different Stages ...
 
Suppress lung cancer progression via up regulation of linc rna-p21
Suppress lung cancer progression via up regulation of linc rna-p21Suppress lung cancer progression via up regulation of linc rna-p21
Suppress lung cancer progression via up regulation of linc rna-p21
 
Low expression of the X-linked ribosomal protein S4 in human serous epithelia...
Low expression of the X-linked ribosomal protein S4 in human serous epithelia...Low expression of the X-linked ribosomal protein S4 in human serous epithelia...
Low expression of the X-linked ribosomal protein S4 in human serous epithelia...
 
Majumder_B_et_al_Nature_Communications_2015
Majumder_B_et_al_Nature_Communications_2015Majumder_B_et_al_Nature_Communications_2015
Majumder_B_et_al_Nature_Communications_2015
 
The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...
The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...
The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...
 
Salivary biomarkers
Salivary biomarkersSalivary biomarkers
Salivary biomarkers
 
ASCB 2014 poster
ASCB 2014  posterASCB 2014  poster
ASCB 2014 poster
 
Alterations of Gut Microbiota From Colorectal Adenoma to Carcinoma
Alterations of Gut Microbiota From Colorectal Adenoma to CarcinomaAlterations of Gut Microbiota From Colorectal Adenoma to Carcinoma
Alterations of Gut Microbiota From Colorectal Adenoma to Carcinoma
 

PaperRozenchan 2015

  • 1. RESEARCH ARTICLE Specific upregulation of RHOA and RAC1 in cancer-associated fibroblasts found at primary tumor and lymph node metastatic sites in breast cancer Patricia Bortman Rozenchan1,2,3 & Fatima Solange Pasini1 & Rosimeire A. Roela1 & Maria Lúcia Hirata Katayama1 & Fiorita Gonzáles Lopes Mundim4 & Helena Brentani5 & Eduardo C. Lyra6 & Maria Mitzi Brentani1 Received: 2 April 2015 /Accepted: 28 June 2015 # International Society of Oncology and BioMarkers (ISOBM) 2015 Abstract The importance of tumor–stromal cell interactions in breast tumor progression and invasion is well established. Here, an evaluation of differential genomic profiles of carcinoma-associated fibroblasts (CAFs) compared to fibro- blasts derived from tissues adjacent to fibroadenomas (NAFs) revealed altered focal adhesion pathways. These data were validated through confocal assays. To verify the possible role of fibroblasts in lymph node invasion, we constructed a tissue microarray consisting of primary breast cancer samples and corresponding lymph node metastasis and compared the ex- pression of adhesion markers RhoA and Rac1 in fibroblasts located at these different locations. Two distinct tissue micro- arrays were constructed from the stromal component of 43 primary tumors and matched lymph node samples, respective- ly. Fibroblasts were characterized for their expression of α- smooth muscle actin (α-SMA) and vimentin. Moreover, we verified the level of these proteins in the stromal compartment from normal adjacent tissue and in non-compromised lymph nodes. Our immunohistochemistry revealed that 59 % of fi- broblasts associated with primary tumors and 41 % of the respective metastatic lymph nodes (p=0.271) displayed posi- tive staining for RhoA. In line with this, 57.1 % of fibroblasts associated with primary tumors presented Rac1-positive stain- ing, and the frequency of co-positivity within the lymph nodes was 42.9 % (p=0.16). Expression of RhoA and Rac1 was absent in fibroblasts of adjacent normal tissue and in compro- mised lymph nodes. Based on our findings that no significant changes were observed between primary and metastatic lymph nodes, we suggest that fibroblasts are active partici- pants in the invasion of cancer cells to lymph nodes and sup- port the hypothesis that metastatic tumor cells continue to depend on their microenvironment. Keywords Breast cancer . Tumor microenvironment . Carcinoma-associated fibroblasts . Rho GTPases . Metastasis Introduction Stromal cells, including fibroblasts, endothelial cells, and im- mune cells, play a critical role in supporting breast cancer (BC) growth, survival, and invasion. Fibroblasts represent the major cell type of the stromal compartment and play an important role in coordinating interactions between stromal and tumor cells by modulating the composition and function of the extracellular matrix. Carcinoma-associated fibroblasts (CAFs) present different characteristics from those of fibro- blasts found in normal breast tissue (NAFs). CAFs form a heterogeneous population [1], express alpha-smooth muscle actin (α-SMA) upon activation [2], and impact BC biological behaviors [3]. * Patricia Bortman Rozenchan pbrozenchan@hotmail.com 1 Radiology and Oncology Department, School of Medicine of São Paulo University, Av. Dr. Arnaldo, 455, sala 4112, São Paulo, SP CEP 01246-903, Brazil 2 Colsan—Blood Bank Beneficent Association, São Paulo, SP, Brazil 3 Gynecological Department, School of Medicine of São Paulo Federal University, São Paulo, SP, Brazil 4 Pathology Department, Vale do Sapucaí University, Pouso Alegre, MG, Brazil 5 Psychiatric Department, School of Medicine of São Paulo University (USP), Rua Ovideo Pires de Campos S/N, São Paulo, SP, Brazil 6 Brazilian Institute of Cancer Control, Av. Alcântara Machado 2576, São Paulo, SP CEP 03102-002, Brazil Tumor Biol. DOI 10.1007/s13277-015-3727-1
  • 2. The importance of interactions between tumor cells and the surrounding stromal cells [4] has been well established. In particular, the migratory/invasive behavior of tumor cells has been reported to be influenced by bidirectional signals be- tween cancer cells and tumor-associated stroma fibroblasts and affects human breast cancer cell adhesion, migration speed, and direction [5, 6]. During cell invasion, fibroblasts use contractile force and proteolytic activity to reorganize col- lagen into linear fibers to generate tracks for migration of cancer cells [7]. Notably, family members of the Rho-small guanosine triphosphatases (GTPases), RhoA, RAC1, and RAC2 binding proteins, have been implicated in CAF- mediated remodeling of the tumor microenvironment in a manner that enhances cancer cell invasion [8]. Indeed, RhoA, Rac1, and Rac2 induce stress fiber formation when overexpressed in fibroblasts [9]. In the present study, we analyzed genes that were differen- tially expressed in primary breast CAFs compared to fibro- blasts that originated from adjacent tissue of benign breast diseases. This analysis revealed numerous differences in genes involved in major functional pathways, including focal adhesion and regulation of actin cytoskeleton and tight junc- tions. Specifically, the GTPase family members RhoA, RAC1, and RAC2, as well as collagens and integrins, which are known to mediate migration and invasiveness, were shown to be differentially regulated in the context of CAFs. To date, increased expression of these proteins, particularly within the epithelial compartment, has been shown to be im- portant for cellular motility, loss of adhesion, invasion, and metastasis [10–13]. Nodal status represents one of the most powerful inde- pendent prognostic indicators of breast carcinoma [14]. Histologically, the presence of fibroblasts in the BC micro- environment of metastatic lymph nodes further reinforces the role that CAFs play in tumor growth and dissemination [15]. Le Bedis et al. [16] suggested that lymph node stroma plays an active part in the process of lymph node metasta- sis by creating a dynamic microenvironment that mimics the environmental conditions present at the primary tumor site. Previous studies by Garcia et al. [17] reported a sim- ilar expression of metalloproteases within primary tumors and the respective lymph nodes. Moreover, tumor–stroma cross-talk seems to influence the metastatic lymph node microenvironment, affecting proliferation and migration of cancer cells [16, 18]. Because the genomic profiles of fibroblasts BC primary and lymph node metastasis have been reported to be similar [19], we hypothesized that similar levels of RhoA and Rac1 expression in the tumor microenvironment at the primary site and at the lymph nodes may represent an advantage to tumor cell behavior. In this study, we determined whether the expres- sion of RhoA and Rac1 in stromal fibroblasts of primary tu- mors was similar to that found in lymph node fibroblasts. Using a tissue microarray (TMA) consisting of primary BC samples and the corresponding lymph node metastasis, we compared the expression level of these markers in fibroblasts residing at the two locations, as well as in adjacent normal tissue of primary tumors and in non-compromised lymph nodes. Materials and methods Primary cell culture tissue samples Malignant and benign breast tissue specimens were obtained from consenting patients undergoing surgery for breast dis- ease. Carcinoma samples were obtained from four patients clinically staged as IIa, and benign samples were obtained from four patients diagnosed as fibroadenoma. All tissue do- nors were patients at Instituto Brasileiro de Controle do Cân- cer, São Paulo, Brazil, a reference center for cancer treatment. This study was approved by the Ethical Institutional Commit- tee, and written explicit informed consent was obtained from all participants. Invasive breast cancer was confirmed histopathologically. Primary cell culture Fibroblasts were obtained from normal adjacent tissue sam- ples from patients with benign breast diseases (NAF) or with primary invasive breast cancer tumors (CAF). H&E- stained, frozen histological sections were prepared from each tissue sample to confirm benignity or malignancy. After adipose tissue removal, tissue was minced in phosphate-buffered saline (PBS), washed twice in PBS (Na2HPO4 10 mM, NaCl 1.37 mM, KCl 27 mM, KH2PO4 2 mM, Thermo Fisher Scientific Inc., MA, USA) and in culture medium, and then chopped into small 1- to 4-mm3 pieces under sterile conditions. A total of 15–30 fragments were transferred to a T25 culture flask (Thermo Fisher Sci- entific Inc.) and covered with Dulbecco’s modified Eagle’s medium (DMEM, Thermo Fisher Scientific Inc.) supple- mented with 20 % FBS (Thermo Fisher Scientific Inc.), 100 μg/mL ampicillin, 100 μg/mL streptomycin, and 2.5 μg/mL fungizone and maintained at 37 °C in a humid- ified atmosphere containing 5 % CO2. Outgrowth of cells was recorded after 10 to 20 days, and the medium was renewed once or twice a week thereafter. After sufficient outgrowth, the tissue fragments were removed and the cells were passaged by mild trypsinization with trypsin 0.5 % (Thermo Fisher Scientific Inc.). Early passages (passage 3) of all fibroblasts were subjected to immunocytochemical evaluation. The prolif- eration rates and description of patients were described before [20]. Tumor Biol.
  • 3. cDNA microarray assembly, hybridization, and analysis Total RNA from four NAFs and four CAFs were extracted by TRIzol reagent (Thermo Fisher Scientific Inc.) and purified with RNeasy minicolumns and reagents (Qiagen, Hilden, Ger- many). RNA microarray analysis was performed with 10 μg of biotin-labelled cRNA target prepared by a linear amplifica- tion method from a pool of samples. The poly (A)+ RNA (mRNA) subpopulation within the total RNA population was primed for reverse transcription by a DNA oligonucleotide containing the T7 RNA polymerase promoter 5′ to a d (T)24 sequence. After second-strand cDNA synthesis, the cDNA served as the template for an in vitro transcription (IVT) reac- tion to produce the target cRNA. This cRNA was hybridized using CodeLinkTM Human Whole Genome 55K Bioarray (GE Healthcare, Buckinghamshire, UK), and the hybridization sig- nals were normalized using the CodeLinkTM System Software Analysis (GE Healthcare) and subjected to a t test with 10,000 permutations. Genes with twofold differential expression levels in the CAF versus NAF comparison were considered differentially expressed. The gene ontology (GO) analysis was performed using the GO Tree Machine tool (GOTM) [21], which identifies hyper-represented categories in our gene lists as well as the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. For both GO and KEGG, we used a hyper-geometric distribution with a p value≤0.05 [Onto-tolls]. Fluorescence microscopy Cells were cultured on glass coverslips in 24-well plates. After 24 h, cells were fixed with 3.5 % paraformaldehyde in PBS at room temperature, permeabilized for 5 min with 0.2 % Triton X-100 in PBS, and blocked with 5 % bovine serum albumin (BSA, Thermo Fisher Scientific Inc.) in PBS for 1 h. The primary chicken polyoclonal RhoA antibody (ab23687, Abcam Antibodies, Cambridge, UK) and mouse monoclonal Rac1 antibody (ab33186, Abcam Antibodies) were incubated overnight at 4 °C. After extensive washes and secondary an- tibody staining (goat anti-mouse IgG, anti-chicken IgG, and anti-rabbit IgG; Sigma-Aldrich, MO, USA), nuclei were stained with 4′-6′-diamidino-2-phenylindole (DAPI, Sigma- Aldrich). Cells were analyzed using a Zeiss 510 META con- focal laser scanning microscope using a 488-nm argon and a 543-nm HeNe laser. Images were acquired using a Plan NeoFluoar 40×/1 lens (Microimaging Inc., Lane Cove, New Zealand). Immunohistochemistry tissue samples This study included 43 breast cancer samples that involved axillary nodes from patients undergoing breast surgery at Hos- pital Samuel Libânio, in Pouso Alegre, MG, Brazil from 1997 to 2005. This study was approved by the Institutional Ethics Committee of Hospital Samuel Libânio (no. 1119/09). The median age of patients was 58 years (range 38–88 years). All patients were diagnosed with invasive ductal carcinomas at clinical stages II and III in 37.2 or 62.8 % of cases, respectively (two unknown). Statuses of the estrogen receptor (ER), pro- gesterone receptor (PR), HER-2, and histological grade (HG) are listed in Table 1. HER-2 was assessed by the HercepTestTM (Dako, Glostrup, Denmark A/S) system. Membrane-based staining cases with scores 0–1 or chromogenic in situ hybrid- ization (CISH) negative were categorized as negative, while cases with score ≥3 were considered as positive. The cases with an immunohistochemistry score of 2 were further con- firmed by CISH analysis (Ventana Medical Systems, Inc., a member of the Roche Group, Tucson, AZ) [22]. Additionally, we included 10 female BC cases (aged be- tween 39–84 years) in spite of analyzing the stromal compart- ment of adjacent normal tissue of primary tumors and not compromised lymph nodes. All samples were subjected to a pathology analysis, where tumor margins were considered to assure consistency within the tissues. The Pathology Reporting of Breast Disease was used as guidelines [23]. Construction of TMA Two distinct TMAs were constructed from the stromal com- ponent of primary tumors and lymph node samples, Table 1 Clinical and pathological parameters of 43 breast cancer patients Characteristic No. of patients (%) Age median (range) 58 (38–88) Clinical stage I 1 (2.3) II 16 (37.2) III 26 (60.5) Histologic grade Grade I 9 (20.9) Grade II 15 (34.9) Grade III 19 (44.2) ER—primary tumor Negative 18 (41.5) Positive 24 (55.8) Missing 1 (2.3) PR—primary tumor Negative 17 (39.5) Positive 21 (48.8) Missing 5 (11.6) HER-2 Negative 33 (76.7) Positive 9 (20.9) Missing 1 (2.3) Tumor Biol.
  • 4. respectively. Representative areas of each component were surrounded by a marking pen at the donor blocks and collect- ed using the Manual Tissue Arrayer I (Beecher Instruments Inc., Sun Prairie, USA). Samples were orderly arranged in a grid, and the first core was represented by a fragment or nor- mal liver used as a reference in both TMAs. The first TMA was built with samples of stromal component of the tumors in order to enable the assessment of fibroblast cells within the desmoplastic contingent of the carcinomas. The second TMA consisted of 43 majorly compromised lymph node samples from cases with axillary lymph node metastasis. Because the width of the metastatic element at the lymph nodes was frequently limited to 0.5 to 1.0 cm, it was seldom possible to separate distinct areas of stromal and epithelial elements. The chosen areas at the donor block in- volved stromal and epithelial elements, which were further evaluated separately. From each of the two TMAs, 3-μm-thick slices were ob- tained and collected on slides with special adhesives (Instrumedics Inc., NJ, USA). Two sets of triplets, separated by a gap of 40 cuts, were submitted for immunohistochemical analyses in order to represent two levels of the same sample. Immunohistochemistry The immunohistochemical reactions were performed using the complex streptavidin-biotin peroxidase (StreptABC, Dako Corporation, Glostrup, Denmark). After deparaffinization of tissue sections, antigen retrieval was performed using a pres- sure cooker in citrate buffer pH 6.0, followed by blocking endogenous peroxidase with hydrogen peroxide solution (3 %). The sections were incubated with primary antibodies, Rac1 (1:1,000; ab33186, Abcam) and RhoA (1:175; ab23687, Abcam), vimentin (1:2,000; clone v9, Dako Corporation), and α-SMA (clone: HHF-35; Cell Marque, CA, USA). After in- cubation with primary antibody and primary blocking, a poly- mer–peroxidase (Novolink, Leica, Wetzlar, Germany) ampli- fication step was performed. Antigen detection was carried out in a solution containing 3,3-diaminobenzidine (Sigma- Aldrich) and 6 % H2O2. Counterstaining was performed with Harris hematoxylin (Merck, NJ, USA). Carcinoma ductal in- vasive of breast was used as positive control for RhoA, and large intestine adenocarcimona was the positive control used for Rac1. Negative controls were performed removing first antibodies. Evaluation of immunohistochemical essays All reactions were assessed by two independent blinded ob- servers. Disparities between the two pathologists were reevaluated by consensus. Results from the epithelial compo- nent and stromal component were reported separately either to carcinomas or to lymph node samples. The presence of RhoA and Rac1 staining was assessed in the stromal population and separately in primary tumors or in lymph node metastases. In relation to the immunohistochem- ical result evaluation, RhoA was scored according to the Allred system [24]. Samples with a score above 4 were con- sidered positive. For Rac1 evaluation, the percentage of pos- itive cells was assessed and classified into three groups: (1) 0 %, (2) 1−33 %, and (3) ≥34 %. Samples with greater than 10 % of cells that were positive for α-SMA and vimentin were classified as positive. For all cases, cells presented in 10 fields at a magnification of ×400 were counted. Statistical methods Correlations between categorical antigen expression and other clinicopathological parameters were studied with the Fisher’s exact test or chi-square test, where appropriate. Spearman’s rank correlation coefficient was calculated to assess the rela- tionships involving categorical antigen expression. All statis- tical tests were two-sided, with significance defined as p<0.05. Analyses were performed using the software SPSS version 10.0 for Windows (SPSS Inc., IL, USA). Results First, we evaluated the gene expression profile of stromal fi- broblasts derived from four benign (NAF) and four malignant (CAF) breast tissues using the CodeLinkTM Human Whole Genome 55K Bioarray. Using a twofold cutoff, we found 1, 111 genes differentially expressed in CAFs compared to NAFs. To further examine the biological functions of these genes, an analysis of the KEGG database revealed differential expression of several pathways, including Boxidative phos- phorylation,^ “focal adhesion,” BMAPK signaling pathway,^ Bleukocyte transendothelial migration,^ Bregulation of actin cytoskeleton,^ and Btight junctions^ (Fig. 1). Interestingly, all of these pathways are involved in important processes like cell/cell communication, migration, and invasiveness. Focal adhesion was ranked the second top significant pathway, and in microarray analysis, we found that both RhoA and Rac1 were upregulated in CAFs compared to NAFs. Using confocal assays, we confirmed that protein levels of RhoA and Rac1 were elevated in carcinoma-associated fibro- blasts (Fig. 2). Strong RhoA staining was localized to the cytoplasm, while Rac1 staining was associated with the plas- ma membrane. To further examine correlations between altered genes found in the microarray data with patterns of protein expres- sion, we analyzed the expression of RhoA and Rac1 in the stromal component of 43 samples of primary breast carcinoma and metastatic ipsilateral axillary lymph nodes by immunohis- tochemistry. In a few metastatic lesions cases, it was not Tumor Biol.
  • 5. Fig. 1 Distribution of differentially expressed genes in CAFs. Representative pathways were identified for genes differentially expressed (black bars) between CAFs and NAFs in our cDNA microarray platform (gray bars). The gene ontology (GO) analysis was performed using the GO Tree Machine tool (GOTM), which identifies hyper- represented categories in our gene lists as well as the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. For both GO and KEGG, we used a hyper-geometric distribution with a p value≤0.05 [Onto-tolls] Fig. 2 CAF-specific staining of RhoA and Rac1. CAF (a, c) and NAF (b, d) confocal photomicrographs of typical fields are shown. Merged images of a cytoplasmatic staining of RhoA (a, b) and cytoplasmatic membrane staining of Rac1 (c, d) validated the gene expression analysis; Rac1 and RhoA were upregulated in CAFs Tumor Biol.
  • 6. possible to count the preferred number of cells. These proteins were also analyzed in fibroblasts of adjacent normal tissue of primary tumors and in non-compromised lymph nodes of 10 breast cancer cases. To distinguish fibroblasts (CAFs) from tumoral cells, we evaluated two fibroblast cell markers: vimentin and α-SMA. Vimentin staining was observed in 37/43 (86.0 %) of fibro- blasts of primary tumors and in 35/41 (85.4 %) of lymph nodal fibroblasts. α-SMA staining was observed in 51.2 % of pri- mary lesions and in 41 % of nodal metastatic lesions (Fig. 3). Positive RhoA staining in stromal cells was seen in 36/43 cases of primary tumors (59 %) and in 25/35 (41 %) of respec- tive metastatic lymph nodes (p=0.271). Similarly, Rac1 stain- ing in stromal cells was observed in 28/39 cases of primary tumors (57.1 %), whereas the frequency of Rac1 staining in stromal cells found in the lymph nodes was 21/38 (42.9 %) (Table 2). In summary, the status of RhoA and Rac1 was similar between primaries and lymph nodes (Fig. 4), and all markers were detected in the cytoplasm. Importantly, RhoA or Rac1 expression was not detected in fibroblasts of adjacent normal tissue or in non-compromised lymph nodes (Fig. 5), suggesting that the presence of RhoA or Rac1 in CAFs may help facilitate tumorigenesis. Discussion A large amount of data have shown that the microenvironment is important to breast carcinoma epithelial cells [25–29]. Ad- ditionally, we have shown that reciprocal changes in gene expression profiles of important cellular function pathways occur when breast epithelial cells were co-cultured with primary fibroblasts, reinforcing the idea that interactions be- tween these two cell types impact cell signaling and behavior [20]. In line with these works, we compared gene expression data generated in CAFs and NAFs to show that a large number of genes present in the focal adhesion pathways, including the GTPAses RhoA and Rac1, were differentially expressed in CAFs. Cell motility and invasiveness require cytoskeleton reorga- nization, which involves formation of the filopodia/ lamellipodia and changes in focal adhesion complexes. Rho proteins are involved in stress fiber formation and focal adhe- sion, while Rac proteins stimulate lamellipodia and membrane-ruffle formation [30–33]. Increasing evidence sug- gests that the RhoA and Rac1 proteins play an important role in cell migration, loss of adhesion, invasion, and metastasis in tumors [10, 34–37]. Building on these studies, we used im- munohistochemistry to investigate whether the RhoA and Rac1 expression patterns were similar in fibroblasts of prima- ry BC tumors and matched lymph node metastases. Fig. 3 CAF-specific α-SMA and vimentin staining at primary at metastatic tumor sites. α-SMA (a, b) and vimentin (c, d) staining in CAFs in the primary tumors (a, c) and in their counterpart lymph node metastasis (b, d). Original magnification ×400 Table 2 Correlation among the proportion of biological marker expression in stromal tissue between the primary tumors and lymph node metastasis of 43 breast cancer patients Variable Primary tumor Lymph node p RhoA Negative 7 (41.2) 10 (58.8) 0.27 Positive 36 (59.0) 25 (41.0) Rac1 Negative 11 (39.3) 17 (60.7) 0.16 Positive 28 (57.1) 21 (42.9) Stroma tissue: only fibroblast cells were analyzed; Values of p (two-sided) less than 0.05 were considered significant Tumor Biol.
  • 7. Early studies noted that RhoA overexpression result- ed in mouse fibroblast transformation [38] and stress fiber formation [39]. It was previously shown that high- ly metastatic mesenchymal sarcoma cells primarily use an ameboid mode of cell invasion that depends on the activity of the Rho family of GTPases [40]. Some au- thors have described the role of Rho GTPases in fibro- blasts [41–43]. Verghese et al. [41] reported that cyto- skeletal regulation by Rho GTPases in breast fibroblasts enhanced migration and invasion in consequence of mir- 26b dysregulation. In agreement with Halon et al. [10], we show that a high proportion of cells in the BC tumor–stromal compartment were positive for RhoA and Rac1. In addition, we have shown that activated fibroblasts, as identified by α-SMA, were pres- ent in the majority of metastatic lymph node cases, whereas non-involved lymph nodes were devoid of myofibroblasts. The epithelial-to-mesenchymal transition (EMT) repre- sents one possible mechanism by which a cancer cell metas- tasizes and may facilitate the re-localization of fibroblasts to the lymph node. Past work has emphasized RhoA as a medi- ator of this process via integrin β1/TGF β activation [44]. Fig. 4 RhoA and Rac1 staining in primaries and in lymph node metastasis. Representative cases depicting RhoA and Rac1 (a, c) in CAFs in the primary tumors and in the respective lymph node metastasis (b, d). Original magnification ×400 Fig. 5 Lack of RhoA and Rac1 staining in fibroblasts of adjacent normal tissue and in non- compromised lymph nodes. Negative expression of RhoA and Rac1 in fibroblasts of adjacent normal tissue (a, c) and non- compromised lymph nodes (b, d). Original magnification ×400 Tumor Biol.
  • 8. Moreover, EMT is a recognized source of CAFs, as it pro- duces myofibroblast cells with enhanced migratory capacity, invasiveness, and increased expression of ECM proteins [45]. In line with these data, we found that 41 % of fibroblasts of lymph nodes origin were positive for RhoA and α-SMA. CAFs may also arrive at lymph nodes in co-migration with tumor cells, referred to as the collective pathway of invasion [46]. Reorganization of collagen fibers by mammary fibro- blasts can create avenues for invasion [5], and the Rho- family of GTP-binding proteins may regulate fibroblast- mediated collagen reorganization [8]. Therefore, RhoA and Rac1 may be involved in both pathways. Our study brings some clarification regarding the role of the microenvironment in lymph node metastasis in BC. How- ever, we acknowledge that our study is limited by the number of samples used in immunohistochemical assays, as well as by the presence of non-distinct histological types, as different subtypes represent different fibroblasts populations [47]. In conclusion, we demonstrate positive expression of RhoA and Rac1 in CAFs in paired primary breast cancer and in the respective lymph node metastasis, suggesting that a similar microenvironment may be present at both sites. Im- portantly, as we were unable to detect positive expression of these proteins in fibroblasts of normal adjacent tissue or in non-committed lymph nodes, our findings may highlight the stroma as an active participant in the metastatic process and suggest that metastatic tumor cells may continue to be depen- dent on their supportive microenvironment. Acknowledgments The authors are grateful to Ana Lúcia Garippo for her technical assistance in confocal microscopy. This research was sup- ported by Fundação de Amparo à Pesquisa no Estado de São Paulo (FAPESP) 01/13513-1, 05/51593-5, 04/04607-8, 05/60333-7, 2014/ 03090-3 and 09/10088-7 and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq). Conflicts of interest None Ethical approval All procedures performed in studies involving hu- man participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Hel- sinki declaration and its later amendments or comparable ethical standards. References 1. Sugimoto H, Mundel TM, Kieran MW, Kalluri R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol Ther. 2006;5(12):1640–6. 2. Xouri G, Christian S. Origin and function of tumor stroma fibro- blasts. Semin Cell Dev Biol. 2010;21(1):40–6. 3. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6(5):392–401. 4. Polyak K, Kalluri R. The role of the microenvironment in mamma- ry gland development and cancer. Cold Spring Harb Perspect Biol. 2010;2(11):a003244. 5. Dang TT, Prechtl AM, Pearson GW. Breast cancer subtype-specific interactions with the microenvironment dictate mechanisms of in- vasion. Cancer Res. 2011;71(21):6857–66. 6. Angelucci C, Maulucci G, Lama G, Proietti G, Colabianchi A, Papi M, et al. Epithelial-stromal interactions in human breast cancer: effects on adhesion, plasma membrane fluidity and migration speed and directness. PLoS One. 2012;7(12):e50804. doi:10.1371/ journal.pone.0050804. 7. Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW, Provenzano PP, et al. Aligned collagen is a prognostic signature for survival in human breast carcinoma. Am J Pathol. 2011;178(3): 1221–32. 8. Rhee S, Grinnell F. Fibroblast mechanics in 3D collagen matrices. Adv Drug Deliv Rev. 2007;59(13):1299–305. 9. Wheeler AP, Ridley AJ. Why three Rho proteins? RhoA, RhoB, RhoC, and cell motility. Exp Cell Res. 2004;301(1):43–9. 10. Halon A, Donizy P, Surowiak P, Matkowski R. ERM/Rho protein expression in ductal breast cancer: a 15 year follow-up. Cell Oncol. 2013;36(3):181–90. 11. Wu YJ, Tang Y, Li ZF, Li Z, Zhao Y, Wu ZJ, Su Q. Expression and significance of Rac1, Pak1 and Rock1 in gastric carcinoma. Asia Pac J Clin Oncol. 2013; doi: 10.1111/ajco.12052 12. Ridley AJ. Rho proteins and cancer. Breast Cancer Res Treat. 2004;84(1):13–9. 13. Sahai E, Marshall CJ. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol. 2003;5(8):711–9. 14. Yenidunya S, Bayrak R, Haltas H. Predictive value of pathological and immunohistochemical parameters for axillary lymph node me- tastasis in breast carcinoma. Diagn Pathol. 2011;6:18. doi:10.1186/ 1746-1596-6-18. 15. Cao Y, Paner GP, Rajan PB. Sentinel node status and tumor char- acteristics: a study of 234 invasive breast carcinomas. Arch Pathol Lab Med. 2005;129(1):82–4. 16. LeBedis C, Chen K, Fallavollita L, Boutros T, Brodt P. Peripheral lymph node stromal cells can promote growth and tumorigenicity of breast carcinoma cells through the release of IGF-I and EGF. Int J Cancer. 2002;100(1):2–8. 17. García MF, González-Reyes S, González LO, Junquera S, Berdize N, Del Casar JM, et al. Comparative study of the expression of metalloproteases and their inhibitors in different localizations with- in primary tumours and in metastatic lymph nodes of breast cancer. Int J Exp Pathol. 2010;91(4):324–34. 18. Montel V, Mose ES, Tarin D. Tumor-stromal interactions recipro- cally modulate gene expression patterns during carcinogenesis and metastasis. Int J Cancer. 2006;119(2):251–63. 19. Del Valle PR, Milani C, Brentani MM, Katayama ML, de Lyra EC, Carraro DM, et al. Transcriptional profile of fibroblasts obtained from the primary site, lymph node and bone marrow of breast can- cer patients. Genet Mol Biol. 2014;37(3):480–9. 20. Rozenchan PB, Carraro DM, Brentani H, et al. Reciprocal changes in gene expression profiles of cocultured breast epithelial cells and primary fibroblasts. Int J Cancer. 2009;125(12):2767–77. 21. Onto-Tools data base [http://vortex.cs.wayne.edu/projects.htm]. 22. Wolff AC, Hammond ME, Hicks DG, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Arch Pathol Lab Med. 2014;138:241–56. 23. Pathology Reporting of Breast Disease. A Joint Document Incorporating the Third Edition of the NHS Breast Screening Programme’s Guidelines for Pathology Reporting in Breast Cancer Screening and the Second Edition of The Royal College of Pathologists’ Minimum Dataset for Breast Cancer Histopathology. NHSBSP Publication No 58. 2005. Tumor Biol.
  • 9. 24. Allred DC, Harvey JM, Berardo M, et al. Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol. 1998;11:155–68. 25. Casbas-Hernandez P, Fleming JM, Troester MA. Gene expression analysis of in vitro cocultures to study interactions between breast epithelium and stroma. J Biomed Biotechnol. 2011; 520987. doi: 10.1155/2011/520987 26. Hawsawi NM, Ghebeh H, Hendrayani SF, et al. Breast carcinoma- associated fibroblasts and their counterparts display neoplastic- specific changes. Cancer Res. 2008;68(8):2717–25. 27. Casey T, Bond J, Tighe S, et al. Molecular signatures suggest a major role for stromal cells in development of invasive breast can- cer. Breast Cancer Res Treat. 2009;114:47–62. 28. Singer CF, Gschwantler-Kaulich D, Fink-Retter A, et al. Differential gene expression profile in breast cancer-derived stro- mal fibroblasts. Breast Cancer Res Treat. 2008;110(2):273–81. 29. Buess M, Nuyten DS, Hastie T, Nielsen T, Pesich R, Brown PO. Characterization of heterotypic interaction effects in vitro to deconvolute global gene expression profiles in cancer. Genome Biol. 2007;8(9):R191. 30. Dummler B, Ohshiro K, Kumar R, Field J. Pak protein kinases and their role in cancer. Cancer Metastasis Rev. 2009;28(1–2):51–63. 31. Arias-Romero LE, Villamar-Cruz O, Pacheco A, Kosoff R, Huang M, Muthuswamy SK, et al. A Rac-Pak signaling pathway is essen- tial for ErbB2-mediated transformation of human breast epithelial cancer cells. Oncogene. 2010;29(43):5839–49. 32. Rider L, Oladimeji P, Diakonova M. PAK1 regulates breast cancer cell invasion through secretion of matrix metalloproteinases in re- sponse to prolactin and three-dimensional collagen IV. Mol Endocrinol. 2013;27(7):1048–64. 33. Shin YJ, Kim EH, Roy A, Kim JH. Evidence for a novel mecha- nism of the PAK1 interaction with the Rho-GTPases Cdc42 and Rac. PLoS One. 2013;8(8):e71495. doi:10.1371/journal.pone. 0071495. 34. Chan CH, Lee SW, Li CF, Wang J, Yang WL, Wu CY, et al. Deciphering the transcriptional complex critical for RhoA gene expression and cancer metastasis. Nat Cell Biol. 2010;12(5):457– 67. 35. Fritz G, Brachetti C, Bahlmann F, Schmidt M, Kaina B. Rho GTPases in human breast tumours: expression and mutation anal- yses and correlation with clinical parameters. Br J Cancer. 2002;87(6):635–44. 36. Chang YW, Marlin JW, Chance TW, et al. RhoA mediates cyclooxygenase-2 signaling to disrupt the formation of adherens junctions and increase cell motility. Cancer Res. 2006;66(24): 11700–8. 37. Spiering D, Hodgson L. Dynamics of the Rho-family small GTPases in actin regulation and motility. Cell Adhes Migr. 2011;5(2):170–80. 38. Khosravi-Far R, Solski PA, Clark GJ, Kinch MS, Der CJ. Activation of Rac1, RhoA, and mitogen-activated protein kinases is required for Ras transformation. Mol Cell Biol. 1995;15(11): 6443–53. 39. Heasman SJ, Ridley AJ. Mammalian Rho GTPases: new insights into their functions from in vivo studies. Nat Rev Mol Cell Biol. 2008;9(9):690–701. 40. Rösel D, Brábek J, Tolde O, Mierke CT, Zitterbart DP, Raupach C, et al. Up-regulation of Rho/ROCK signaling in sarcoma cells drives invasion and increased generation of protrusive forces. Mol Cancer Res. 2008;6(9):1410–20. 41. Verghese ET, Drury R, Green CA, Holliday DL, Lu X, Nash C, et al. MiR-26b is down-regulated in carcinoma-associated fibro- blasts from ER-positive breast cancers leading to enhanced cell migration and invasion. J Pathol. 2013;231(3):388–99. 42. Berenjeno IM, Bustelo XR. Identification of the Rock-dependent transcriptome in rodent fibroblasts. Clin Transl Oncol. 2008;10(11): 726–38. 43. Sells MA, Pfaff A, Chernoff J. Temporal and spatial distribution of activated Pak1 in fibroblasts. J Cell Biol. 2000;151(7):1449–58. 44. Tse JC, Kalluri R. Mechanisms of metastasis: epithelial-to- mesenchymal transition and contribution of tumor microenviron- ment. J Cell Biochem. 2007;101(4):816–29. 45. Aboussekhra A. Role of cancer-associated fibroblasts in breast can- cer development and prognosis. Int J Dev Biol. 2011;55(7–9):841– 9. 46. Gaggioli C, Hooper S, Hidalgo-Carcedo C, et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9(12):1392–400. 47. Tchou J, Kossenkov AV, Chang L, Satija C, Herlyn M, Showe LC, et al. Human breast cancer associated fibroblasts exhibit subtype specific gene expression profiles. BMC Med Genomics. 2012;5:39. Tumor Biol.