ArticlePDF Available

Silencing of GM3 synthase suppresses lung metastasis of murine breast cancer cells

Authors:
  • Qingdao University of Science and Technology

Abstract and Figures

Gangliosides are sialic acid containing glycosphingolipids that are ubiquitously distributed on vertebrate plasma membranes. GM3, a precursor for most of the more complex ganglioside species, is synthesized by GM3 synthase. Although total ganglioside levels are significantly higher in breast tumor tissue than in normal mammary tissue, the roles played by gangliosides in breast cancer formation and metastasis are not clear. To investigate the roles of gangliosides in breast tumor development, GM3 synthase was silenced in the highly metastatic 4T1 cells and over-expressed in the non-metastatic 67NR cells. The behavior of breast cancer cells was examined in vitro using migration assay, invasion assay, and soft agar assay. Tumor formation and metastasis in vivo were examined using a well established mouse mammary tumor model. GM3 synthase silencing in 4T1 cells significantly inhibited cell migration, invasion and anchorage-independent growth in vitro, and lung metastasis in vivo. In addition, over-expression of GM3 synthase in nonmetastatic 67NR cells significantly induced cell migration and anchorage-independent growth. Further studies indicated that activation of the phosphoinositide-3 kinase/Akt pathway, and consequently inhibition of nuclear factor of activated T cell (NFAT)1 expression, could be the mechanism underlying the suppression of breast cancer migration/invasion induced by GM3 synthase silencing. Our findings indicate that GM3 synthase silencing suppressed lung metastasis in murine breast cancer cells. The molecular mechanism that underlies GM3 synthase mediated migration and invasion was inhibition of the phosphoinositide-3 kinase/Akt pathway. The findings suggest that GM3 synthase may be of value as a therapeutic target in breast cancer.
Content may be subject to copyright.
Open Access
Available online http://breast-cancer-research.com/content/10/1/R1
Page 1 of 12
(page number not for citation purposes)
Vol 10 No 1
Research article
Silencing of GM3 synthase suppresses lung metastasis of murine
breast cancer cells
Yuchao Gu*, Junhua Zhang*, Wenyi Mi, Jing Yang, Feng Han, Xinzhi Lu and Wengong Yu
Department of Molecular Biology, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao, China
* Contributed equally
Corresponding author: Wengong Yu, yuwg66@ouc.edu.cn
Received: 5 Aug 2007 Revisions requested: 18 Sep 2007 Revisions received: 24 Oct 2007 Accepted: 3 Jan 2008 Published: 3 Jan 2008
Breast Cancer Research 2008, 10:R1 (doi:10.1186/bcr1841)
This article is online at: http://breast-cancer-research.com/content/10/1/R1
© 2008 Gu et al.; licensee BioMed Central Ltd.
This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Background Gangliosides are sialic acid containing
glycosphingolipids that are ubiquitously distributed on
vertebrate plasma membranes. GM3, a precursor for most of the
more complex ganglioside species, is synthesized by GM3
synthase. Although total ganglioside levels are significantly
higher in breast tumor tissue than in normal mammary tissue, the
roles played by gangliosides in breast cancer formation and
metastasis are not clear.
Methods To investigate the roles of gangliosides in breast
tumor development, GM3 synthase was silenced in the highly
metastatic 4T1 cells and over-expressed in the non-metastatic
67NR cells. The behavior of breast cancer cells was examined
in vitro using migration assay, invasion assay, and soft agar
assay. Tumor formation and metastasis in vivo were examined
using a well established mouse mammary tumor model.
Results GM3 synthase silencing in 4T1 cells significantly
inhibited cell migration, invasion and anchorage-independent
growth in vitro, and lung metastasis in vivo. In addition, over-
expression of GM3 synthase in nonmetastatic 67NR cells
significantly induced cell migration and anchorage-independent
growth. Further studies indicated that activation of the
phosphoinositide-3 kinase/Akt pathway, and consequently
inhibition of nuclear factor of activated T cell (NFAT)1
expression, could be the mechanism underlying the suppression
of breast cancer migration/invasion induced by GM3 synthase
silencing.
Conclusion Our findings indicate that GM3 synthase silencing
suppressed lung metastasis in murine breast cancer cells. The
molecular mechanism that underlies GM3 synthase mediated
migration and invasion was inhibition of the phosphoinositide-3
kinase/Akt pathway. The findings suggest that GM3 synthase
may be of value as a therapeutic target in breast cancer.
Introduction
Gangliosides are sialic acid containing glycosphingolipids that
are ubiquitously distributed on vertebrate plasma membranes
[1]. They participate in the regulation of various cellular func-
tions, including cell proliferation, apoptosis, migration, and
invasion [2-4]. Numerous studies have demonstrated that
abnormal ganglioside expression is strongly associated with
the malignancy of cancer cells [5]. The ganglioside content is
upregulated in some metastasizing tumor cells, such as lym-
phoma cells, fibrosarcoma cells, and melanoma cells [6-8].
However, there is an inverse relation between metastasis
properties and ganglioside content in some types of tumors,
for instance bladder tumor and liver carcinoma [4,9,10]. Some
observations indicate that the effects of gangliosides on tumor
metastasis are dependent on the ganglioside species and/or
tumor-type specificity. For example, the effects of a given gan-
glioside, such as GM3, on metastatic potential differ between
tumor cell types [10-12]. On the other hand, the metastatic
potential of a given tumor cell type, such as melanoma cells,
may be enhanced or inhibited by different gangliosides
[3,11,13]. Hence, there is a need to define the involvement
and molecular mechanisms of gangliosides in metastasis of
each cancer type.
Among the various gangliosides, GM3 contains the simplest
ganglioside oligosaccharide and it serves as a precursor for
CMP = cytidine 5'-monophosphate; DME-10 = Dulbecco's modified Eagles medium supplemented with 10% fetal calf serum; EGFP = enhanced
green fluorescent protein; GAPDH = glyceraldehyde-3-phosphate dehydrogenase; GM3S = CMP-N-acetylneuraminic acid:lactosylceramide 2,3-sia-
lyltransferase; NFAT = nuclear factor of activated T cell; PI3K = phosphoinositide-3 kinase; PTEN = phosphatase and tensin homolog; RT-PCR =
reverse transcription polymerase chain reaction; siRNA = small interfering RNA.
Breast Cancer Research Vol 10 No 1 Gu et al.
Page 2 of 12
(page number not for citation purposes)
most of the more complex ganglioside species (for example,
GD3, GM2, GD2, and so on) [14]. GM3 is synthesized by
transfer of sialic acid from cytidine 5'-monophosphate (CMP)-
sialic acid to nonreduced terminal galactose residue of lacto-
sylceramide through the α-2,3-glycosyl bond, and the reaction
is catalyzed by GM3 synthase (CMP-N-acetylneuraminic
acid:lactosylceramide 2,3-sialyltransferase [EC 2.4.99.9]; also
known as ST3Gal V, GM3S [the term used in the present
report], and Siat9) [15]. Overexpression and suppression of
GM3S expression are approaches used to determine the
effects of endogenous gangliosides on the metastatic process
[2,16].
Total gangliosides in breast tumor tissues and sera from
breast cancer patients were significantly higher than in those
from healthy individuals [17,18]. GM3 and some other gan-
gliosides (such as GD3 and GM2) have been reported to be
potential targets for breast cancer immunotherapy [19-22],
chemotherapy [23], and radiotherapy [24]. Although ganglio-
side levels have been demonstrated to be elevated in breast
cancer, there are no reports in the literature regarding the roles
played by gangliosides in breast cancer metastasis.
In this study, in order to investigate the roles played by endog-
enous gangliosides in breast cancer formation and metastasis,
GM3S was silenced or over-expressed in different breast
tumor cell lines. The effects of GM3S expression on the malig-
nant properties of breast tumor cells and the molecular mech-
anisms of ganglioside-mediated migration and invasion were
evaluated.
Materials and methods
Cells and cell cultures
67NR and 4T1 (kindly provided by Dr Fred R Miller at Kar-
manos Cancer Institute) are mammary tumor cell lines charac-
terized by enhanced lung metastatic potential that were
derived from a single, spontaneously arising mouse mammary
tumor [25]. The nonmetastatic 67NR cells fail to leave the pri-
mary site, whereas the 4T1 cells can metastasize to lung, liver,
bone, and brain via the hematogenous route. They were cul-
tured in Dulbecco's modified Eagles medium supplemented
with 10% fetal calf serum (DME-10), 1 mmol/l mixed nones-
sential amino acids, and 2 mmol/l L-glutamine.
Plasmid construction
The dual promoter small interfering RNA (siRNA) expression
vector pMEHMpuro was constructed based on a self-inacti-
vating murine stem cell virus plasmid, namely pMSCVpuro.
First, the HindIII restriction site of pMSCVpuro was deleted by
digesting the plasmid with HindIII, blunting the 3' recessed
ends and re-ligating. The enhanced green fluorescent protein
(EGFP) gene was then inserted into the BglII/XhoI sites of
HindIII restriction site deleted pMSCVpuro vector; the result-
ing construct was named pMSCVpuro-EGFP. Then, the dual
promoter siRNA expression cassette was constructed as
described previously [26] and inserted into the XhoI/EcoRI-
digested pMSCVpuro-EGFP plasmid to form pMEHMpuro. In
order to generate siRNA vectors, three pairs of oligonucle-
otides, corresponding to different regions of murine GM3S
encoding gene, were annealed and inserted into BglII/HindIII
double digested pMEHMpuro: siGM3S1, 5'-AGC TTA AAA
AGT AAG GTT GAA CAG TGC GCC TTT TTA-3' and 5'-GAT
CTA AAA AGG CGC ACT GTT CAA CCT TAC TTT TTA-3';
siGM3S2, 5'-AGC TTA AAA AGA CTG CCT TCG ACA TCC
TTC TTT TTA-3' and 5'-GAT CTA AAA AGA AGG ATG TCG
AAG GCA GTC TTT TTA-3'; and siGM3S3, 5'-AGC TTA AAA
AGT GTG ACC ACA GAG ACC AAG TTT TTA-3' and 5'-
GAT CTA AAA ACT TGG TCT CTG TGG TCA CAC TTT
TTA-3'. This process yielded the vectors pMEHM-siGM3S1
(siGM3S1), pMEHM-siGM3S2 (siGM3S2), and pMEHM-
siGM3S3 (siGM3S3), respectively. In addition, annealed oli-
gonucleotides (5'-AGC TTA AAA AGT TCC GTA TGT TGC
ATC ACC TTT TTA-3' and 5'-GAT CTA AAA AGG TGA TGC
AAC ATA CGG AAC TTT TTA-3') that did not match any
known mouse gene was inserted into BglII/HindIII double
digested pMEHMpuro, yielding pMEHM-siMock. The full-
length GM3S encoding gene was amplified from the total
RNA of 4T1 cells using RT-PCR and inserted into XhoI/EcoRI
double digested pMSCVpuro, resulting in the GM3S expres-
sion vector pMSCV-GM3S. All of the resulting constructs
were confirmed by DNA sequencing.
DNA transfection and selection
4T1 cells were transfected with various GM3S-targeting RNA
interference and mock plasmids. 67NR cells were transfected
with GM3S and pMSCVpuro vectors. Transfection was car-
ried out using LipofectAMINE 2000 (Invitrogen, Carlsbad, CA,
USA), and the transfected cells were cultured in the medium
supplied with 4 μg/ml puromycin for 2 weeks. The obtained
cells were named 4T1-siMock, 4T1-siGM3S1, 4T1-siGM3S2,
4T1-siGM3S3, 67NR-Control and 67NR-GM3S.
RNA isolation and real-time RT-PCR
Total RNA was isolated from cells using Trizol (Invitrogen) and
reverse transcription was carried out using the High Capacity
cDNA Archive Kit (Applied Biosystems, Foster City, CA, USA).
Real-Time PCR was carried out using Power SYBR Green
Master PCR mix (Applied Biosystems, Warrington, UK) in trip-
licate, and the reactions were conducted on a 7500 real-time
PCR system (Applied Biosystems, Singapore). The relative
quantitative analysis was normalized to endogenous control
glyceraldehyde-3-phosphate dehydrogenase (GAPDH). The
mouse GM3S forward primer was 5'-GCT TCA AGC AAT
GGT AAA AAA TGA-3', and its reverse primer was 5'-TTC
TGC CAC TTG CTT CCA AA-3'. The mouse phosphatase
and tensin homolog (PTEN) forward primer was 5'-TGA AGA
CCA TAA CCC ACC ACA-3', and its reverse primer was 5'-
TCA TTA CAC CAG TCC GTC CCT-3'. Mouse GAPDH for-
ward primer was 5'-AAT TCA ACG GCA CAG TCA AGG-3',
Available online http://breast-cancer-research.com/content/10/1/R1
Page 3 of 12
(page number not for citation purposes)
and its reverse primer was 5'-TGT TAG TGG GGT CTC GCT
CC-3'.
Immunoblotting (IB) analysis
Cells were lysed in lysis buffer (50 mmol/l Tris-HCl [pH 7.4],
150 mmol/l NaCl, 1% NP40, 1 mmol/l EDTA, 1 mmol/l
Na3VO4, 10 mmol/l NaF) containing a protease inhibitor cock-
tail (Roche, Nutley, NJ, USA). Protein samples (50 μg) were
separated by 12% SDS-PAGE and transferred to Immobilon-
P membranes (Millipore, Bedford, MA, USA). Antibodies to
phosphorylated and total Akt (Cell Signaling, Beverly, MA,
USA), phosphorylated (Ser380/Thr382/383) and total PTEN
(Cell Signaling), NFAT1 (Santa Cruz, CA, USA), and GAPDH
(Santa Cruz) were used, with detection by ECL-detecting rea-
gent (Amersham Biosciences, Buckinghamshire, UK). Quanti-
fication of the blots was conducted using Image-Pro Plus
software (version 6.0; Media Cybernetics, Bethesda, MD,
USA).
Flow cytometry analysis
The flow cytometry procedure was as described previously
[27]. Briefly, cells were trypsinized and incubated with anti-
GM3 or anti-GD3 monoclonal antibodies (Seikagaku Corpora-
tion, Tokyo, Japan) and then labeled with rat anti-mouse IgM-
P-RE (Southern Biotechnology Associates, Birmingham, AL,
USA). A total of 1 × 104 labeled cells were analyzed using a
cytometer (Becton Dickinson, Franklin Lakes, NJ, USA).
Cell proliferation assay
Cells were seeded 1 × 104 per well in a 96-well plate. Cells
were allowed to grow for 24 hours. Then, 20 μl of 3-(4,5-
dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) (5
mg/ml) was added to each well. After 4 hours of incubation at
37°C, cells were lysed by addition of 200 μl dimethylsulfoxide.
Absorbance was measured at 570 nm using a Rainbow micro-
plate reader (Tecan, Groding/Salzburg, Austria).
Anchorage-independent cell growth
Cells (5 × 103 to 1 × 104) were suspended in 1 ml top agar
medium (DME-10 supplied with 0.4% agar), in the presence
or absence of phosphoinositide-3 kinase (PI3K)-specific inhib-
itor LY294002 (Merck, Nottingham, UK). The cell suspension
was then overlaid onto 1.5 ml bottom agar medium (DME-10
supplied with 0.8% agar) in six-well tissue culture plates in
triplicate. DME-10, with or without LY294002, was added to
the plates every 3 days as a feeder layer. On day 12, the
number of colonies was counted in six random fields at 40×
magnification.
Cell migration and invasion assay
Cell migration was assayed using Transwell chambers (6.5
mm; Corning, New York, USA) with 8 μmol/l pore membranes.
The lower chamber was filled with 600 μl NIH-3T3 condi-
tioned medium containing 20 μg/ml fibronectin (BD Bio-
sciences, Bedford, MA, USA) with or without 2 μmol/l
LY294002. Cells (5 × 104) were suspended with 100 μl
upper medium (Dulbecco's modified Eagles medium with 1%
fetal calf serum) and plated into the upper chamber with or
without 2 μmol/l LY294002. After 16 hours, the number of
cells appearing by crystal violet staining on the undersurface
of the polycarbonate membranes was scored visually in five
random fields at 100× magnification using a light microscope.
For invasion assays, the upper face of the membrane was cov-
ered with 70 μl Matrigel (1 mg/ml; BD Biosciences). The inva-
sion assay procedure was the same as for the migration assay,
except that the incubation time of the experiment was pro-
longed to 24 hours.
Primary tumor growth and lung metastases assay
These procedures were performed as described previously
[25,28] with minor modifications. Female BALB/c mice, aged
8 to 10 weeks, were used in the experiment. In brief, mice (six
to eight per group) were anesthetized with sodium pentobar-
bital (50 mg/kg body weight), and tumor cells (5 × 105) in 10
μl DME-10 were injected into the mammary gland. The weight
of the primary tumors and the number of metastatic nodules on
the lung surface were evaluated 30 days after the tumor cells
injection. The animals were housed and cared for in accord-
ance with the guidelines established by the National Science
Council of Republic of China. The lung tissues (six lungs per
group) from in vivo experiments described above were fixed in
10% neutral buffered formalin and embedded in paraffin, and
then thick sections (4.0 μm; three sections per lung tissue)
were cut and stained with hematoxylin and eosin.
Statistic analysis
All of the results were repeated in at least three independent
experiments and consistently yielded similar results. Data were
analyzed by the Student-Newman-Keuls test using the SPSS
11.0 software program (SPSS Inc., Chicago, IL, USA). P <
0.05 was considered statistically significant. Results are
expressed as mean ± standard error of the mean.
Results
GM3S is silenced or overexpressed in breast cancer cell
lines
We found that the expression of GM3S mRNA was greater in
the highly metastatic 4T1 tumor cells than in the nonmetastatic
67NR cells (Figure 1a). These observations led us to regard
GM3S to be a potential candidate for orchestrating metastasis
in breast cancer. To determine whether GM3S played an
important role in breast cancer malignancy, expression of
GM3S in the highly metastatic 4T1 cell line was silenced by
RNA interference. The expression of the GM3S siRNAs
siGM3S2 and siGM3S3, but not that of siGM3S1, drastically
reduced the expression of GM3S mRNA in 4T1 cells (Figure
1b). The results of flow cytometry analysis indicate that GM3S
knockdown significantly suppressed the expression of the
ganglioside GM3 (Figure 2a) and the more complex
Breast Cancer Research Vol 10 No 1 Gu et al.
Page 4 of 12
(page number not for citation purposes)
ganglioside GD3 (Figure 2b). On the other hand, GM3S was
over-expressed in 67NR cells. The expression of GM3S was
upregulated (Figure 1c) and the levels of expression of GM3
(Figure 2c) and GD3 (Figure 2d) gangliosides were enhanced.
These GM3S-reconstructed cells enabled us to identify the
effects of GM3S expression on the malignant properties of
breast cancer cells, and these cell lines also appeared to be
useful for analyzing the roles played by gangliosides in breast
cancer cells.
GM3S expression enhances the anchorage-independent
growth of breast cancer cells
To investigate whether GM3S expression affected cell prolif-
eration and anchorage-independent growth, MTT and soft
agar colony assays were conducted. Silencing of GM3S in
4T1 cells and over-expression of GM3S in 67NR cells did not
affect the proliferation (Figure 3a,b). The results were also ver-
ified by cell counting experiments (data not shown). Interest-
ingly, GM3S knockdown in 4T1 cells significantly reduced the
number of colonies and slightly decreased the colony size
(Figure 3c), whereas 67NR cells over-expressing GM3S
formed much larger colonies than did control cells, and the
number of colonies was slightly increased (Figure 3d). These
results indicated that endogenous GM3S was important for
maintaining the anchorage-independent growth of breast
tumor cells.
GM3S expression enhances migration and invasion of
breast cancer cells
To investigate the role played by GM3S in breast cancer cell
metastasis, we conducted migration and invasion assays in
vitro. As shown in Figure 4a and 4b, the migration and invasion
abilities were inhibited by silencing of GM3S in 4T1 cells.
Over-expression of GM3S in noninvasive 67NR cells
enhanced their ability to migrate (Figure 4c). However, over-
expression of GM3S was unable to transform noninvasive
67NR cells into invasive ones (data not shown). These find-
ings suggest that GM3S expression is essential but not suffi-
cient for breast cancer cell migration and invasion.
Figure 1
Analysis of GM3S expression of breast tumor cell lines by real-time RT-PCRAnalysis of GM3S expression of breast tumor cell lines by real-time RT-PCR. (a) GM3S mRNA expression was higher in metastatic 4T1 cells than in
noninvasive 67NR cells. (b, c) Relative GM3S mRNA expression in (b) GM3S-silenced 4T1 cells and in (c) GM3S over-expressing 67NR cells.
CMP, cytidine 5'-monophosphate; GM3S, CMP-N-acetylneuraminic acid:lactosylceramide 2,3-sialyltransferase.
Available online http://breast-cancer-research.com/content/10/1/R1
Page 5 of 12
(page number not for citation purposes)
Inhibition of GM3S expression suppresses lung
metastases of breast cancer cells
Based on the roles played by GM3S expression in migration/
invasion and anchorage-independent growth of breast cancer
cells described above, we next examined the effects of GM3S
silencing on tumor formation and metastasis of breast cancer
cells. Consistent with the results in vitro, GM3S silencing in
4T1 cells had no effect on primary tumor weight (Figure 5a)
but it dramatically reduced the number of visible metastatic
nodules on the lung surface of tumor-bearing mice (Figure
5b,c). Histologic analyses confirmed that the number of
micrometastatic lesions was significantly reduced in lungs of
mice carrying 4T1-siGM3S2 and 4T1-siGM3S3 tumors (Fig-
ure 5d).
GM3S promotes breast cancer cells migration and
invasion through the PI3K/Akt pathway
To investigate the molecular mechanism underlying GM3S-
mediated migration and invasion, some metastasis-associated
signaling pathways were assayed by immunoblotting. We
found that suppression of GM3S significantly enhanced the
phosphorylation of Akt at two key residues, namely Thr308
and Ser473 (Figure 6a). Further studies demonstrated that the
PI3K specific inhibitor LY294002 (2 μmol/l) restored Akt
phosphorylation to the level exhibited by 4T1-siMock cells
(Figure 6b) and partially compensated for the inhibition of cell
migration/invasion induced by the GM3S knockdown (Figure
6c).
PTEN is a major negative regulator of the PI3K/Akt signaling
pathway. In the present study, GM3S inhibition suppressed
the expression and phosphorylation of PTEN in 4T1 cells (Fig-
ure 7a,b). These findings suggest that the inhibition of PTEN
expression at least partially contributed to activation of the
PI3K/Akt pathway in 4T1 cells in which GM3S had been
knocked down.
We further investigated the downstream effectors of PI3K/Akt
pathway. GM3S knockdown inhibited NFAT1 expression (Fig-
ure 8a), and inhibition of the PI3K/Akt pathway induced
NFAT1 expression (Figure 8b). In conclusion, as illustrated in
Figure 9, GM3S knockdown suppressed PTEN expression
and resulted in activation of Akt signaling, which reduced the
expression of NFAT1 and inhibited cell migration and invasion.
PI3K/Akt pathway has no effect on anchorage-
independent growth of breast tumor cells
To test whether activation of the PI3K/Akt pathway was
responsible for the anchorage-independent growth affected
by GM3S knockdown in 4T1 cells, we inactivated the PI3K/
Akt pathway with LY294002. The colony forming abilities of
both GM3S-knockdown 4T1 cells and the mock transfected
cells were unaffected by inhibition of the PI3K/AKT pathway
(Figure 10), indicating that the anchorage-independent
growth caused by GM3S was not associated with the PI3K/
Akt pathway. Extra pathway(s) may exist to compensate for the
PI3K/Akt pathway with respect to anchorage-independent
survival of breast tumor cells.
Discussion
Breast cancer is the most common malignant disease of
women. In women with breast cancer, it is not the primary
tumor but its metastasis to distant sites that is the ultimate
cause of death. Improving our understanding of the molecular
mechanisms that underlie the metastatic process might
improve clinical management of the disease [29].
It has been demonstrated that levels of total gangliosides and
GM3 (N-glycolylneuraminic acid) in breast tumor tissue were
significantly higher than in normal tissues [18,24], and ganglio-
side content was fourfold higher in the invasive human breast
cancer cells MDA-MB-231 than in noninvasive MCF-7 cells
[29]. In the present study, the results of flow cytometry analy-
sis indicated that GM3 and GD3 gangliosides were
significantly higher in the highly metastatic 4T1 cells than in
the nonmetastatic 67NR cells. These studies suggested that
gangliosides might play roles in breast cancer formation and
metastasis.
Figure 2
Analysis of ganglioside content on cell surface by flow cytometryAnalysis of ganglioside content on cell surface by flow cytometry.
Expression levels of (a) GM3 and (b) GD3 gangliosides on the surface
of GM3S-silenced 4T1 cells using anti-GM3 and anti-GD3 monoclonal
antibody (shaded peaks: 4T1-siGM3S2; unshaded peaks: 4T1-
siMock). Expression levels of (c) GM3 and (d) GD3 gangliosides on
the surface of GM3S over-expressing 67NR cells using anti-GM3 and
anti-GD3 monoclonal antibody (shaded peaks: 67NR-Control;
unshaded peaks: 67NR-GM3S). CMP, cytidine 5'-monophosphate;
GM3S, CMP-N-acetylneuraminic acid:lactosylceramide 2,3-sialyltrans-
ferase.
Breast Cancer Research Vol 10 No 1 Gu et al.
Page 6 of 12
(page number not for citation purposes)
Metastasis is a rather complex process that occurs through a
series of steps that include invasion, intravasation, transport
through the circulatory system, arrest at a secondary site, and
the extravasation and growth in a secondary organ [30]. The
findings presented here suggest that GM3S expression is a
multistep modulator of breast cancer cell metastasis. First,
gain in motility and invasiveness is essential for most steps in
metastasis (the initial step, intravasation, and extravasation).
Our findings indicate that GM3S expression played important
roles in these steps by affecting ability to migrate/invade. In
addition, anchorage-independent growth is thought to be
among the fundamental properties of malignant cells [31,32].
In carcinomas, invasion, intravasation, and extravasation are
either deprived of matrix or exposed to foreign matrix
components. All of these events normally trigger apoptotic
processes [30]. In the present study we demonstrated that
GM3S significantly enhanced anchorage-independent growth
of breast tumor cells. Based on these findings, we hypothe-
sized that GM3S expression could affect almost all steps in
breast cancer metastasis.
Some other studies of the roles played by gangliosides in can-
cer progression support our hypothesis. GD3 and GD2 gan-
gliosides are upregulated in small cell lung cancer cells, and
these gangliosides were able to enhance proliferation and
invasion of small cell lung cancer [27]. In melanoma cells, GD3
has also been demonstrated to promote proliferation and inva-
sion [3]. In many types of tumor cells (for example, erythro-
leukemia, lymphoma, neuroblastoma, melanoma, glioblastoma,
and renal cell carcinoma cells), gangliosides may be shed into
the tumor cell microenvironment and there inhibit antitumor
immune responses [33-36]. It has also been demonstrated
that gangliosides shed from tumor cells can induce cell
migration and in vivo angiogenesis [37-40]. Ganglioside lev-
els in sera from breast cancer patients were also significantly
higher than in sera from healthy individuals [17]; this implies
Figure 3
Analysis of cell proliferation and anchorage-independent growthAnalysis of cell proliferation and anchorage-independent growth. Proliferation of (a) GM3S-silenced 4T1 cells and (b) GM3S over-expressing 67NR
cells was analyzed by MTT assay. Anchorage-independent growth of (c) GM3S-silenced 4T1 cells (5 × 103 cells/well) and (d) GM3S over-express-
ing 67NR cells were analyzed by soft agar assays (1 × 104 cells per well). *P < 0.05, **P < 0.01. CMP, cytidine 5'-monophosphate; GM3S, CMP-
N-acetylneuraminic acid:lactosylceramide 2,3-sialyltransferase.
Available online http://breast-cancer-research.com/content/10/1/R1
Page 7 of 12
(page number not for citation purposes)
that shed breast cancer gangliosides play a role in accelerat-
ing tumor progression.
Although gangliosides affect cell migration and invasion in
some types of tumor cells, only a few investigations have been
conducted that focused on the molecular mechanisms
involved. Hamamura and coworkers [3] demonstrated that
GD3 promoted melanoma cell invasion through activating
p130Cas and paxillin; however, invasion of the human kerati-
nocyte-derived SCC12 cell line was suppressed by GM3
ganglioside via inhibition of matrix metalloproteinase-9 activa-
tion, disrupting its association with integrin [41]. Some gan-
Figure 4
Migration and invasion assaysMigration and invasion assays. The relative (a) migration and (b) invasion assays for GM3S-silenced 4T1 cells. (c) The relative migration assays for
GM3S over-expressing 67NR cells. **P < 0.01. CMP, cytidine 5'-monophosphate; GM3S, CMP-N-acetylneuraminic acid:lactosylceramide 2,3-sia-
lyltransferase.
Figure 5
Tumor formation and lung metastases 30 days after tumor implantationTumor formation and lung metastases 30 days after tumor implantation. (a) Primary tumor weights. (b) The average numbers of lung metastatic nod-
ules. (c) Representative photos of the lungs. The arrows point to the metastatic nodules in lung. (d) Representative hematoxylin and eosin staining
sections of the lungs were photographed at 40× magnification. *P < 0.05. M, metastatic nodule; N, normal.
Breast Cancer Research Vol 10 No 1 Gu et al.
Page 8 of 12
(page number not for citation purposes)
Figure 6
Involvement of the PI3K/Akt in suppression of breast cancer migration/invasionInvolvement of the PI3K/Akt in suppression of breast cancer migration/invasion. Activation of the PI3K/Akt pathway is the causative mechanism for
the suppression of breast cancer migration/invasion induced by GM3S silencing. (a) Upper panels: the expression and activation of Akt detected by
immunoblotting with anti-Akt, anti-p-Akt Ser (473), and anti-p-Akt Thr (308) antibodies. Lower panels: quantitation of these blots after normalization
with the GAPDH blot. (b) Upper panels: PI3K inhibitor LY294002 restored the phosphorylation of Akt in 4T1-siGM3S2 cells. Lower panels: the
quantitation of the blots after normalization with the GAPDH blot. (c) The migration and invasion abilities of GM3S-silenced 4T1 cells were
enhanced by LY294002. *P < 0.05, **P < 0.01. CMP, cytidine 5'-monophosphate; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; GM3S,
CMP-N-acetylneuraminic acid:lactosylceramide 2,3-sialyltransferase; PI3K, phosphoinositide-3 kinase.
Available online http://breast-cancer-research.com/content/10/1/R1
Page 9 of 12
(page number not for citation purposes)
gliosides, such as GM3 and GM2, inhibited cell motility
facilitated by the tetraspanins CD9 and CD82 in some tumor
cell lines (for example, colorectal, haptotactic, and bladder
cancer cells) [42-45].
In this study we found that a novel mechanism (silencing of
GM3S) inhibited migration and invasion of breast cancer cells
through activation of the PI3K/Akt pathway. It is generally
accepted that the PI3K/Akt axis promotes tumorigenesis by
enhancing the survival capacity of cancer cells [46]. However,
some recently published evidence indicates that Akt can
inhibit breast cancer cell migration and invasion [47]. Previous
reports have well demonstrated that GM3 gangliosides can
inhibit Akt signaling, and this inhibition occurs mainly through
three processes. First, many studies have demonstrated that
GM3 gangliosides inhibit phosphorylation of the epidermal
growth factor receptor and result in inhibition of PI3K/Akt sig-
naling in varied cell types [48-51]. Second, GM3 and some
other gangliosides interact with integrin and consequently
inhibit the integrin/integrin-linked kinase/Akt signaling pathway
[52-54]. Finally, GM3 treatment markedly increases PTEN
expression, which results in inhibition of Akt signaling in colon
cancer cells [55]. Here, we demonstrated that GM3S silenc-
ing markedly suppressed PTEN expression and subsequently
activated the PI3K/Akt pathway in 4T1 cells. Furthermore, the
PI3K specific inhibitor LY294002 partially restored the cell
migration/invasion ability inhibited by GM3S knockdown in
4T1 cells. This finding clearly indicates that GM3S-associated
breast cancer migration and invasion occurred, at least partly,
through the PI3K/Akt pathway.
Previous studies revealed that the NFAT transcription factor
promoted invasion of breast cancer cells [56,57]. In this report
we demonstrated that GM3S silencing suppressed NFAT1
expression and that inhibition of the PI3K/Akt pathway signifi-
cantly enhanced expression of NFAT1. These findings sug-
gest that inhibition of NFAT1 expression via the activation of
PI3K/Akt signaling plays a causal role in the suppression of
migration and invasion that is induced by GM3S knockdown
in murine breast cancer cells.
Conclusion
This study demonstrates that GM3S silencing can suppress
cell migration, invasion, anchorage-independent growth, and
lung metastasis in murine breast cancer cells. The molecular
mechanism underlying the GM3S-mediated migration and
invasion was found to be inhibition of the PI3K/Akt pathway.
To our knowledge, this report is the first to elucidate the
involvement of GM3S and mechanisms by which it influences
the malignant properties of breast cancer cells.
Competing interests
The authors declare that they have no competing interests.
Authors' contributions
YG and JZ participated in designing the study, conducted cell
line transfection, immunoblotting analysis and animal experi-
ments, and drafted the manuscript. WM conducted the flow
cytometry analysis and immunoblotting analysis. JY partici-
pated in vector construction. FH and XL participated in the
Figure 7
Expression of PTEN was inhibited by GM3S silencingExpression of PTEN was inhibited by GM3S silencing. (a) Relative
PTEN expression in GM3S-knockdown 4T1 cells analyzed by real-time
RT-PCR. (b) Upper panels: expression and activation of PTEN
detected by immunoblotting. Lower panels: quantitation of these blots
after normalization with the GAPDH blot. *P < 0.05, **P < 0.01. CMP,
cytidine 5'-monophosphate; GAPDH, glyceraldehyde-3-phosphate
dehydrogenase; GM3S, CMP-N-acetylneuraminic acid:lactosylcera-
mide 2,3-sialyltransferase; p-PTEN, phosphor-PTEN Ser380/Thr382/
383; PTEN, phosphatase and tensin homolog.
Breast Cancer Research Vol 10 No 1 Gu et al.
Page 10 of 12
(page number not for citation purposes)
Figure 8
Expression of NFAT1 was inhibited through the PI3K/Akt pathway in GM3S-knockdown 4T1 cellsExpression of NFAT1 was inhibited through the PI3K/Akt pathway in GM3S-knockdown 4T1 cells. (a) Upper panels: expression of NFAT1 was
detected by immunoblotting. Lower panels: quantitation of these blots after normalization with the GAPDH blot. (b) Upper panels: expression of
NFAT1 was enhanced by the PI3K inhibitor LY294002. Lower panels: quantitation of these blots after normalization with the GAPDH blot. *P <
0.05, **P < 0.01. CMP, cytidine 5'-monophosphate; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; GM3S, CMP-N-acetylneuraminic
acid:lactosylceramide 2,3-sialyltransferase; NFAT, nuclear factor of activated T cell; PI3K, phosphoinositide-3 kinase.
Figure 9
Effects of GM3S on Akt-associated cell migration/invasion in breast cancer cellsEffects of GM3S on Akt-associated cell migration/invasion in breast
cancer cells. This schematic diagram shows that GM3S silencing
reduced expression of gangliosides and activated Akt signaling via inhi-
bition of PTEN expression (the major negative regulator of the PI3K/Akt
signaling). It also shows that activation of Akt signaling inhibits cell
migration and invasion, probably via downregulation of NFAT1. CMP,
cytidine 5'-monophosphate; GM3S, CMP-N-acetylneuraminic acid:lac-
tosylceramide 2,3-sialyltransferase; PI3K, phosphoinositide-3 kinase;
PTEN, phosphatase and tensin homolog.
Figure 10
Effects of PI3K/Akt signal on the anchorage-independent growth of breast cancer cellsEffects of PI3K/Akt signal on the anchorage-independent growth of
breast cancer cells. The anchorage-independent growth of both
GM3S-knockdown 4T1 cells and the mock transfected 4T1 cells (5 ×
103 cells/well) were not affected by the inhibition of the PI3K/AKT path-
way. CMP, cytidine 5'-monophosphate; GM3S, CMP-N-acetyl-
neuraminic acid:lactosylceramide 2,3-sialyltransferase; PI3K,
phosphoinositide-3 kinase.
Available online http://breast-cancer-research.com/content/10/1/R1
Page 11 of 12
(page number not for citation purposes)
design of the study and conducted the statistical analysis. WY
conceived of the study, participated in its design and coordi-
nation, and helped to draft the manuscript. All authors read
and approved the final manuscript.
Acknowledgements
We thank Fred R Miller for providing the murine breast tumor cell lines,
Meiyu Geng and Jing Li for technical assistance, and Guanpin Yang for
critical review of the manuscript. This work was supported by the
National Basic Research Program of China (973 Program;
2003CB716402).
References
1. Huwiler A, Kolter T, Pfeilschifter J, Sandhoff K: Physiology and
pathophysiology of sphingolipid metabolism and signaling.
Biochim Biophys Acta 2000, 1485:63-99.
2. Watanabe R, Ohyama C, Aoki H, Takahashi T, Satoh M, Saito S,
Hoshi S, Ishii A, Saito M, Arai Y: Ganglioside G(M3) overexpres-
sion induces apoptosis and reduces malignant potential in
murine bladder cancer. Cancer Res 2002, 62:3850-3854.
3. Hamamura K, Furukawa K, Hayashi T, Hattori T, Nakano J,
Nakashima H, Okuda T, Mizutani H, Hattori H, Ueda M, et al.: Gan-
glioside GD3 promotes cell growth and invasion through
p130Cas and paxillin in malignant melanoma cells. Proc Natl
Acad Sci USA 2005, 102:11041-11046.
4. Kawamura S, Ohyama C, Watanabe R, Satoh M, Saito S, Hoshi S,
Gasa S, Orikasa S: Glycolipid composition in bladder tumor: a
crucial role of GM3 ganglioside in tumor invasion. Int J Cancer
2001, 94:343-347.
5. Dyatlovitskaya EV, Kandyba AG: Sphingolipids in tumor metas-
tases and angiogenesis. Biochemistry (Mosc) 2006,
71:347-353.
6. Ruggieri S, Mugnai G, Mannini A, Calorini L, Fallani A, Barletta E,
Mannori G, Cecconi O: Lipid characteristics in metastatic cells.
Clin Exp Metastasis 1999, 17:271-276.
7. Laferte S, Fukuda MN, Fukuda M, Dell A, Dennis JW: Glycosphin-
golipids of lectin-resistant mutants of the highly metastatic
mouse tumor cell line, MDAY-D2. Cancer Res 1987,
47:150-159.
8. Saha S, Mohanty KC: Correlation of gangliosides GM2 and
GM3 with metastatic potential to lungs of mouse B16
melanoma. J Exp Clin Cancer Res 2003, 22:125-134.
9. Kloppel TM, Morre DJ, Jacobsen LB: Ganglioside patterns of
metastatic and non-metastatic transplantable hepatocellular
carcinomas of the rat. J Supramol Struct 1979, 11:485-492.
10. Satoh M, Ito A, Nojiri H, Handa K, Numahata K, Ohyama C, Saito
S, Hoshi S, Hakomori SI: Enhanced GM3 expression, associ-
ated with decreased invasiveness, is induced by brefeldin A in
bladder cancer cells. Int J Oncol 2001, 19:723-731.
11. Deng W, Li R, Guerrera M, Liu Y, Ladisch S: Transfection of glu-
cosylceramide synthase antisense inhibits mouse melanoma
formation. Glycobiology 2002, 12:145-152.
12. Sawada M, Moriya S, Saito S, Shineha R, Satomi S, Yamori T, Tsu-
ruo T, Kannagi R, Miyagi T: Reduced sialidase expression in
highly metastatic variants of mouse colon adenocarcinoma 26
and retardation of their metastatic ability by sialidase
overexpression. Int J Cancer 2002, 97:180-185.
13. Thomas CP, Buronfosse A, Portoukalian J, Fertil B: The ganglio-
sides as a possible molecular coupling factor between the
proportion of radiosensitive cells in vitro and the metastatic
potential in vivo within a human melanoma cell line. Br J
Cancer 1997, 75:639-649.
14. van Echten G, Sandhoff K: Ganglioside metabolism. Enzymol-
ogy, topology, and regulation. J Biol Chem 1993,
268:5341-5344.
15. Fukumoto S, Miyazaki H, Goto G, Urano T, Furukawa K, Furukawa
K: Expression cloning of mouse cDNA of CMP-NeuAc:Lactos-
ylceramide alpha2,3-sialyltransferase, an enzyme that initiates
the synthesis of gangliosides. J Biol Chem 1999,
274:9271-9276.
16. Uemura S, Kabayama K, Noguchi M, Igarashi Y, Inokuchi J: Sialyla-
tion and sulfation of lactosylceramide distinctly regulate
anchorage-independent growth, apoptosis, and gene expres-
sion in 3LL Lewis lung carcinoma cells. Glycobiology 2003,
13:207-216.
17. Wiesner DA, Sweeley CC: Circulating gangliosides of breast-
cancer patients. Int J Cancer 1995, 60:294-299.
18. Marquina G, Waki H, Fernandez LE, Kon K, Carr A, Valiente O,
Perez R, Ando S: Gangliosides expressed in human breast
cancer. Cancer Res 1996, 56:5165-5171.
19. Fukumoto H, Nishio K, Ohta S, Hanai N, Fukuoka K, Ohe Y, Sugi-
hara K, Kodama T, Saijo N: Effect of a chimeric anti-ganglioside
GM2 antibody on ganglioside GM2-expressing human solid
tumors in vivo. Int J Cancer 1999, 82:759-764.
20. Carr A, Rodriguez E, Arango Mdel C, Camacho R, Osorio M, Gabri
M, Carrillo G, Valdes Z, Bebelagua Y, Perez R, et al.: Immuno-
therapy of advanced breast cancer with a heterophilic ganglio-
side (NeuGcGM3) cancer vaccine. J Clin Oncol 2003,
21:1015-1021.
21. Bada A, Casaco Parada A, Arteaga M, Martinez J, Leon A, Santana
E, H ernandez O , Orphee R, Go nzalez A , Mesa C, et al.: Toxicity of
a GM3 cancer vaccine in Macaca fascicularis monkey: a 12-
month study. Hum Exp Toxicol 2002, 21:263-267.
22. Guthmann MD, Castro MA, Cinat G, Venier C, Koliren L, Bitton RJ,
Vazquez AM, Fainboim L: Cellular and humoral immune
response to N-Glycolyl-GM3 elicited by prolonged immuno-
therapy with an anti-idiotypic vaccine in high-risk and meta-
static breast cancer patients. J Immunother (1997) 2006,
29:215-223.
23. Basu S, Ma R, Boyle PJ, Mikulla B, Bradley M, Smith B, Basu M,
Banerjee S: Apoptosis of human carcinoma cells in the pres-
ence of potential anti-cancer drugs: III. Treatment of Colo-205
and SKBR3 cells with: cisplatin, tamoxifen, melphalan, betu-
linic acid, L-PDMP, L-PPMP, and GD3 ganglioside. Glycoconj J
2004, 20:563-577.
24. Oliva JP, Valdes Z, Casaco A, Pimentel G, Gonzalez J, Alvarez I,
Osorio M, Velazco M, Figueroa M, Ortiz R, et al.: Clinical evi-
dences of GM3 (NeuGc) ganglioside expression in human
breast cancer using the 14F7 monoclonal antibody labelled
with (99 m)Tc. Breast Cancer Res Treat 2006, 96:115-121.
25. Aslakson CJ, Miller FR: Selective events in the metastatic proc-
ess defined by analysis of the sequential dissemination of
subpopulations of a mouse mammary tumor. Cancer Res
1992, 52:1399-1405.
26. Zheng L, Liu J, Batalov S, Zhou D, Orth A, Ding S, Schultz PG: An
approach to genomewide screens of expressed small interfer-
ing RNAs in mammalian cells. Proc Natl Acad Sci USA 2004,
101:135-140.
27. Yoshida S, Fukumoto S, Kawaguchi H, Sato S, Ueda R, Furukawa
K: Ganglioside G(D2) in small cell lung cancer cell lines:
enhancement of cell proliferation and mediation of apoptosis.
Cancer Res 2001, 61:4244-4252.
28. Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come
C, Savagner P, Gitelman I, Richardson A, Weinberg RA: Twist, a
master regulator of morphogenesis, plays an essential role in
tumor metastasis. Cell 2004, 117:927-939.
29. Weigelt B, Peterse JL, van 't Veer LJ: Breast cancer metastasis:
markers and models. Nat Rev Cancer 2005, 5:591-602.
30. Mehlen P, Puisieux A: Metastasis: a question of life or death.
Nat Rev Cancer 2006, 6:449-458.
31. Freedman VH, Shin SI: Cellular tumorigenicity in nude mice:
correlation with cell growth in semi-solid medium. Cell 1974,
3:355-359.
32. Cifone MA: In vitro growth characteristics associated with
benign and metastatic variants of tumor cells. Cancer Metas-
tasis Rev 1982, 1:335-347.
33. Ladisch S, Gillard B, Wong C, Ulsh L: Shedding and immu-
noregulatory activity of YAC-1 lymphoma cell gangliosides.
Cancer Res 1983, 43:3808-3813.
34. Ladisch S, Kitada S, Hays EF: Gangliosides shed by tumor cells
enhance tumor formation in mice. J Clin Invest 1987,
79:1879-1882.
35. Alfonso M, Lanne B, Ifversen P, Vazquez AM, Perez R, Portoukalian
J, Zeuthen J: Antigen-specific primary immune response of
human B-lymphocytes after in vitro immunization with GM3
ganglioside. Hum Antibodies Hybridomas 1995, 6:102-112.
36. Potapenko M, Shurin GV, de Leon J: Gangliosides as
immunomodulators. Adv Exp Med Biol 2007, 601:195-203.
Breast Cancer Research Vol 10 No 1 Gu et al.
Page 12 of 12
(page number not for citation purposes)
37. Birkle S, Zeng G, Gao L, Yu RK, Aubry J: Role of tumor-associ-
ated gangliosides in cancer progression. Biochimie 2003,
85:455-463.
38. Kim CW, Lee HM, Lee TH, Kang C, Kleinman HK, Gho YS: Extra-
cellular membrane vesicles from tumor cells promote angio-
genesis via sphingomyelin. Cancer Res 2002, 62:6312-6317.
39. Lang Z, Guerrera M, Li R, Ladisch S: Ganglioside GD1a
enhances VEGF-induced endothelial cell proliferation and
migration. Biochem Biophys Res Commun 2001,
282:1031-1037.
40. Manfredi MG, Lim S, Claffey KP, Seyfried TN: Gangliosides influ-
ence angiogenesis in an experimental mouse brain tumor.
Cancer Res 1999, 59:5392-5397.
41. Wang XQ, Sun P, Paller AS: Ganglioside GM3 inhibits matrix
metalloproteinase-9 activation and disrupts its association
with integrin. J Biol Chem 2003, 278:25591-25599.
42. Todeschini AR, Dos Santos JN, Handa K, Hakomori SI: Ganglio-
side GM2-tetraspanin CD82 complex inhibits met and its
cross-talk with integrins, providing a basis for control of cell
motility through glycosynapse. J Biol Chem 2007,
282:8123-8133.
43. Mitsuzuka K, Handa K, Satoh M, Arai Y, Hakomori S: A specific
microdomain ('glycosynapse 3') controls phenotypic conver-
sion and reversion of bladder cancer cells through GM3-medi-
ated interaction of alpha3beta1 integrin with CD9. J Biol Chem
2005, 280:35545-35553.
44. Kawakami Y, Kawakami K, Steelant WF, Ono M, Baek RC, Handa
K, Withers DA, Hakomori S: Tetraspanin CD9 is a 'proteolipid',
and its interaction with alpha 3 integrin in microdomain is pro-
moted by GM3 ganglioside, leading to inhibition of laminin-5-
dependent cell motility. J Biol Chem 2002, 277:34349-34358.
45. Ono M, Handa K, Sonnino S, Withers DA, Nagai H, Hakomori S:
GM3 ganglioside inhibits CD9-facilitated haptotactic cell
motility: coexpression of GM3 and CD9 is essential in the
downregulation of tumor cell motility and malignancy. Bio-
chemistry 2001, 40:6414-6421.
46. Vivanco I, Sawyers CL: The phosphatidylinositol 3-Kinase AKT
pathway in human cancer. Nat Rev Cancer 2002, 2:489-501.
47. Toker A, Yoeli-Lerner M: Akt signaling and cancer: surviving but
not moving on. Cancer Res 2006, 66:3963-3966.
48. Wada T, Hata K, Yamaguchi K, Shiozaki K, Koseki K, Moriya S, Miy-
agi T: A crucial role of plasma membrane-associated sialidase
in the survival of human cancer cells. Oncogene 2007,
26:2483-2490.
49. Sottocornola E, Misasi R, Mattei V, Ciarlo L, Gradini R, Garofalo T,
Berra B, Colombo I, Sorice M: Role of gangliosides in the asso-
ciation of ErbB2 with lipid rafts in mammary epithelial HC11
cells. Febs J 2006, 273:1821-1830.
50. Wang XQ, Sun P, Paller AS: Ganglioside GM3 blocks the acti-
vation of epidermal growth factor receptor induced by integrin
at specific tyrosine sites. J Biol Chem 2003, 278:48770-48778.
51. Mirkin BL, Clark SH, Zhang C: Inhibition of human neuroblast-
oma cell proliferation and EGF receptor phosphorylation by
gangliosides GM1, GM3, GD1A and GT1B. Cell Prolif 2002,
35:105-115.
52. Sun P, Wang XQ, Lopatka K, Bangash S, Paller AS: Ganglioside
loss promotes survival primarily by activating integrin-linked
kinase/Akt without phosphoinositide 3-OH kinase signaling. J
Invest Dermatol 2002, 119:107-117.
53. Wang XQ, Sun P, Paller AS: Inhibition of integrin-linked kinase/
protein kinase B/Akt signaling: mechanism for ganglioside-
induced apoptosis. J Biol Chem 2001, 276:44504-44511.
54 . T oledo MS, Su zuki E , Hand a K, Ha komor i S: Effect of ganglioside
and tetraspanins in microdomains on interaction of integrins
with fibroblast growth factor receptor. J Biol Chem 2005,
280:16227-16234.
55. Choi HJ, Chung TW, Kang SK, Lee YC, Ko JH, Kim JG, Kim CH:
Ganglioside GM3 modulates tumor suppressor PTEN-medi-
ated cell cycle progression – transcriptional induction of
p21(WAF1) and p27(kip1) by inhibition of PI-3K/AKT pathway.
Glycobiology 2006, 16:573-583.
56. Chen M, O'Connor KL: Integrin alpha6beta4 promotes expres-
sion of autotaxin/ENPP2 autocrine motility factor in breast
carcinoma cells. Oncogene 2005, 24:5125-5130.
57. Yiu GK, Toker A: NFAT induces breast cancer cell invasion by
promoting the induction of cyclooxygenase-2. J Biol Chem
2006, 281:12210-12217.
... Suppression in the PTEN activity subsequently promoted Akt phosphorylation and reduced expression of NFAT1 (Nuclear factor of activated T cells). Reduced activity of transcription factor NFAT1 explains the molecular events behind the reduced tumorigenesis in GM3 synthase knocked down cells (Gu et al., 2008). The role of GD3 in regulating metastasis and invasion was extensively investigated in cancer (Liu et al., 2018). ...
Article
Full-text available
Gangliosides are glycosphingolipids with prevalence in nervous tissue and their involvement in certain neuronal diseases have been widely known. Interestingly, many recent studies highlighted their importance in the development and progression of various cancers through orchestration of multiple attributes of tumorigenesis, i.e., promoting migration, invasion, escaping the host immune system, and influencing other cancer hallmarks. Therefore, the multidimensional role of gangliosides in different cancers has established them as potential cancer targets. However, the tremendous structural complexity and functional heterogeneity are the major challenges in ganglioside research. Moreover, despite numerous immunotherapeutic attempts to target different gangliosides, it has failed to yield consistent results in clinical trials owing to their poor immunogenicity, a broad range of cross-reactivity, severe side effects, lack of uniform expression as well as heterogeneity. The recent identification of selective O-acetylated ganglioside expression in cancer tissues, but not in normal tissues, has strengthened their potential as a better and specific target for treating cancer patients. It was further supported by reduced cross-reactivity and side effects in clinical trials, although poor immunogenicity remains a major concern. Therefore, in addition to characterization and identification of the biological importance of O-acetylated gangliosides, their specific and efficient targeting in cancer through engineered antibodies is an emerging area of glycobiology research. This review highlights the modulatory effect of select gangliosides on different hallmarks of cancer and presents the overall development of ganglioside targeted immunotherapies along with recent progress. Here, we have also discussed its potential for future modifications aimed towards improvement in ganglioside-based cancer therapies.
... Gangliosides of glycosphingolipid were reported to be involved in the process of EMT to induce cancer cells lose their epithelial features to gain mesenchymal characteristics [29]. ST3GAL5 is key enzyme for the synthesis of most complex gangliosides, Gu et al. observed that silencing ST3GAL5 could reduce the migration, invasion and anchorage-independent growth of murine breast cancer cell (4T1) in vitro as well as lung metastasis in vivo [30]. While other pathways including nucleotide excision repair, base excision repair, cell cycle, DNA replication, basal transcription factors, P53 signaling pathway, mismatch repair and non-homologous end joining (NHEJ) pathway, were elevated in the low-score group. ...
Preprint
Full-text available
Aim To establish a novel lipid metabolism-related (LMR) prognostic model for gastric cancer (GC) and explore the potential mechanism of natriuretic peptide receptor-3 (NPR3) in the process of GC metastasis. Method LMR genes were identified from the Gene Set Enrichment Analysis (GSEA) and mRNA expression profile were download from The Cancer Genome Atlas (TCGA) database. We used the R package “limma” to obtain the LMR differentially expressed genes (DEGs) between GC and adjacent tissues. Consensus clustering was then performed based on the expression of LMR DEGs using the R package “ConsensusClusterPlus”. We adopted the weighted correlation network analysis (WGCNA) to obtain the best module related to metabolic subtypes. A prognostic model based on 6 LMR genes (FBLN7, NPY1R, VTN, NPR3, EPHB3 and AUH) was established through least absolute shrinkage and selection operator (LASSO) penalized Cox regression analysis based on progression-free interval (PFI). In addition, we verified the NPR3 expression in several GC cell lines by quantitative Real-time PCR and Western Blotting, and explored the effect of NPR3 on GC cell migration using the wound healing assay and transwell test. We performed immunohistochemistry (IHC), H&E and collagen staining on 42 GC tissues to clarify the clinical significance of NPR3 in gastric cancer. Results 2 LMR subtypes (C1 and C2) were confirmed using consensus clustering of 153 LMR-DEGs. Compared with C1, C2 was associated with a worse prognosis, especially in terms of PFI (HR: 1.64, 95%CI: 1.15–2.33, P < 0.001). Using WGCNA and univariate cox regression, 558 genes were screened out to build and optimize the model. Finally, a novel predictive formula system based on 6 genes (FBLN7, NPY1R, VTN, NPR3, EPHB3 and AUH) were constructed and the time-dependent area under the receiver operating characteristic curve (time-ROC, 1/3/5 years) was 0.79/0.77/0.71 and 0.73/0.68/0.64 in the training set (N = 214) and validation set (N = 141), respectively. In addition, we found that NPR3 over-expression could promote the migration of GC cells. And its expression was higher in tumor tissues than in paracancerous tissues and patients with high expression of NPR3 were more likely have the vascular invasion (OR: 5.056, 95%CI: 1.159–22.060, p = 0.031) and higher stage (OR: 5.100, 95%CI: 1.336–19.470, p = 0.017). Conclusion We established a novel LMR prognostic model predicting the prognosis of gastric cancer, and found that NPR3 can promote the tumor migration and vascular invasion of gastric cancer.
... GM3 synthase induces anchorage-independent growth and promotes migration and invasion of cancer cells (Gu et al., 2008), whereas suppression of GM3 synthase has been reported to inhibit cancer cell migration and invasion. DHC is a precursor of GM3 and DHC is also increased in Exon 20 mutant by twofold compared with the parental cell line (Fig. 3), indicating increased GM3 synthase in the mutant. ...
Article
Lipids play crucial biological roles in health and disease, including in cancers. The phosphatidylinositol 3-kinase (PI3K) signaling pathway is a pivotal promoter of cell growth and proliferation in various types of cancer. The somatic mutations in PIK3CA, the gene coding for the catalytic subunit p110α of PI3K, are frequently present in cancer cells, including breast cancer. Although the most prominent mutants, represented by single amino acid substitutions in the helical domain in exon 9 (E545K) and the kinase domain in exon 20 (H1047R) are known to cause a gain of PI3K function, activate AKT signaling and induce oncogenic transformation, the effect of these mutations on cellular lipid profiles has not been studied. We carried out untargeted lipidomics using liquid chromatography-tandem mass spectrometry to detect the lipid alterations in mammary gland epithelial MCF10A cells with isogenic knockin of these mutations. A total of 536 species of lipids were analyzed. We found that the levels of monosialogangliosides, signaling molecules known to enhance cell motility through PI3K/AKT pathway, were significantly higher in both mutants. In addition, triglycerides and ceramides, lipid molecules known to be involved in promoting lipid droplet production, cancer cell migration and invasion, were increased, whereas lysophosphatidylcholines and phosphatidylcholines that are known to inhibit cancer cell motility were decreased in both mutants. Our results provide novel insights into a potential link between altered lipid profile and carcinogenesis caused by the PIK3CA hotspot mutations. In addition, we suggest untargeted lipidomics offers prospects for precision/personalized medicine by unpacking new molecular substrates of cancer biology.
... The ST3GAL5 gene regulating GM3 synthesis was identified as essential with the MB model and GM3 was found abundantly in MB by Chang et al. [37]. Even though GM3 showed apoptotic features in many cancer types, suppressing GM3 synthase led to anti-cancer effects in MB [38,39]. Particularly, in the Group 4 type of MB, the ST3GAL5 gene was highly active in comparison to the control group and other subtypes [40]. ...
Article
Full-text available
Medulloblastoma (MB), occurring in the cerebellum, is the most common childhood brain tumor. Because conventional methods decline life quality and endanger children with detrimental side effects, computer models are needed to imitate the characteristics of cancer cells and uncover effective therapeutic targets with minimum toxic effects on healthy cells. In this study, metabolic changes specific to MB were captured by the genome-scale metabolic brain model integrated with transcriptome data. To determine the roles of sphingolipid metabolism in proliferation and metastasis in the cancer cell, 79 reactions were incorporated into the MB model. The pathways employed by MB without a carbon source and the link between metastasis and the Warburg effect were examined in detail. To reveal therapeutic targets for MB, biomass-coupled reactions, the essential genes/gene products, and the antimetabolites, which might deplete the use of metabolites in cells by triggering competitive inhibition, were determined. As a result, interfering with the enzymes associated with fatty acid synthesis (FAs) and the mevalonate pathway in cholesterol synthesis, suppressing cardiolipin production, and tumor-supporting sphingolipid metabolites might be effective therapeutic approaches for MB. Moreover, decreasing the activity of succinate synthesis and GABA-catalyzing enzymes concurrently might be a promising strategy for metastatic MB.
... These suggest caution when using ST6GalNAc2 as a possible biomarker for predicting metastases in ER-negative breast cancers [90]. Moreover, sialic acid-containing GM3 has been reported to reduce phosphoinositide-3 kinase/serine/threonine protein kinase B (PI3K/Akt) signaling to increase breast and colon cancer migration and invasion via inhibiting EGFR phosphorylation, upregulating phosphatase and tensin homolog (PTEN) expression, and interacting with integrins [92]. On the contrary, downregulation of ST3Gal4 is associated with malignant progression [60,93] in part by activating PI3K/Akt pathway in renal cell carcinoma [60]. ...
Article
Full-text available
Sialylation is an enzymatic process that covalently attaches sialic acids to glycoproteins and glycolipids and terminates them by creating sialic acid-containing glycans (sialoglycans). Sialoglycans, usually located in the outmost layers of cells, play crucial biological roles, notably in tumor transformation, growth, metastasis, and immune evasion. Thus, a deeper comprehension of sialylation in cancer will help to facilitate the development of innovative cancer therapies. Cancer sialylation-related articles have consistently increased over the last four years. The primary subjects of these studies are sialylation, cancer, immunotherapy, and metastasis. Tumor cells activate endothelial cells and metastasize to distant organs in part by the interactions of abnormally sialylated integrins with selectins. Furthermore, cancer sialylation masks tumor antigenic epitopes and induces an immunosuppressive environment, allowing cancer cells to escape immune monitoring. Cytotoxic T lymphocytes develop different recognition epitopes for glycosylated and nonglycosylated peptides. Therefore, targeting tumor-derived sialoglycans is a promising approach to cancer treatments for limiting the dissemination of tumor cells, revealing immunogenic tumor antigens, and boosting anti-cancer immunity. Exploring the exact tumor sialoglycans may facilitate the identification of new glycan targets, paving the way for the development of customized cancer treatments.
... Some GMs, such as GM1 and GM3, exhibit predominantly anti-proliferative effects in different cancers, such as colon, bladder, gliomas, and leukemias, by modulating cell cycle progression through PTEN and p53 stimulation, mitigating PDGF-mediated MAPK activation, and promoting apoptosis through BAX and BAD upregulation [235,[258][259][260][261]. Furthermore, GM3 and GM1 expression have consistently been reported to weaken metastatic potential in gastrointestinal and ovarian carcinomas by inhibiting cell motility and MMP-9mediated migration [262][263][264]. Interestingly, GM3 is also able to induce MMP-2-mediated invasion of melanoma, and positively correlates with Ki-67 status in breast cancer, which is puzzling when considering that GM3 synthase silencing has been shown to decrease breast cancer metastases in vivo via NFAT1 inhibition [265][266][267][268][269]. Similarly, GM3 enhances sensitivity to EGFR-TK inhibitors in lung cancer while promoting resistance to classic chemotherapeutic agents in this same neoplasm, suggesting it has divergent roles within specific cellular pathways, such as apoptosis [270,271]. ...
Article
Full-text available
Simple Summary Cancer is an incredibly smart disease, so much so that it can entirely reprogram our cells’ metabolism to ensure its survival and dissemination. Lymphomas are a particularly interesting group of cancers because they are very diverse and exhibit complex pathogenic mechanisms that can make them clinically challenging. Many of these mechanisms involve the dysregulation of a biologically relevant family of lipids known as sphingolipids. These molecules are involved in virtually every cellular process and therefore studying them in this context is crucial. However, sphingolipid biochemistry is intricate and the synergistic and antagonic effects of different sphingolipid species with one another has long puzzled scientists. This duality is also observed in cancer, but research specifically focusing on lymphomas is limited. Could novel biomarkers and therapies against lymphomas be hiding within this pathway? Abstract Lymphomas are a highly heterogeneous group of hematological neoplasms. Given their ethiopathogenic complexity, their classification and management can become difficult tasks; therefore, new approaches are continuously being sought. Metabolic reprogramming at the lipid level is a hot topic in cancer research, and sphingolipidomics has gained particular focus in this area due to the bioactive nature of molecules such as sphingoid bases, sphingosine-1-phosphate, ceramides, sphingomyelin, cerebrosides, globosides, and gangliosides. Sphingolipid metabolism has become especially exciting because they are involved in virtually every cellular process through an extremely intricate metabolic web; in fact, no two sphingolipids share the same fate. Unsurprisingly, a disruption at this level is a recurrent mechanism in lymphomagenesis, dissemination, and chemoresistance, which means potential biomarkers and therapeutical targets might be hiding within these pathways. Many comprehensive reviews describing their role in cancer exist, but because most research has been conducted in solid malignancies, evidence in lymphomagenesis is somewhat limited. In this review, we summarize key aspects of sphingolipid biochemistry and discuss their known impact in cancer biology, with a particular focus on lymphomas and possible therapeutical strategies against them.
... Ganglioside GM3 is widely distributed in essentially all types of animal cells, and overexpressed in several types of human cancers, such as melanomas, lung, and brain cancers [106,107]. Its detection at the surface of tumor cells is an important factor in determining the metastatic phenotype [108,109]. It has been demonstrated that GM3 density may affect antigenicity. ...
Article
Full-text available
Simple Summary Aberrant glycosylation is a common feature of many cancers, and it plays crucial roles in tumor development and biology. Cancer progression can be regulated by several physiopathological processes controlled by glycosylation, such as cell–cell adhesion, cell–matrix interaction, epithelial-to-mesenchymal transition, tumor proliferation, invasion, and metastasis. Different mechanisms of aberrant glycosylation lead to the formation of tumor-associated carbohydrate antigens (TACAs), which are suitable for selective cancer targeting, as well as novel antitumor immunotherapy approaches. This review summarizes the strategies developed in cancer immunotherapy targeting TACAs, analyzing molecular and cellular mechanisms and state-of-the-art methods in clinical oncology. Abstract Aberrant glycosylation is a hallmark of cancer and can lead to changes that influence tumor behavior. Glycans can serve as a source of novel clinical biomarker developments, providing a set of specific targets for therapeutic intervention. Different mechanisms of aberrant glycosylation lead to the formation of tumor-associated carbohydrate antigens (TACAs) suitable for selective cancer-targeting therapy. The best characterized TACAs are truncated O-glycans (Tn, TF, and sialyl-Tn antigens), gangliosides (GD2, GD3, GM2, GM3, fucosyl-GM1), globo-serie glycans (Globo-H, SSEA-3, SSEA-4), Lewis antigens, and polysialic acid. In this review, we analyze strategies for cancer immunotherapy targeting TACAs, including different antibody developments, the production of vaccines, and the generation of CAR-T cells. Some approaches have been approved for clinical use, such as anti-GD2 antibodies. Moreover, in terms of the antitumor mechanisms against different TACAs, we show results of selected clinical trials, considering the horizons that have opened up as a result of recent developments in technologies used for cancer control.
Article
Aberrant glycosylation is a key mechanism employed by cancer cells to evade immune surveillance, induce angiogenesis and metastasis, among other hallmarks of cancer. Sialic acids, distinctive terminal glycan structures located on glycoproteins or glycolipids, are prominently upregulated across various tumor types, including colorectal cancer (CRC). Sialylated glycans modulate anti-tumor immune responses through their interactions with Siglecs, a family of glycan-binding receptors with specificity for sialic acid-containing glycoconjugates, often resulting in immunosuppression. In this paper, we investigated the immunomodulatory function of ST3Gal5, a sialyltransferase that catalyzes the addition of α2-3 sialic acids to glycosphingolipids, since lower expression of ST3Gal5 is associated with better survival of CRC patients. We employed CRISPR/Cas9 to knock out the ST3Gal5 gene in two murine CRC cell lines MC38 and CT26. Glycomics analysis confirmed the removal of sialic acids on glycolipids, with no discernible impact on glycoprotein sialylation. Although knocking out ST3Gal5 in both cell lines did not affect in vivo tumor growth, we observed enhanced levels of regulatory T cells in CT26 tumors lacking ST3Gal5. Moreover, we demonstrate that the absence of ST3Gal5 affected size and blood vessel density only in MC38 tumors. In summary, we ascertain that sialylation of glycosphingolipids has a limited influence on the anti-tumor immune response in CRC, despite detecting alterations in the tumor microenvironment, possibly due to a shift in ganglioside abundance.
Article
Full-text available
Background: Malignant cells may arise from dedifferentiation of mature cells and acquire features of the progenitor cells. Definitive endoderm from which liver is derived, expresses glycosphingolipids such as SSEA3, Globo H, and SSEA4. Herein, we evaluated the potential prognosis value of the three glycosphingolipids and biological functions of SSEA3 in hepatocellular carcinoma (HCC). Methods: The expression of SSEA3, Globo H, and SSEA4 in tumor tissues obtained from 382 patients with resectable HCC was examined by immunohistochemistry staining. Epithelial mesenchymal transition (EMT) and their related genes were analyzed by transwell assay and qRT-PCR, respectively. Results: Kaplan Meier survival analysis showed significantly shorter relapse-free survival (RFS) for those with higher expression of SSEA3 (P < 0.001), Globo H (P < 0.001), and SSEA4 (P = 0.005) and worse overall survival (OS) for those with high expression of either SSEA3 (P < 0.001) or SSEA4 (P = 0.01). Furthermore, multivariable Cox regression analysis identified the SSEA3 as an independent predictor for RFS (HR: 2.68, 95% CI: 1.93-3.72, P < 0.001) and OS (HR: 2.99, 95% CI: 1.81-4.96, P < 0.001) in HCC. Additionally, SSEA3-ceramide enhanced the EMT of HCC cells, as reflected by its ability to increase migration, invasion and upregulate the expression of CDH2, vimentin, fibronectin, and MMP2, along with ZEB1. Moreover, ZEB1 silencing abrogated the EMT-enhancing effects of SSEA3-ceramide. Conclusions: Higher expression of SSEA3 was an independent predictor for RFS and OS in HCC and promoted EMT of HCC via upregulation of ZEB1.
Article
Full-text available
Gangliosides are sialylated glycolipids, mainly present at the cell surface membrane, involved in a variety of cellular signaling events. During malignant transformation, the composition of these glycosphingolipids is altered, leading to structural and functional changes, which are often negatively correlated to patient survival. Cancer cells have the ability to shed gangliosides into the tumor microenvironment, where they have a strong impact on anti-tumor immunity and promote tumor progression. Since most ganglioside species show prominent immunosuppressive activities, they might be considered checkpoint molecules released to counteract ongoing immunosurveillance. In this review, we highlight the current state-of-the-art on the ganglioside-mediated immunomodulation, specified for the different immune cells and individual gangliosides. In addition, we address the dual role that certain gangliosides play in the tumor microenvironment. Even though some ganglioside species have been more extensively studied than others, they are proven to contribute to the defense mechanisms of the tumor and should be regarded as promising therapeutic targets for inclusion in future immunotherapy regimens.
Article
Full-text available
Expression cloning of a cDNA for the α2,3-sialyltransferase (GM3 synthase) (EC 2.4.99.-) gene was performed using a GM3-lacking mouse fibroblast line L cell and anti-GM3 monoclonal antibody. Plasmids from a cDNA library generated with poly(A)+ RNA of a mouse fibrosarcoma line CMS5j and pdl3027 (polyoma T antigen) were co-transfected into L cells. The isolated cDNA clone pM3T-7 predicted a type II membrane protein with 13 amino acids of cytoplasmic domain, 17 amino acids of transmembrane region, and a large catalytic domain with 329 amino acids. Introduction of the cDNA clone into L cells resulted in the neo-synthesis of GM3 and high activity of α2,3-sialyltransferase. Among glycosphingolipids, only lactosylceramide showed significant activity as an acceptor, indicating that this gene product is a sialyltransferase specific for the synthesis of GM3. An amino acid sequence deduced from the cloned cDNA showed the typical sialyl motif with common features among α2,3-sialyltransferases. Among various mouse tissues, brain, liver, and testis showed relatively high expression of a 2.3-kilobase mRNA, whereas all tissues, more or less, expressed this gene.
Article
Full-text available
The role of tumor cell membrane gangliosides in tumor formation was probed using a series of cloned murine AKR lymphoma cell lines. Tumor formation was directly related to high expression and shedding of membrane gangliosides. In vivo, as little as 1 pmol of purified total gangliosides of highly tumorigenic cells, injected intradermally with poorly tumorigenic cells (which lacked and did not shed gangliosides), markedly increased the tumorigenicity of these cells in syngeneic normal mice. Thus, gangliosides shed by tumor cells are a previously unrecognized, extremely potent enhancer of tumor formation in vivo.
Article
Full-text available
YAC-1 lymphoma cells, both when cultured in vitro and when passaged in ascites form in vivo, synthesize gangliosides (means of 22.1 and 14.7 nmol lipid-bound sialic acid isolated per 10(8) cells, respectively) with potent inhibitory effects on mitogen- and antigen-induced lymphoproliferation: 10 to 30 nmol highly purified YAC-1 gangliosides/ml caused greater than 90% inhibition of proliferative responses of murine lymphocytes to concanavalin A, lysozyme (a soluble specific antigen), and allogeneic cells (mixed-lymphocyte response). Measureable quantities of these gangliosides were shed by the tumor cells in vitro and also were recovered from the ascites fluid in vivo. Furthermore, the gangliosides isolated from ascites fluid (mean of 15.3 nmol/ml) had inhibitory activity of a magnitude similar to that of the gangliosides isolated from the tumor cells. Therefore, significant inhibition of normal lymphoproliferative responses by tumor-derived gangliosides occurred at ganglioside concentrations which are actually present in the fluid surrounding the tumor cells in vivo. These results support the hypothesis that shedding of gangliosides may serve to protect tumor cells from host immune destruction.
Article
In previous investigations, we correlated levels of sialic acid, gangliosides, and ganglioside glycosyltransferases with tumorigenesis over a 24-week continuum of growth of hepatocellular neoplasms of the rat induced by the carcinogen N-2-fluorenylacetamide. However, metastatic tumors developed only rarely and were not analyzed. To investigate surface changes associated with metastasis, well-differentiated and poorly differentiated hepatocellular carcinomas were transplanted to syngeneic recipient rats. From those, several metastatic and nonmetastatic isolates were obtained and compared. Both total and ganglioside sialic acid amounts in transplantable hepatomas were elevated above control liver values but were significantly lower for metastatic lines than for nonmetastatic lines. The nonmetastatic lines were characterized by ganglioside patterns depleted in the precursor ganglioside GM3 (sialic acid-galactose-glucose-ceramide) and elevated in the products of the monosialoganglioside pathway. In contrast, metastatic isolates exhibited a restoration of GM3 and nearer normal amounts of other gangliosides. The findings point to differences in sialic acid-containing glycolipids, comparing metastatic and nonmetastatic hepatocellular carcinomas, and further extend the concept that ganglioside alterations do not cause tumorigenesis but are the end result of a cascade of events which apparently continue beyond the onset of metastasis.
Article
To identify selective steps in metastasis, those that eliminate nonmetastatic tumor cells more efficiently than metastatic cells, we have evaluated the sequential dissemination of tumor cells from a mammary fatpad, using both metastatic (4T1 and 66cl4) and nonmetastatic (67NR, 168FARN, and 4TO7) subpopulations of a single mouse mammary tumor. Each of these variant subpopulations is resistant to one or more selective drugs so they could be quantitatively identified by colony formation in selective media. We found that the 2 metastatic cell lines metastasized by different routes and that the nonmetastatic tumor cell lines failed at different points in dissemination. Line 67NR did not leave the primary site; clonogenic tumor cells were not detected in the nodes, blood, or lungs during the experiment (7 weeks). Tumor line 168FARN disseminated from the primary tumor because clonogenic cells were cultured from the draining lymph nodes throughout the experiment. However, dissemination essentially stopped in the node as cells were rarely isolated from blood, lungs, or lives. Whether 168FARN cells failed to reach these tissues or were killed very rapidly after traversing the lymph node is unknown. Line 4TO7 cells disseminated via the blood and were consistently recovered from lungs by day 19 but failed to proliferate. This panel of 5 subpopulations thus identifies different points of selective failure in tumor cell dissemination and should be valuable in the assessment of antimetastatic therapies.
Article
Neutral and acidic glycolipids in MDAY-D2, a highly metastatic murine tumor cell line, were examined and compared with glycolipids of MDW4 and D33W25-1, two lectin-resistant mutants of MDAY-D2 from distinct genetic complementation classes. D33W25-1 remained highly metastatic while MDW4 cells were found to be nonmetastatic (Dennis, J. W., Donaghue, T., Florian, M., and Kerbel, R. S., Nature (Lond.), 292: 242-245, 1981 and Dennis, J. W. et al., Cancer Res., 46: 4594-4600, 1986). Glycolipid structures were identified by fast-atom bombardment mass spectrometry, methylation analysis, exoglycosidase treatment, and immunostaining. The metastatic MDAY-D2 was found to contain GM3, GM2, IV3GalNAc-GM1b, and high levels of GM1a, GM1b, and GD1a. MDW4 showed a 3-fold decrease in total ganglioside content compared to MDAY-D2 and a corresponding increase in the precursor, glucosylceramide. MDW4 was deficient in GM1 and accumulated GM2 and NeuNG-GM2, indicating a lack of gangliosides having NeuNAc alpha 2-3Gal beta 1-3 terminal sequence. Neosynthesis of GD3 was also observed in MDW4. The metastatic mutant D33W25-1 had a similar pattern of gangliosides as that found in MDAY-D2 cells with N-glycolyl rather than N-acetyl neuraminic acid. These results suggest that the metastatic property of these cell lines may be related to the level of ganglioside, and that the substitution of N-glycolyl for N-acetyl neuraminic acid does not reduce metastatic capacity.
Article
Cultured cells derived from either normal or malignant tissues of several species have been tested by injection into the immune-deficient nude mouse in order to determine the cellular properties which are associated with tumorigenicity in vivo. Results show that one in vitro property consistently correlated with neoplastic growth in nude mice is the ability of the cell to form spherical colonies in a semi-solid growth medium such as methyl cellulose suspension. Cellular tumorigenicity is not determined solely by the malignancy of the tissue of origin, since cells derived from nonmalignant tissues become tumorigenic when they are no longer anchorage dependent for growth. In addition, acquisition of infinite growth potential in heteropioid cell lines is not in itself sufficient to confer tumorigenic capacity on the cells. These results suggest that the degree of cell growth in methyl cellulose is a useful parameter in vitro for predicting tumorigenicity in the animal, and also demonstrate the potential usefulness of the nude mouse for analysis of cellular malignancy irrespective of the tissue or species of origin.
Article
Tumors show a wide range of growth rates in vivo. In general, nonmetastatic (benign) tumors appear to grow slowly, while metastasizing (malignant) tumors proliferate rapidly and contain a large number of dividing cells. In order to analyze if there is a direct relationship between metastatic potential and growth rate (without the influence of host factors), in vitro analyses are necessary. In vitro studies that compared monolayer doubling times and in vivo tumorigenicity rarely demonstrated a correlation between the two parameters. Differences in monolayer growth rates were observed among tumors, but the differences were unrelated to in vivo growth. In contrast, some tumor systems demonstrated a correlation between in vivo growth and/or malignant potential and growth properties observed during anchorage independent growth. Evidence is presented to show that in particular tumor systems, an analysis of colony size and colony morphology during growth in suspension can be used to study the relationship between malignant potential and growth properties.