Detection of Neu5Gc Gangliosides in NSC Lung Carcinoma

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

Detection of N-glycolyated gangliosides

in non-small-cell lung cancer using GMR8


monoclonal antibody
Nobuyoshi Hayashi,1 Hirofumi Chiba,1,7 Koji Kuronuma,1 Shinji Go,2 Yoshihiro Hasegawa,1 Motoko Takahashi,3
Shinsei Gasa,4 Atsushi Watanabe,5 Tadashi Hasegawa,6 Yoshio Kuroki,2 Jinichi Inokuchi2 and Hiroki Takahashi1
1
Third Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo; 2Division of Glycopathology, Institute of Molecular
Biomembranes and Glycobiology, Tohoku Pharmaceutical University, Miyagi, Sendai; Departments of 3Biochemistry, 4Chemistry, 5Second Department of
Surgery, 6Clinical Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan

(Received March 12, 2012 ⁄ Revised September 5, 2012 ⁄ Accepted September 13, 2012 ⁄ Accepted manuscript online September 24, 2012 ⁄ Article first published online October 30, 2012)

Gangliosides are glycosphingolipids found on the cell surface. kinase.(4) GM3 is a ganglioside that binds to the extracellular
They act as recognition molecules or signal modulators and regu- domain of EGFR and inhibits its dimerization without inhibit-
late cell proliferation and differentiation. N-glycolylneuraminic ing ligand binding.(5) Kawashima et al. described the signifi-
acid (NeuGc)-containing gangliosides have been detected in some cance of carbohydrate–carbohydrate interactions between
neoplasms in humans, although they are usually absent in nor- N-glycans with N-acetylglucosamine (GlcNAc) termini on
mal human tissues. Our aim was to evaluate the presence of EGFR and oligosaccharides on GM3.(6)
NeuGc-containing gangliosides including GM3 (NeuGc) and assess NeuAc and N-glycolylneuraminic acid (NeuGc) are the most
their relationship with the prognosis of non-small-cell lung can- common types of sialic acids found in vertebrates. The only
cer (NSCLC). NeuGc-containing ganglioside expression in NSCLC structural difference between them is a single oxygen atom at
tissues was analyzed immunohistochemically using the mouse the C-5 position of NeuGc, which is catalyzed by the cytidine
monoclonal antibody GMR8, which is specific for gangliosides monophospho-N-acetylneuramic acid hydroxylase (CMAH).(7)
with NeuGc alpha 2,3Gal-terminal structures. On the basis of In general, NeuGc is abundant in mammals other than humans,
NeuGc-containing ganglioside expression, we performed survival in whom CMAH is inactivated due to a mutation in the
analysis. We also investigated the differences in the effects of CMAH gene.(8,9) However, the composition and production of
GM3 (N-acetylneuraminic acid [NeuAc]) and GM3 (NeuGc) on inhi- gangliosides is altered in many tumor types.(10,11) It is known
bition of epidermal growth factor receptor (EGFR) tyrosine kinase that anti-NeuGc-containing ganglioside antibodies recognize
in A431 cells. As a result, the presence of NeuGc-containing gan- tumor tissues in breast cancer, Wilms tumor, melanoma and
gliosides was evident in 86 of 93 (93.5%) NSCLC samples. The neuroblastoma.(11–14)
NSCLC patients with high NeuGc-containing ganglioside expres- A mouse GMR8 monoclonal antibody (mAb) was generated
sion had a low overall survival rate and a significantly low pro- by immunizing mice with purified GM3 (NeuGc).(15) GMR8
gression-free survival rate. In the in vitro study, the inhibitory mAb binds specifically to gangliosides with NeuGc alpha
effect of GM3 on EGFR tyrosine kinase in A431 cells after expo- 2,3Gal-terminal structures, such as GM3 (NeuGc), IV3NeuGc
sure to GM3 (NeuGc) was lower than that after exposure to GM3 alpha-Gg4Cer, IV3NeuGc alpha-nLc4Cer, V3NeuGc alpha-
(NeuAc). In conclusion, NeuGc-containing gangliosides including Gb5Cer and GD1a (NeuGc, NeuGc). Furthermore, it does not
GM3 (NeuGc) are widely expressed in NSCLC, and NeuGc-contain- recognize any other ganglioside with internal NeuGc alpha
ing ganglioside expression is associated with patient survival. 2,3Gal-terminal structures, such as GM2 (NeuGc) and GM1
The difference in the effects of GM3 (NeuGc) and GM3 (NeuAc) (NeuGc), corresponding gangliosides with NeuAc alpha
on the inhibition of EGFR tyrosine kinase might contribute to 2,3Gal-terminal structures and neutral glycolipids. Thus, the
improvement in the prognosis of NSCLC patients. (Cancer Sci epitope structures recognized by mAb are exclusively NeuGc
2013; 104: 43–47) alpha 2,3Gal-terminal structures.
In the present study, we evaluated the presence of NeuGc-
containing gangliosides in NSCLC by immunohistochemistry

L ung cancer is the most frequently diagnosed major cancer


worldwide.(1) The World Health Organization classification
of lung cancer identifies squamous cell carcinoma, adenocarci-
using GMR8 mAb and performed survival analysis based on
NeuGc-containing ganglioside expression.

noma, large cell carcinoma and small cell carcinoma as its


four major types. These tumors are commonly divided into Materials and Methods
small-cell lung cancer and non-small-cell lung cancer
Patients. Ninety-three NSCLC patients who underwent pri-
(NSCLC) depending on differences in their biology and treat-
ment. The low rate of cure for NSCLC can be attributed to the mary tumor resection at Sapporo Medical University between
high metastasis rate at diagnosis and the lack of effective treat- July 2003 and October 2006 were included in the present
ments to cure such a metastatic disease. study. Informed consent was obtained from all patients. The
Gangliosides are ubiquitous membrane-associated glycos- Human Ethics Review Committees of Sapporo Medical Uni-
phingolipids containing at least one sialic acid residue. They versity approved the study protocol (approval no. 219-3).
Immunohistochemistry. We determined N-glycolyated gangli-
act as recognition molecules or signal modulators and regulate
cell proliferation and differentiation.(2,3) Delay in cell growth oside expression in NSCLC tissues using mouse GMR8
of the human epidermoid carcinoma cell line A431 is caused
by GM3 (N-acetylneuraminic acid [NeuAc])-mediated inhi-
bition of epidermal growth factor receptor (EGFR) tyrosine 7
To whom correspondence should be addressed.
E-mail: hchiba@sapmed.ac.jp

doi: 10.1111/cas.12027 Cancer Sci | January 2013 | vol. 104 | no. 1 | 43–47
© 2012 Japanese Cancer Association
mAb.(13) Paraffin-embedded tumor tissues were cut into 5-lm- Protect tubes (Qiagen, Duesseldorf, Germany) immediately
thick sections and placed on glass slides. These sections were after pulmonary resections of RNA stabilization, and total
deparaffinized and pretreated by boiling the slides in citrate buf- RNA was extracted from specimens using the single-step
fer (pH 6.8) for 10 min. The sections were then immersed in method of RNA isolation by acid guanidinum thiocyanate-phe-
0.3% hydrogen peroxide for 30 min to block endogenous tissue nol-chloroform extraction.(17) Reverse transcription of total
peroxidase activity. After blocking non-specific proteins by RNA was performed to generate the first strand cDNA. For
incubation in diluted normal serum for 30 min, sections were detection of cDNA of human CMP-sialic acid hydroxylase, we
incubated with mouse GMR8 mAb (1:200) at 4°C overnight. performed reverse–transcription polymerase chain reaction
Subsequently, sections were biotinylated with anti-mouse IgM (RT-PCR) assays using the primer set based on sequences of
for 30 min, followed by incubation with avidin DH–biotinylat- CMP-sialic acid; 5′-CCAGTCAGGAAGTC-3′ (forward primer,
ed horseradish peroxidase complex (Vector Labs, Burlingame, the lesion upper 92-bp deletion) and 5′-GGTTGGAGGAC-
CA, USA) for 30 min at room temperature. Finally, the peroxi- CAG-3′ (reverse, the lesion lower 92-bp deletion primer).(8)
dase reaction was developed using diaminobenzidine as the Amplification was initiated by preincubation for 15 min at 94°
chromogen. Slides were counterstained with hematoxylin. C for the initial activation, followed by 30 cycles of denatur-
Next, we evaluated NeuGc-containing ganglioside expres- ation for 1 min at 94°C, and primer annealing for 1 min at 54°
sion. We defined GMR8-positive cells as cells with complete or C and elongation for 2 min at 72°C using Hotstar Taq Master
partly stained cytoplasmic membranes. Three 0.25 9 0.25 mm Mix Kit (Qiagen). A 20 lL aliquot of each reaction mixture
squares were randomly marked in the tumor area. The mean was electrophoresed through 1% agarose gel and stained using
percentage of GMR8-positive cells among the total number of ethidium bromide for visualization. DNA fragments were
tumor cells was determined for each NSCLC sample. The eval- extracted with agarose gel using the QIAquick Gel Extraction
uation was performed by two independent observers. Kit (Qiagen). DNA sequencing was performed with the use of
TLC immunostaining of gangliosides. Gangliosides were dye terminator chemistry and a Capillary ABI 3100 sequencer
extracted from approximately 10 mm blocks of formalin-fixed (Applied Biosystems, Tokyo, Japan).
tumor and normal tissues using a chloroform-methanol (2:1) Statistical analysis. The statistical analysis with categorical
solution. Gangliosides extracted from tissues were immuno- data was done using the Pearson’s Chi-squared test. Kaplan–
stained on a high-performance thin-layer chromatography plate Meier plots and log-rank analysis were used to determine the
(E, Merck, Darmstadt, Germany) to determine the ganglioside significance of differences in progression-free and overall surviv-
fraction. The solvent system used for developing chromato- als. P < 0.05 (two-tailed) was considered statistically significant.
grams was chloroform-methanol-water (60:35:8, v/v/v). Gan- Progression-free survival was the time between diagnosis and
gliosides were visualized with orcinol stain. The plate was air the date of recurrence or remaining alive, while overall survival
dried, blocked with phosphate-buffered saline (PBS) containing was the time between diagnosis and the date of death or the date
3% bovine serum albumin for 1 h, and incubated with mouse when patients were last known to be alive. In the survival analy-
GMR8 mAb and mouse anti-GM3 (NeuAC) mAb M2590 sis, only data before 31 December 2010 was considered.
(Cosmo Bio Co. Ltd, Tokyo, Japan) for 1 day at 4°C. Bound
primary antibodies were visualized using the Vectastain kit Results
(Vector Labs) according to the manufacturer’s instructions.
In vitro GM3 (NeuGc)- and GM3 (NeuAc)-mediated inhibition of Patient characteristics. To investigate NeuGc-containing gan-
EGFR phosphorylation. Human ovarial epidermoid carcinoma glioside expression in NSCLC tissues, we examined expression
A431 cells known to overexpress EGFR in the membrane levels in surgically resected tumor tissues of 93 NSCLC
were used in the present study. A431 cells were cultured in patients treated at Sapporo Medical University Hospital and
Dulbecco’s modified Eagle medium (DMEM; Sigma-Aldrich- retrospectively reviewed their clinical records. Patient demo-
Japan; number D5796, Tokyo, Japan) containing 10% heat-inac- graphics are shown in Table 1.
tivated fetal bovine serum (Japan Bio Serum; number 82225, N-glycolyated ganglioside expression in NSCLC tissues. To
Fukuyama, Japan) and 100 lg/mL sodium pyruvate at 37°C in confirm that lipid components were recognized by GMR8
5% CO2. An in vitro EGFR phosphorylation assay was per- mAb, immunostaining was performed after removal of lipid
formed as described by Kawashima et al.(6) and Zhou et al.(16) components from lung tissues using a chloroform-methanol
Aliquots of solution containing 250 lM GM3 (NeuAc) (Calbio- (2:1) solution. As shown in Figure 1(C,D) staining was lost
chem; number 345733, Darmstadt, Germany) (average molecu- after removal of the lipid components. Therefore, the
lar weight = 1280) and 250 lM GM3 (NeuGc) (Alex number molecules immunohistochemically detected by GMR8 were
302-019-MC01) (average molecular weight = 1264) in a chlo- gangliosides.
roform-methanol solution were dried under vacuum. Ethanol
was added and the sample was dried again. Serum-free DMEM Table 1. Demographic data of the non-small-cell lung cancer
(400 lL) was added to completely dried GM3 (NeuAc) or GM3 patients
(NeuGc), mixed using a vortex mixer for 3 h and sonicated for
No. patients evaluable 93
30 min at 4°C. A431 cells were cultured separately in 12-well
Mean age (range) (years) 66.3 (40–81)
plates in DMEM containing 10% fetal bovine serum and starved
Male/female (n) 58/35
in serum-free DMEM for 24 h before confluence, then washed
Tumor histology (n)
with serum-free DMEM and incubated in serum-free DMEM
Adenocarcinoma 60
containing GM3 (NeuAc) or GM3 (NeuGc) for 3 h at 37°C.
Squamous cell carcinoma 22
Cells were washed with PBS to induce phosphorylation, incu-
Large cell carcinoma 6
bated for 1 h in 400 lL of EGF (10 ng/mL) or 400 lL of EGF
Other* 5
(100 ng/mL) at 4°C, washed and lysed in 100 lL of lysis buffer
Pathological stage
(1% SDS, 5 mM EDTA, 5 mM EGTA, 1 mM Na3VO4) for
I 59
30 min at 4°C. The cell lysate was subjected to SDS-PAGE fol-
II 10
lowed by western blotting using anti-EGFR and phosphotyro-
III 20
sine (PY20) antibodies.
IV 4
Sequencing of cDNA of CMP-NeuAc hydroxylase in NSCLC
tissues. Lung cancer tissues were stored in RNAlater Tissue *Pleomorphic carcinoma (n = 3); adenosquamous carcinoma (n = 2).

44 doi: 10.1111/cas.12027
© 2012 Japanese Cancer Association
A B

C D

Fig. 1. N-glycolyated ganglioside expression in non-small-cell lung cancer (NSCLC). (A) Positive N-glycolyated ganglioside expression with GMR8
staining in NSCLC. (B) Negative N-glycolyated ganglioside expression with GMR8 staining in NSCLC. (C) Positive N-glycolyated ganglioside expres-
sion with GMR8 staining in NSCLC (not the same as the sample shown in Fig. 1A). (D) After removal of lipid components by incubating with chlo-
roform–methanol solution (2:1), negative N-glycolyated ganglioside expression with GMR8 staining in NSCLC (same sample as shown in Fig. 1C).

Positive and negative GMR8 expression in NSCLC tissues is GMR8-positive cells was not significantly correlated with the
shown in Figure 1(A,B). No NeuGc-containing ganglioside pathological stage of NSCLC.
expression was detected in normal lung tissues. If more than Immunoblotting of gangliosides on TLC plates. Using tumor
5% of the tumor cells were GMR8 positive, then NeuGc-con- tissues, we analyzed the composition of gangliosides recog-
taining ganglioside expression was considered positive. Neu- nized by GMR8 mAb. Using four NSCLC tissues that were
Gc-containing gangliosides were expressed in 86 of 93 positive for NeuGc-containing ganglioside expression in the
(93.5%) NSCLC samples. Details of the percentage of GMR8- immunohistochemistry study, we performed TLC immuno-
positive cells are shown in Table 2. In addition, Kaplan–Meier staining of gangliosides extracted from both normal and cancer
analysis was conducted to determine whether a difference in tissues. N-glycolyated gangliosides (GD1a [NeuGc] and GM3
the percentage of GMR8-positive cells was associated with [NeuGc]) were detected only in cancer tissues using GMR8
prognosis. Pathological stage IV patients whose prognosis was mAb (Fig. 3A), whereas GM3 (NeuAc) was found in both nor-
obviously poor were excluded from analysis. For survival anal- mal and cancer tissues using mouse anti-GM3 (NeuAC) mAb
ysis, the group with  80% GMR8-positive cells was sepa- (M2590 mAb) (Fig. 3B).
rated from the group with <80% GMR8-positive cells (Fig. 2). In vitro GM3 (NeuGc) and GM3 (NeuAc)-mediated inhibition of
We found that progression-free survival was significantly low EGFR phosphorylation. We investigated the difference in the
in the group with a higher percentage of GMR8-positive cells effects of GM3 (NeuAc) and GM3 (NeuGc) on inhibition of
(P < 0.05) and the overall survival was low, although this was EGFR tyrosine kinase. The density ratio of anti-PY20 to anti-
not statistically significant (P = 0.156). The percentage of EGFR bands is shown in the bar graph (Fig. 4). As shown in
Figure 4, the inhibitory effect of GM3 (NeuAc) and GM3
(NeuGc) on EGFR tyrosine kinase induced by 10 or 100 ng
EGF in A431 cell membrane fractions was reduced after expo-
Table 2. N-glycolyated ganglioside expression in tumor cells of sure to GM3 (NeuGC) was lower than that after exposure to
non-small-cell lung cancer GM3 (NeuAC).
cDNA sequences of CMAH in NSCLC tissues. In three cancer
No. cases (%)
OS PFS tissues that were positive for NeuGc-containing ganglioside
(months) (months) expression, we analyzed the cDNA sequence of the CMAH
gene and found a 92-bp deletion in it and a frameshift muta-
High N-glycolyated ganglioside expression
tion in the stop codon (TGA), as previously reported by Chou
80~100% positive cells in 15 (17.0) 52.4 47.5
et al.(8)
tumor cells
Low N-glycolyated ganglioside expression
60~79% positive cells in 15 (17.0) 55.9 56.4 Discussion
tumor cells
In the present study, we demonstrated that N-glycolyated gan-
40~59% positive cells in 12 (13.6) 55.4 55.4
gliosides were extensively expressed in NSCLC, although they
tumor cells
have never been previously detected in normal human tissues.
20~39% positive cells in 17 (19.3) 58.6 57.8
We used GMR8 mAb, which specifically reacts with ganglio-
tumor cells
sides having NeuGc alpha 2,3Gal-terminal structures, generated
6~19% positive cells in 16 (18.2) 56.9 56.9
by immunizing mice with purified GM3 (NeuGc). In contrast,
tumor cells
van Cruijsen et al.(18) and Blanco et al.(19) reported that
0~5% positive cells in 13 (14.8) 58.8 58.8
NSCLC tissues stained positive in a tissue microarray analysis
tumor cells
using another anti-GM3 (NeuGc) mouse mAb, 14F7 mAb.
OS, overall survival; PFS, progression-free survival. Their studies demonstrated that GM3 (NeuGc) expression was

Hayashi et al. Cancer Sci | January 2013 | vol. 104 | no. 1 | 45


© 2012 Japanese Cancer Association
(A) (B) 100 Low N-glycolyated gangliosides expression
100 Low N-glycolyated gangliosides expression
Median overall survival 57.5 months
Median progression free survival 57.4 months

80

Progression-free survival (%)


80

Overall survival (%)


60 60

40 40
High N
N-glycolyated
glycolyated gangliosides expression
High N-glycolyated gangliosides expression Median overall survival 52.4 months
Median progression-free survival 47.5 months
20 20
Log rank P = 0.156
Log rank P < 0.01

0 0

0 20 40 60 80 100 0 20 40 60 80 100
Time after primary tumor resection (months) Time after primary tumor resection (months)

Fig. 2. Kaplan–Meier plots for (A) progression-free survival and (B) overall survival based on N-glycolyated ganglioside expression in non-small-
cell lung cancer patients.

(A)

(B)

Fig. 3. (A) TLC immunostaining using GMR8 mAb. Normal tissues = 1, 2, 3 and 4; cancer tissues = 5, 6, 7 and 8. (B) TLC immunostaining using
anti-GM3 (NeuAc) mAb, M2590. Normal tissues = 1, 2, 3 and 4; cancer tissues = 5, 6, 7 and 8.

associated with a better prognosis in NSCLC patients. On the sequences of the CMAH gene in human NSCLC tissues were
contrary, our results indicated that patients with high NeuGc- the same as those in normal tissues. Thus, CMAH enzyme
containing ganglioside expression had a low overall survival activity in human NSCLC tissues is considered to be inactive,
rate and a significantly low progression-free survival rate. The similar to that in normal tissues. This suggests the existence of
reason for this discrepancy might be that GMR8 mAb have a an alternative pathway for NeuGc-containing gangliosides, dif-
different specificity for recognizing molecules compared with ferent from the normal pathway that is mediated by CMAH
the 14F7 mAb. In NSCLC tissues, our immunostaining of gan- enzyme activity. Yin et al. reported a hypoxic culture induced
gliosides on TLC demonstrated that at least two NeuGc-con- NeuGc-containing ganglioside expression on human cells as a
taining gangliosides, GM3 (NeuGc) and GD1a (NeuGc), were possible candidate for the alternative pathway because a hyp-
recognized by GMR8 mAb. In addition, our investigation used oxic culture markedly induced mRNA for the sialic acid trans-
93 patients, which is a larger sample size compared with that in porter sialin.(21) This might be the cause of NeuGc-containing
their investigation, which used 26 patients. Thus, differences in ganglioside expression in NSCLC tissues because tumor cells
the specificity of the antibodies and the number of patients in a hypoxic environment promote incorporation of NeuGc-type
might lead to differences in results. sialic acid in extracellular fluid via activation of the sialic acid
Human tissues and body fluids contain little or undetectable transporter. Selection of hypoxia-resistant cancer cells by the
amounts of NeuGc, whereas corresponding samples from chim- hypoxic environment in locally advanced tumor sites is known
panzees and bonobos express high levels.(20) Human deficiency to result in the culture expansion of cancer cells with higher
of NeuGc is due to a mutation in the CMAH gene.(7–9) Human invasive and metastatic activities.(22,23) Overall, NSCLC with
CMAH is inactive because of the deletion of the region corre- high NeuGc-containing ganglioside expression might occur in a
sponding to the N-terminal domain of the hydroxylase, which severely hypoxic environment, which might be related to poor
is essential for enzyme activity.(9) We observed that cDNA survival as suggested earlier.

46 doi: 10.1111/cas.12027
© 2012 Japanese Cancer Association
after exposure to GM3 (NeuGc) was lower than that after
exposure to GM3 (NeuAc). The difference in the inhibition of
EGFR tyrosine kinase between the two types may be associ-
ated with the different manner of GM3 binding to EGFR via
carbohydrate–carbohydrate interaction. Furthermore, our in
vitro results were consistent with the results of survival analy-
sis, indicating that prognosis is poor for NSCLC patients with
high NeuGc-containing ganglioside expression. Thus, inhibi-
tion of EGFR tyrosine kinase with GM3 is weakened in
NSCLC tissues with high NeuGc-containing ganglioside
expression, possibly leading to an environment where a tumor
can proliferate easily.
Our immunohistochemical studies showed that NeuGc-con-
taining gangliosides including GM3 (NeuGc) are widely
expressed in NSCLC tissues. Their expression could be associ-
ated with patient survival due to the difference of GM3 (NeuGc)
and GM3 (NeuAc) on the effects on inhibition of EGFR tyrosine
Fig. 4. In vitro inhibitory effect of GM3 (N-glycolyl neuraminic acid kinase. Further study is required to determine the expression
[NeuGc]) and GM3 (N-acetylneuraminic acid [NeuAc]) on epidermal ratio of GM3 (NeuGc) and GM3 (NeuAc) in NSCLC.
growth factor receptor (EGFR) tyrosine phosphorylation. Phosporyla-
tion was analyzed using western blotting with mAb PY20 in the upper
This work decribes that NeuGc-containing gangliosides
section. The density of band analyzed as a percentage of the control including GM3 (NeuGc) are widely expressed in NSCLC and
value is shown in the lower section. NeuGc-containing ganglioside expression is associated with
patient survival. It might be useful for NSCLC therapy to
decrease the NeuGc-containing ganglioside ratio in NSCLC tissues.
Epidermal growth factor receptor belongs to the erbB trans-
membrane receptor family and plays a role in cell proliferation Acknowledgments
and differentiation at the epithelial surface.(24) Delay in cell
growth of the human epidermoid carcinoma cell line A431 is The authors are grateful to Dr I. Kawashima at Tokyo Metropolitan
caused by GM3 (NeuAc)-mediated inhibition of EGFR tyro- Institute of Medical Science for his valuable comments on the present
sine kinase.(2) GM3 (NeuAc) inhibits EGFR tyrosine kinase study.
via carbohydrate–carbohydrate interactions between N-glycans
with GlcNAc termini on EGFR and oligosaccharides on Disclosure Statement
GM3.(6) In the present study, we found that the inhibitory
effect of GM3 on EGFR tyrosine kinase reduced in A431 cells The authors declare no competing financial interests.

References 13 Scursoni AM, Galluzzo L, Camarero S et al. Detection of N-glycolyl GM3


ganglioside in neuroectodermal tumors by immunohistochemistry: an attrac-
1 Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA tive vaccine target for aggressive pediatric cancer. Clin Dev Immunol 2011;
Cancer J Clin 2005; 55: 74–108. 2011: 245181.
2 Bremer EG, Schlessinger J, Hakomori S. Ganglioside-mediated modulation 14 Carr A, Mullet A, Mazorra Z et al. A mouse IgG1 monoclonal antibody spe-
of cell growth. J Biol Chem 1986; 261: 2434–40. cific for N-glycolyl GM3 ganglioside recognized breast and melanoma
3 Miljan EA, Meuillet EJ, Mania-Farnell B et al. Interaction of the extracellu- tumor. Hybridoma 2000; 19: 241–7.
lar domain of the epidermal growth factor receptor with gangliosides. J Biol 15 Ozawa H, Kawashima I, Tai T. Generation of murine monoclonal antibodies
Chem 2002; 277: 10108–13. specific for N-glycolylneuraminic acid-containing gangliosides. Arch Bio-
4 Yoon SJ, Nakayama K, Hikita T et al. Epidermal growth factor receptor chem Biophys 1992; 294: 427–33.
tyrosine kinase is modulated by GM3 interaction with N-linked GlcNAc ter- 16 Zhou Q, Hakomor S, Kitamura K et al. GM3 directly inhibits tyrosine phos-
mini of the receptor. Proc Natl Acad Sci USA 2006; 103: 18987–91. phorylation and de-N-acetyl-GM3 directly enhances serine phosphorylation
5 Wang XQ, Sun P, Paller AS. Ganglioside GM3 blocks the activation of of epidermal growth factor receptor, independently of receptor–receptor
epidermal growth factor receptor induced by integrin at specific tyrosine interaction. J Biol Chem 1994; 269: 1959–65.
sites. J Biol Chem 2003; 277: 47028–34. 17 Chomcynski P, Sacchi N. Single-step method of RNA isolation by acid
6 Kawashima N, Yoon SJ, Itoh K et al. Tyrosine kinase activity of epidermal guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem 1987;
growth factor receptor is regulated by GM3 binding through carbohydrate to 162: 156–9.
carbohydrate interaction. J Biol Chem 2009; 284: 6147–55. 18 van Cruijsen H, Ruiz MG, van der Valk P et al. Tissue micro array analysis
7 Schlenzka W, Shaw L, Kelm S et al. CMP-N-acetylneuramic acid hydroxy- of ganglioside N-glycolyl GM3 expression and signal transducer and activa-
lase; the first cytosolic Rieske iron-sulphur protein to be described in Euk- tor of transcription (STAT)-3 activation in relation to dendritic cell infiltra-
arya. FEBS Lett 1996; 385: 197–200. tion and microvessel density in non-small cell lung cancer. BMC Cancer
8 Chou HH, Hayakawa T, Diaz S et al. Inactivation of CMP-N-acetylneuramic 2009; 9: 180.
acid hydroxylase occurred prior to brain expansion during human evolution. 19 Blanco R, Rengifo CE, Cedeño M, Frómeta M, Rengifo E, Carr A. Immuno-
Proc Natl Acad Sci USA 2002; 99: 11736–41. reactivity of the 14F7 Mab (raised against N-Glycolyl GM3 Ganglioside) as
9 Irie A, Koyama S, Kozutsumi Y et al. The molecular basis for the a positive prognostic factor in non-small-cell lung cancer. Patholog Res Int
absence of N-glycolylneuramic acid in humans. J Biol Chem 1998; 273: 2012; 2012: 235418.
15866–71. 20 Muchmor EA, Diaz S, Varki A. A structural difference between the cell sur-
10 Noguchi M, Suzuki T, Kabayama K et al. GM3 synthase gene is a novel faces of humans and the great apes. Am J Phys Anthropol 1998; 107: 187–98.
biomarker for histological classification and drug sensitivity against epider- 21 Yin J, Hashimoto A, Izawa M et al. Hypoxic culture induces expression of sia-
mal growth factor receptor tyrosine kinase inhibitors in non-small cell lung lin, a sialic acid transporter, and cancer-associated gangliosides containing
cancer. Cancer Sci 2007; 98: 1625–32. non-human sialic acid on human cancer cells. Cancer Res 2006; 66: 2937–45.
11 Marquina G, Waki H, Fernadez LE et al. Gangliosides expressed in human 22 Semenza GL. Hypoxia and cancer. Cancer Metastasis 2007; 26: 223–4.
breast cancer. Cancer Res 1996; 56: 5165–71. 23 Harris AL. Hypoxia – a key regulatory factor in tumor growth. Nat Rev Can-
12 Scursoni AM, Galluzzo L, Camarero S et al. Detection and characterization cer 2002; 2: 38–47.
of N-glycolyated gangliosides in Wilms tumor by immunohistochemistry. 24 Ullrich A, Schlessinger J. Signal transduction by receptors with tyrosine
Pediatr Dev Pathol 2010; 13: 18–23. kinase activity. Cell 1990; 61: 203–12.

Hayashi et al. Cancer Sci | January 2013 | vol. 104 | no. 1 | 47


© 2012 Japanese Cancer Association

You might also like