ArticlePDF Available

BCAT1 Overexpression is an Indicator of Poor Prognosis in Patients with Urothelial Carcinomas of the Upper Urinary Tract and Urinary Bladder

Authors:

Abstract and Figures

Amino acid biosynthesis is one of the cardinal events of carcinogenesis that has not been investigated in urothelial carcinoma (UC). By data mining a published transcriptomic database of UCs of urinary bladder (UBUCs) (GSE31684), we identified branched-chain amino acid transaminase 1 (BCAT1) as the most significantly stepwise upregulated gene during tumor progression among those associated with amino acid biosynthetic process (GO:0008652). Accordingly, we analyzed BCAT1 transcript and protein expression with their clinicopathological significance. We used real time RT-PCR to detect BCAT1 transcript levels in 20 UCs of upper tract (UTUCs) and 20 UBUCs, respectively. Immunohistochemical study was performed to determine BCAT1 protein expression in 340 UTUCs and 295 UBUCs. Higher BCAT1 transcript levels were associated with higher pT status in both groups (p<0.05). BCAT1 protein overexpression was also significantly associated with adverse clinicopathological features, e.g., advanced pT stage, nodal metastasis, high pathological grade and etc. (p<0.05). BCAT1 overexpression predicted worse disease-specific survival and metastasis-free survival in both univariate and multivariate analyses (p≤0.001). BCAT1 overexpression is associated with advanced tumor status, and implies adverse clinical outcomes of UCs, suggesting its role in tumor progression and could serve as a prognostic biomarker and a novel therapeutic target in UC. This article is protected by copyright. All rights reserved. This article is protected by copyright. All rights reserved.
Content may be subject to copyright.
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
304
J
Jo
ou
ur
rn
na
al
l
o
of
f
C
Ca
an
nc
ce
er
r
2016; 7(3): 304-313. doi: 10.7150/jca.13638
Research Paper
Necdin Overexpression Predicts Poor Prognosis in Pa-
tients with Urothelial Carcinomas of the Upper Urinary
Tract and Urinary Bladder
I-Wei Chang1,2, Yu-Hui Wang3,4, Wen-Jeng Wu5,6,7,8,9, Peir-In Liang10, Wei-Ming Li5,6,7,8,9, Bi-Wen Yeh5,6,7,8,9,
Ting-Feng Wu11, Hong-Lin He1, Steven Kuan-Hua Huang12,, Chien-Feng Li4,11,13,14,
1. Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan;
2. School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan;
3. Institute of Bioinformatics and Biosignal Transduction, National Cheng Kung University, Tainan, Taiwan;
4. Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan;
5. Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan;
6. Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan;
7. Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan;
8. Department of Urology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan;
9. Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan;.
10. Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan;
11. Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan;
12. Department of Urology, Chi Mei Medical Center;
13. Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan;
14. National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.
Corresponding authors: Chien-Feng Li, M.D., PhD., Department of Pathology, Chi Mei Medical Center, Tainan 701, Taiwan., E-mail: angelo.p@yahoo.com.tw,
or Steven Kuan-Hua Huang., M.D., Ph.D., Department of Urology, Chi Mei Medical Center, Tainan 701, Taiwan., E-mail: skhsteven@gmail.com
© Ivyspring International Publisher. Reproduction is permitted for personal, noncommercial use, provided that the article is in whole, unmodified, and properly cited. See
http://ivyspring.com/terms for terms and conditions.
Received: 2015.08.23; Accepted: 2015.10.28; Published: 2016.01.10
Abstract
Background and Aims: Oncogenesis is a multistep process, resulting from the accumulations of
multiple mutations. Of these mutations, self-sufficiency in growth signals, i.e., disruption of cell growth
regulation, is the first episode. Nonetheless, the genes associated with cell growth dysregulation have
seldom been systematically evaluated in either urothelial carcinomas of upper urinary tract (UTUC) or
urothelial carcinomas of urinary baldder (UBUC). By data mining a published transcriptomic dataset of
UBUCs (GSE31684), we identified the NDN gene as one of the most significant of those associated with
the regulation of cell growth and found this gene is associated with advanced tumor status and meta-
static disease (GO:0001558). Accordingly, we analyzed NDN transcript and protein expression with
their clinicopathological significance.
Materials and Methods: We used real time RT-PCR to detect NDN transcript levels in 27 UTUCs
and 27 UBUCs, respectively. Immunohistochemical study was performed to determine NDN protein
(a.k.a. Necdin) expression evaluated by H-score method in 340 UTUCs and 295 UBUCs. NDN ex-
pression was further correlated with clinicopathological features and disease-specific survival (DSS) and
metastasis-free survival (MeFS).
Results: NDN transcriptional level was significantly higher in UCs of both sites with stepwise more
advanced pT statuses. Through immunohistochemistry, we found NDN protein expression was sig-
nificantly associated with adverse clinicopathological parameters, e.g., advanced pT status, nodal me-
tastasis, high grade histological patterns, and frequent mitotses (all P<0.05). In univariate analysis, NDN
overexpression not only predicted worse DSS and MeFS in both the UTUC and UBUC groups, it also
served as an independent prognostic factor for DSS and MeFS in multivariate analysis (all P<0.05).
Conclusions: NDN may play an important role in tumor progression in UC and could serve as a
prognostic biomarker and a potential novel therapeutic target in UC.
Key words: NDN gene, Necdin, Urothelial carcinoma, Prognosis.
Ivyspring
International Publisher
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
305
Introduction
Urothelial carcinoma (UC) is the most prevalent
histological type of malignancy throughout the uri-
nary tract, from the upper urinary tract (UT) to the
urinary bladder (UB) [1]. The former consists of the
renal pelvis and the ureter. In contrast to urothelial
carcinoma of the urinary bladder (UBUC), which is
the seventh most common cancer in the United States
[2], urothelial carcinoma of the upper urinary tract
(UTUC) is much uncommon and accounts for only 5%
to 10% of all UCs [3]. Nevertheless, the incidence of
UTUC in Taiwan is unusually high, especially in
southern Taiwan and areas of endemic “Black-foot
disease” [4-6]. Arsenic-contaminated drinking water
may contribute to the prevalence of UTUC in Taiwan
[6]. Etiologically, all UCs, regardless of anatomical
location, are attributed to identical carcinogens, such
as cigarette smoking, and aromatic amines, e.g. ben-
zidine, β-naphtylanine [7-9]. However, certain popu-
lations are particularly predisposed to UTUC. For
instance, patients with analgesic nephropathy [10],
Chinese herb nephropathy [11, 12], and Balkans
nephropathy [11, 13] are more susceptible to UTUC
than UBUC. In spite of the fact that UTUCs usually
have higher stage and grade than UBUCs, their clini-
cal behavior is similar after balancing the stages and
grades [14]. In addition, the previous study revealed
that gene expression profiles of both UBUCs and
UTUCs are much alike [15]. These findings indicate
that carcinogensis of UCs from both anatomical loca-
tions may participate in a common molecular path-
way.
Cancer is essentially a disease of regulation of
cell growth; genes that regulate cell growth must be
altered in order to transform normal cells into cancer
cells [16]. Oncogenesis is a multistep process, result-
ing from the accumulations of multiple mutations. Of
these mutations, self-sufficiency in growth signals, i.e.,
disruption of cell growth regulation is the first epi-
sode [17]. Nonetheless, the genes associated with cell
growth dysregulation have seldom been systemati-
cally evaluated in either UTUC or UBUC. By data
mining documented transcript expression profiles
(GSE31684) in the Gene Expression Omnibus (GEO,
National Center for Biotechnology Information
(NCBI), Bethesda, MD, USA) with a special focus on
growth regulation (GO:0001558), we found that tran-
scription of the NDN gene was significantly upregu-
lated from early tumor development and associated
stepwise with tumor progression. This evidence sug-
gests that the NDN gene plays an important role in
tumorigenesis and its progression.
The NDN gene encodes Necdin protein, a
member of the melanoma-associated antigen gene
(MAGE) family [18] that was first identified in neu-
rally differentiated mouse stem cells of P19 embryonal
carcinoma cell lines treated with retinoic acid [19].
Necdin was originally recognized as a suppressor of
cell proliferation in postmitotic neurons, leading the
differentiated neurons into permanent withdrawal
from the cell cycle due to constitutive and lifelong
expression of Necdin [20]. Moreover, NDN transcripts
and their encoded proteins showed downregulation
or low expression in previous UBUC cell lines and
human UBUC tissue studies [21]. Hence, we con-
ducted this study to elucidate the expression of Nec-
din protein and NDN mRNA in UCs from both ana-
tomical sites, as well as their association with clini-
copathological parameters and clinical outcomes.
Materials and Methods
Data mining the GEO to identify the most al-
tered transcripts in UCs
We performed data mining of the GEO of NCBI,
identifying dataset GSE31684 (http://www.ncbi.nlm.
nih.gov/geo/query/acc.cgi?acc=GSE31684) in order
to analyze radical cystectomy specimens from 93 pa-
tients with UBUC using the Affymetrix GeneChip
Human Genome U133 Plus 2.0 Array. To analyze all
probe sets, we used Nexus Expression 3 statistical
software (BioDiscovery, El Segundo, CA, USA)
without preselection or filtering. Under supervision,
our comparative analysis examined the statistical sig-
nificance of differentially expressed transcripts on the
basis of primary tumor status (pT) and the develop-
ment of metastatic events. We performed functional
profiling using transcriptomes of high-stage UCs
(pT2-pT4) with metastases and low-stage UCs
(pTa-pT1) devoid of metastasis, focusing on those
related to the regulation of cell growth (GO:0001558).
We further analyzed survival patterns by dichoto-
mizing all cases into high-expression and
low-expression clusters for computing the prognostic
significance of the selected genes.
Patients and tumor specimens
The Institutional Review Board of Chi Mei
Medical Center approved this study (IRB971006). For
immunohistochemical study and survival analysis,
we enrolled 635 consecutive cases diagnosed as con-
ventional UC between 1996 and 2004, from the ar-
chives of the Department of Pathology, Chi-Mei
Medical Center. Of these cases, 340 tumors originated
from the UT and 295 arose from the UB. Other histo-
logical classifications as well as variants of UC were
excluded. The criteria for clinicopathological evalua-
tion were essentially identical to those in our previous
work [22]. Detailed information is provided in the
online supplementary material.
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
306
Transcriptional levels of the NDN gene
The materials for genetic examination were
gathered from macrodissection of fresh tumor tissue.
For quantification of NDN mRNA expression, we ex-
tracted total RNAs, quantified them, and submitted
them for reverse-transcription from 27 UTUCs and 27
UBUCs (both were composed of 9 cases of pTa; 9 pT1;
9 pT2-4), respectively. Using pre-designed TaqMan
assay reagents (Applied Biosystems, Waltham, MA,
USA), we measured mRNA abundance of NDN
(Hs00267349_s1) with the ABI StepOnePlus™ System,
as previously described [23]. We calculated the fold
expression of NDN relative to normal urothelium by
comparative Ct method, after normalization to
POLR2A (Hs01108291_m1) as the internal control.
Immunohistochemical staining and scoring of
NDN
Tissue sections underwent standard procedures
for deparaffinization, rehydration and antigen re-
trieval. Afterwards, the sections were incubated with
a primary antibody targeting NDN (TA506975, 1:150;
OriGene, Rockville, MD, USA) for an hour. Normal
brain tissue with or without incubation of primary
antibody were run in parallel as positive and negative
control, respectively. We scored NDN immunoreac-
tivity based on the combination of the percentage and
intensity of positivity of the immunostain in tumoral
nuclei in order to generate an H-score, which was
calculated using the following equation:
H-score=ΣPi(i +1), where i is the intensity of stained
tumor cells (0-3+), and Pi is the percentage of stained
tumor cells for each intensity varying from 0% to
100%. This formula produced a score range from
100-400, where 100 indicated that 100% of tumor cells
were negative and 400 indicated 100% of tumor cells
were strongly stained (3+) [24, 25].
Statistical analysis
We performed statistics using SPSS V.14.0 soft-
ware (SPSS Inc., Chicago, IL, USA). The median
H-score of NDN immunoreactivity was used as the
cut-off to dichotomize the study cohort, separating
cases into high expression and low expression groups.
We used Pearson’s χ2 test to compare NDN expres-
sion status and various categorical clinicopathological
parameters. Mann-Whitney U test was used to com-
pare NDN expression and numerical ones, i.e., mitotic
rate. The end points analyzed were disease-specific
survival (DSS) and metastasis-free survival (MeFS),
calculated from the date of tumor resection to the date
the event developed. Patients lost to follow-up were
censored on the latest follow-up date. We performed
univariate survival analyses using Kaplan-Meier plots
and compared them by log-rank test. Those parame-
ters with univariate P<0.1 were enrolled into multi-
variate tests using Cox proportional hazards regres-
sion. For all analyses, we used two-sided tests of sig-
nificance, with P<0.05 considered statistically signifi-
cant.
Results
NDN is a significant differentially upregulated
transcript linked with regulation of cell growth
in UBUCs
From the transcriptomic profiling of 93 UBUC
cases (GSE31684), which contains 78 high-stage
(T2-T4) and 15 low-stage (Ta-T1) cases, 28 with meta-
static disease and 49 without metastasis, we identified
six probes covering two transcripts associated with
regulation of cell growth (GO:0001558) (Figure 1). As
shown in Table 1, both transcripts, i.e., IGFBP5 and
NDN, were significantly upregulated with a log2 ratio
of 0.337 to 2.6399-fold and 0.7821-fold upregulation,
respectively, comparing high-stage to low-stage (all
P<0.005). IGFBP5 and NDN upregulation also showed
statistically significant association with metastatic
disease in all probes (all P0.0001). Comparing
high-expression (n=37) to low-expression (n=56)
clusters, high expression levels of NDN transcripts
significantly predicted worse disease-specific survival
(Figure S1, P=0.0059). We had investigated IGFBP5 in
a previous study [26]; however, NDN has not previ-
ously been systematically studied in UCs. Hence, we
further characterized the endogenous expression lev-
els and clinical significance of both NDN transcript
and its protein in UC.
Higher NDN mRNA transcriptional levels in
UBUCs and UTUCs with advanced pT stage
In the examined 27 UBUCs and 27 UTUCs, NDN
transcripts were significantly more abundant in tu-
mors with stepwise more advanced pT status, from
pTa to pT1 to pT2-4 (all P<0.05), suggesting it plays a
role in tumor progression (Figure 2).
Clinicopathological findings on UTUCs and
UBUCs
Clinicopathological features of the patients with
UTUCs and UBUCs are listed in Table 2. In the UTUC
group, no obvious sex predilection was noted despite
there being slightly more women (n=182, 53.5%). In
contrast, the majority of patients with UBUC were
male (n=216, 73.2%). For both types of UC, most pa-
tients’ age at diagnosis was older than 65 years
(n=202, 59.4% for UTUC and n=174, 59.0% for UBUC).
In the UTUC group, synchronous multifocal tumors
occurred in 62 patients (18.2%); both the renal pelvis
and ureter were involved in 49 (14.4%) of these. In
both UC groups, most tumors were classified as high
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
307
histological grade (n=284, 83.5% for UTUC and n=239,
81% for UBUC). Advanced pT stages (pT2-T4) were
seen in 46.8% (n=159) and 41.7% (n=123) of patients
with UTUCs and UBUCs, respectively. Nodal metas-
tasis was detected in 8.2% (n=28) and 9.8% (n=29) of
UTUC and UBUC patients, respectively. Vascular
invasion was noted in 31.2% of UTUC (n=106) and
16.6% of UBUC cases (n=49); while perineurial inva-
sion was observed in 5.6% of UTUC (n=19) and 6.8%
of UBUC cases (n=20). In addition, about half of the
tumors showed ten or more mitoses per ten high
power fields (n=167, 49.1% for UTUC and n=156,
52.9% for UBUC).
Correlations between immunoreactivity of
NDN and clinicopathological parameters in
UTUCs and UBUCs
NDN showed variable nuclear expression in
UCs from both sites. The median H-score for each
group was 105 and 370 for the UTUCs and UBUCs,
respectively. As Table 2 demonstrates, after dichoto-
mizing tumors into low and high NDN expression,
increased NDN expression in UC of both anatomical
sites was significantly associated with stepwise in-
creases of pT status (Figure 3, UTUC, P=0.003; UBUC,
P<0.001), lymph node metastasis (UTUC, P =0.002;
UBUC, P=0.012), high grade histological patterns
(UTUC, P=0.019; UBUC, P=0.003) and higher mitotic
rate (UTUC, P=0.004; UBUC, P=0.005). Increased
NDN expression was significantly associated with
vascular invasion and perineural invasion only in the
UBUC group (P=0.008 and 0.001, respectively).
Survival analysis for UTUC and UBUC pa-
tients
The univariate and multivariate analyses of rela-
tionships between clinical outcomes and miscellane-
ous clinicopathological parameters in both UTUC and
UBUC patients are illustrated in Tables 3-4. In mul-
tivariate analysis, multifocality (P=0.007), nodal me-
tastasis (P<0.001), high histological grade (P=0.007)
and perineurial invasion (P=0.001) are independent
prognostic factors for poor DSS in the UTUC group.
Similarly, inferior MeFS was significantly associated
with multifocality (P=0.002), nodal metastasis
(P=0.001), high histological grade (P=0.030), vascular
invasion (P<0.001) and perineurial invasion (P=0.014)
in patients with UTUCs. In UTUCs, advanced pT
status was significantly associated with worse DSS
and MeFS in univariate (P<0.0001) but not in multi-
variate analyses. Tumor location and vascular inva-
sion in UTUC group correlated with poorer patient
DSS in univariate analysis only (P=0.0079 and
P<0.0001, respectively).
Figure 1. Analysis of gene expression in urothelial carcinoma using a published
transcriptomic dataset (GSE31684). Clustering analysis of genes focused on the
regulation of cell growth (GO:0001558) revealed NDN is one of the most significantly
upregulated genes associated with increments of pT status and metastatic disease.
Tissue specimens from tumors with different pT statuses are indicated on top of the
heatmap, and expression levels of upregulated and downregulated genes are repre-
sented as a spectrum of brightness or red and green, respectively. Those with
unaltered mRNA transcriptional levels are coded black.
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
308
In the UBUC group, multivariate analysis re-
vealed advanced pT status and higher mitotic rate
were significantly associated with both dismal DDS
and MeFS (all P<0.05). Lymph node metastasis was
significantly associated with poor MeFS (P=0.037) in
multivariate analysis, as well. In UBUCs, high histo-
logical grade, and vascular and perineural invasion
were significantly associated with both adverse DSS
and MeFS in univariate analysis (all P<0.005), but not
in multivariate analysis.
Prognostic significance of NDN expression in
UC
As shown in Tables 3-4 and Figure 4, in uni-
variate analysis, either the UTUC or UBUC group
with NDN overexpression had significantly worse
DSS and MeFS (P<0.005 for all). Notably, in multi-
variate analysis, high NDN nuclear expression was
still an independent prognosticator predicting more
dismal DSS and MeFS for all UCs (P<0.05 for all).
Table 1. Summary of differentially expressed genes associated with regulation of cell growth (GO:0001558) and showed positive as-
sociations to cancer invasiveness and metastasis in the transcriptome of urothelial carcinoma of urinary bladder (GSE31684).
Probe
Comparing T2-4 to
Ta-T1
#
Gene Symbol
Biological Process
Molecular Function
log ratio
p-value
log ratio
p-value
1554741_s_at
0.4313
<0.0001
0.3919
<0.0001
IGFBP5
regulation of cell growth, signal transduction
growth factor binding, insu-
lin-like growth factor binding
201666_at
1.1435
0.0001
1.0825
<0.0001
IGFBP5
regulation of cell growth, signal transduction
growth factor binding, insu-
lin-like growth factor binding
205131_x_at
0.337
0.0008
0.2968
0.0001
IGFBP5
regulation of cell growth, signal transduction
growth factor binding, insu-
lin-like growth factor binding
205168_at
0.7821
0.0044
1.0228
<0.0001
NDN
axon extension involved in development, axonal
fasciculation, axonogenesis, central nervous system
development, glial cell migration, negative regula-
tion of cell proliferation, nerve growth factor re-
ceptor signaling pathway, nervous system devel-
opment, neuron development, neuron migration,
regulation of cell growth, regulation of progression
through cell cycle, regulation of transcription;
DNA-dependent, respiratory gaseous exchange,
sensory perception of pain, transcription
DNA binding, gamma-tubulin
binding, protein binding
205782_at
0.8325
0.0003
0.8815
<0.0001
IGFBP5
regulation of cell growth, signal transduction
growth factor binding, insu-
lin-like growth factor binding
207426_s_at
2.6399
<0.0001
1.4678
<0.0001
IGFBP5
regulation of cell growth, signal transduction
growth factor binding, insu-
lin-like growth factor binding
#, Meta., distal metastasis developed during follow-up; Non-Meta.: no metastatic event developed.
Table 2. Correlations between NDN expression and other important clinicopathological parameters in urothelial carcinomas.
Parameter
Category
Upper Urinary Tract Urothelial Carcinoma
Urinary Bladder Urothelial Carcinoma
Case No. (%)
NDN Expression
p-value
Case No. (%)
NDN Expression
p-value
Low (%)
High (%)
Low (%)
High (%)
Gender
Male
158 (46.5)
86 (54.4)
72 (45.6)
0.128
216 (73.2)
109 (50.5)
107 (49.5)
0.719
Female
182 (53.5)
84 (46.2)
98 (53.8)
79 (26.8)
38 (48.1)
41 (51.9)
Age (years)
< 65
138 (40.6)
70 (50.7)
68 (49.3)
0.825
121 (41.0)
59 (48.8)
62 (51.2)
0.759
≥ 65
202 (59.4)
100 (49.5)
102 (50.5)
174 (59.0)
88 (50.6)
86 (49.4)
Tumor location
Renal pelvis
141 (41.5)
71 (50.4)
70 (49.6)
0.685
-
-
-
-
Ureter
150 (44.1)
72 (48.0)
78 (52.0)
-
-
-
-
Renal pelvis & ureter
49 (14.4)
27 (55.1)
22 (44.9)
-
-
-
-
Multifocality
Single
278 (81.8)
138 (49.6)
140 (50.4)
0.779
-
-
-
-
Multifocal
62 (18.2)
32 (51.6)
30 (48.4)
-
-
-
-
Primary tumor (pT)
pTa
89 (26.2)
56 (62.9)
33 (37.1)
0.003*
84 (28.5)
62 (73.8)
22 (26.2)
<0.001*
pT1
92 (27.0)
49 (53.3)
43 (46.7)
88 (29.8)
48 (54.5)
40 (45.5)
pT2-pT4
159 (46.8)
65 (40.9)
94 (59.1)
123 (41.7)
37 (30.0)
86 (70.0)
Nodal metastasis
Negative (pN0)
312 (91.8)
164 (52.6)
148 (47.4)
0.002*
266 (90.2)
139 (52.3)
127 (47.7)
0.012*
Positive (pN1-pN3)
28 (8.2)
6 (21.4)
22 (78.6)
29 (9.8)
8 (27.6)
21 (72.4)
Histological grade
Low grade
56 (16.5)
36 (64.3)
20 (35.7)
0.019*
56 (19.0)
38 (67.9)
18 (32.1)
0.003*
High grade
284 (83.5)
134 (47.2)
150 (52.8)
239 (81.0)
109 (45.6)
130 (54.4)
Vascular invasion
Absent
234 (68.9)
122 (52.1)
112 (47.9)
0.242
246 (83.4)
131 (53.3)
115 (46.7)
0.008*
Present
106 (31.1)
48 (45.3)
58 (54.7)
49 (16.6)
16 (32.7)
33 (67.3)
Perineural invasion
Absent
321 (94.4)
164 (51.1)
157 (48.9)
0.098
275 (93.2)
144 (52.4)
131 (47.6)
0.001*
Present
19 (5.6)
6 (31.6)
13 (68.4)
20 (6.8)
3 (15.0)
17 (85.0)
Mitotic rate (per 10
high power fields)
#
340
9.66+/-7.315
14.96+/-15.324
0.004*
295
11.41+/-10.286
17.38+/-16.469
0.005*
#, Mann-Whitney U test; *, Statistically significant.
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
309
Table 3. Univariate log-rank and multivariate analyses for Disease-specific and Metastasis-free Survivals in upper urinary tract urothelial
carcinoma.
Parameter
Category
Case No.
(%)
Disease-specific Survival
Metastasis-free Survival
Univariate analysis
Multivariate analysis
Univariate analysis
Multivariate analysis
No. of event
(%)
p-value
R.R.
95% C.I.
p-value
No. of event
(%)
p-value
R.R.
95% C.I.
p-value
Gender
Male
158 (46.5)
28 (17.7)
0.8286
-
-
-
32 (20.3)
0.7904
-
-
-
Female
182 (53.5)
33 (18.1)
-
-
-
38 (20.9)
-
-
-
Age (years)
< 65
138 (40.6)
26 (18.8)
0.9943
-
-
-
30 (21.7)
0.8470
-
-
-
≥ 65
202 (59.4)
35 (17.3)
-
-
-
40 (19.8)
-
-
-
Tumor side
Right
177 (52.1)
34 (19.2)
0.7366
-
-
-
38 (21.5)
0.3074
-
-
-
Left
154 (45.3)
26 (16.9)
-
-
-
32 (20.8)
-
-
-
Bilateral
9 (2.6)
1 (11.1)
-
-
-
0 (0)
-
-
-
Tumor loca-
tion
Renal pelvis
141 (41.5)
24 (17.0)
0.0079*
1
-
0.840
31 (22.0)
0.0659
-
-
-
Ureter
150 (44.1)
22 (14.7)
0.876
0.472-1.625
25 (16.7)
-
-
-
Renal pelvis &
ureter
49 (14.4)
15 (30.6)
1.286
0.607-7.438
14 (28.6)
-
-
-
Multifocality
Single
278 (81.8)
48 (17.3)
0.0026*
1
-
0.007*
52 (18.7)
0.0127*
1
-
0.002*
Multifocal
62 (18.2)
18 (29.0)
2.821
1.321-6.024
18 (29.0)
2.402
1.381-4.4
178
Primary
tumor (pT)
pTa
89 (26.2)
2 (2.2)
<0.0001*
1
-
0.071
4 (4.5)
<0.0001*
1
-
0.227
pT1
92 (27.0)
9 (9.8)
3.042
0.646-14.324
15 (16.3)
2.344
0.756-7.2
67
pT2-pT4
159 (46.8)
50 (31.4)
5.159
1.150-23.153
51 (32.1)
2.497
0.795-7.8
46
Nodal me-
tastasis
Negative
(pN0)
312 (91.8)
42 (13.5)
<0.0001*
1
-
<0.001*
55 (17.6)
<0.0001*
1
-
0.001*
Positive
(pN1-pN3)
28 (8.2)
19 (67.9)
5.071
2.753-9.340
15 (53.6)
2.888
1.552-5.3
72
Histological
grade
Low grade
56 (16.5)
4 (7.1)
0.0215*
1
-
0.007*
3 (5.4)
0.0027*
1
-
0.030*
High grade
284 (83.5)
57 (20.0)
3.853
1.441-10.298
67 (23.6)
3.818
1.139-12.
792
Vascular
invasion
Absent
234 (68.9)
24 (10.3)
<0.0001*
1
-
0.059
26 (11.1)
<0.0001*
1
-
<0.001*
Present
106 (31.1)
37 (34.9)
1.774
0.978-3.218
44 (41.5)
2.960
1.625-5.3
92
Perineural
invasion
Absent
321 (94.4)
50 (15.6)
<0.0001*
1
-
0.001*
61 (19.0)
<0.0001*
1
-
0.014*
Present
19 (5.6)
11 (57.9)
3.463
1.661-7.222
9 (47.4)
2.562
1.208-5.4
34
Mitotic rate
(per 10 high
power fields)
< 10
173 (50.9)
27 (15.6)
0.167
-
-
30 (17.3)
0.0823
-
-
>= 10
167 (49.1)
34 (20.4)
-
-
40 (24.0)
-
-
NDN ex-
pression
Low
170 (50.0)
17 (10.0)
0.0002*
1
-
0.032*
24 (14.1)
0.0027*
1
-
0.033*
High
170 (50.0)
44 (25.9)
1.893
1.058-3.385
46 (27.1)
1.731
1.044-2.8
69
* Statistically significant.
Table 4. Univariate log-rank and multivariate analyses for Disease-specific and Metastasis-free Survivals in urinary bladder urothelial
carcinoma.
Parameter
Category
Case No.
(%)
Disease-specific Survival
Metastasis-free Survival
Univariate analysis
Multivariate analysis
Univariate analysis
Multivariate analysis
No. of
event (%)
p-value
R.R.
95% C.I.
p-value
No. of
event (%)
p-value
R.R.
95% C.I.
p-value
Gender
Male
216 (73.2)
41 (19.0)
0.4446
-
-
-
60 (27.8)
0.2720
-
-
-
Female
79 (26.8)
11 (13.9)
-
-
-
16 (20.3)
-
-
-
Age (years)
< 65
121 (41.0)
17 (14.0)
0.1136
-
-
-
31 (25.6)
0.6875
-
-
-
≥ 65
174 (59.0)
35 (20.1)
-
-
-
45 (25.9)
-
-
-
Primary tumor
(pT)
pTa
84 (28.5)
1 (1.4)
<0.0001*
1
-
<0.0001*
4 (4.8)
<0.0001*
1
-
0.011*
pT1
88 (29.8)
9 (10.2)
3.817
1.748-8.333
23 (26.1)
4.631
1.311-16.364
pT2-pT4
123 (41.7)
42 (34.1)
21.277
2.823-200.00
49 (39.8)
6.688
1.889-23.677
Nodal metastasis
Negative (pN0)
266 (90.2)
41 (15.4)
0.0002*
1
-
0.387
61 (22.9)
<0.0001*
1
-
0.037*
Positive
(pN1-pN3)
29 (9.8)
11 (37.9)
1.364
0.675-2.758
15 (51.7)
1.939
1.042-3.607
Histological grade
Low grade
56 (19.0)
2 (3.6)
0.0013*
1
-
0.889
5 (8.9)
0.0007*
1
-
0.711
High grade
239 (81.0)
50 (20.9)
0.895
0.188-4.266
71 (29.7)
0.809
0.264-2.481
Vascular invasion
Absent
246 (83.4)
37 (15.0)
0.0024*
1
-
0.185
54 (22.0)
0.0001*
1
-
0.835
Present
49 (16.6)
15 (30.6)
0.629
0.317-1.249
22 (44.9)
1.067
0.580-1.964
Perineural invasion
Absent
275 (93.2)
44 (16.0)
0.0001*
1
-
0.131
66 (24.0)
0.0007*
1
-
0.347
Present
20 (6.8)
8 (40.0)
1.908
0.824-4.420
10 (50.0)
1.429
0.679-3.009
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
310
Parameter
Category
Case No.
(%)
Disease-specific Survival
Metastasis-free Survival
Univariate analysis
Multivariate analysis
Univariate analysis
Multivariate analysis
No. of
event (%)
p-value
R.R.
95% C.I.
p-value
No. of
event (%)
p-value
R.R.
95% C.I.
p-value
Mitotic rate (per 10
high power fields)
< 10
139 (47.1)
12 (8.6)
<0.0001*
1
-
0.011*
23 (16.5)
<0.0001*
1
-
0.019*
>= 10
156 (52.9)
40 (25.6)
2.409
1.219-4.761
53 (34.0)
1.871
1.109-3.156
NDN expression
Low
147 (49.8)
10 (6.8)
<0.0001*
1
-
0.012*
21 (14.3)
<0.0001*
1
-
0.021*
High
148 (50.2)
42 (28.4)
2.477
1.220-5.028
55 (37.2)
1.852
1.098-3.123
* Statistically significant.
Figure 2. Quantitative real-time PCR (qPCR) analysis showed that NDN mRNA expression was significantly increased stepwise in both urothelial carcinomas of the
upper urinary tract (left panel) and urinary bladder (right panel) along with more advanced primary pT status, from pTa to pT1 and pT1 to pT2-4 (all P<0.05).
Figure 3. NDN immunostaining on representative sections revealed stepwise increase of NDN expression from non-tumoral urothelium (A), non-invasive (B), superficially
infiltrating (pT1) (C) and deeply infiltrating (pT2-4) urothelial carcinoma (D).
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
311
Figure 4. Kaplan-Meier plots disclosed the predictive value of NDN immunoreactivity for disease-specific survival (DSS) and metastasis-free survival (MeFS) in both groups
of UTUC (A and B for DSS and MeFS, respecitvely) and UBUC (C and D for DSS and MeFS, respectively) (all P<0.005).
Discussion
Urothelial carcinoma is prevalent worldwide
and has high recurrence rates [27]. Pathological stag-
ing is still the most decisive prognosticator of out-
comes for bladder cancer, where five-year survival
rates are approximately 60% to 85% for T2 bladder
cancers after cystectomy, but decrease rapidly to 40%
to 60% for T3 tumors [28]. In spite of the introduction
of cisplatin-based combination chemotherapy, the
prognosis is still poor for advanced stage bladder
cancers; so far bladder cancer is ranked as the thir-
teenth most common cause of cancer death in the
world (GLOBOCAN 2012 http://globocan.iarc.fr).
Therefore, developing more potent therapeutic regi-
mens is of crucial importance for high-risk patients.
Every cancer has ten hallmarks: sustaining pro-
liferative signaling, evading growth suppressors,
avoiding immune destruction, enabling replicative
immortality, tumor-promoting inflammation, acti-
vating invasion and metastasis, inducing angiogene-
sis, genome instability and mutation, resisting cell
death, and deregulating cellular energetics [29].
Among these characteristics, sustaining proliferative
signaling, that is, cell growth dysregulation, serves as
the first step of oncogenesis.
The maternally imprinted gene, NDN, is associ-
ated with Prader-Willi syndrome [30]. Necdin, en-
coded by the NDN gene, is a 325-amino acid protein
belonging to the MAGE family [18, 19]. The function
of Necdin protein is similar to Rb, which has been
shown to interact with viral oncogenes such as simian
virus 40 (SV40) large T antigen and adenovirus E1A,
as well as E2F1 and p53 [31, 32]. Nevertheless, Necdin
also bears oncogenic properties. Necdin’s inhibitory
effect on p53-mediated growth arrest and suppressive
impact on p53-dependent apoptosis have also been
demonstrated to be oncogenic functions [33, 34]. In
line with this, there have been divergent results for
Necdin expression in different cancers of each study.
The scarcity of Necdin expression in brain tumor cell
lines [35], as well as its decreased expression in mel-
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
312
anomas [36] and bladder cancer cell lines and tumors
[21] suggest NDN may be a tumor suppressor gene.
Conversely, upregulation and loss of imprinting of
Necdin in pancreatic cancers have been demonstrated
by more recent studies [37, 38]. The paradoxical re-
sults of experiments designed to recognize the role of
Necdin in oncogenesis indicate that it has complex
functions, which may be due to different microenvi-
ronments and cellular contexts [34]. Our current in-
vestigation proves the possible oncogenic role of NDN
in UCs, which is not consistent with a previous study
that found NDN was downregulated in bladder can-
cer cell lines and tumors [21]. A study by Chapman, et
al. [21] first observed that NDN was induced to be
downregulated in normal human urothelial cells
transducted by hTERT, the catalytic subunit of te-
lomerase. After that, the authors found NDN tran-
scripts were downregulated in 26 of 28 (92.9%) blad-
der cancer cell lines and 35 of 58 (60%) bladder cancer
tumors. However, the researchers neither correlated
the NDN transcription levels with clinicopathological
significance nor analyzed their association with sur-
vival. In addition, to the best of our knowledge, the
current investigation is the first to inspect the rela-
tionship between NDN expression of both mRNA and
protein levels, along with clinical outcomes in UTUC.
Conclusion
In sum, our work demonstrates that NDN gene
and NDN protein (a.k.a. Necdin) plays an influential
role in tumor progression of UCs. High expression of
Necdin in both UTUC and UBUC is associated with
worse clinicopathological parameters. Overexpres-
sion of Necdin is also an independent prognosticator
of inferior DSS and MeFS in both groups. Further re-
search to elucidate the details of the biological role of
the NDN gene and Necdin in carcinogenesis of UC is
essential for exploring the potential of NDN-targeted
therapy for UCs, as we illustrated the hopeful targets
for new strategies for UC therapy lately [39-41].
Supplementary Material
Supplementary methods and Figure S1.
http://www.jcancer.org/v07p0304s1.pdf
Acknowledgements
The authors are grateful to the BioBank at Chi
Mei Medical Center for providing tumor samples.
Grant Support
This work was sponsored by E-DA Hospital
(EDAHP104014 to I-W Chang), Taiwan Ministry of
Science and Technology (NSC99-2320-B-384-001-MY2
and NSC101-2320-B-384 -001 -MY3), Ministry of
Health and Welfare (DOH102-TD-M-111-102001 and
MOHW104- TDU-B-212124-003, from Health and
Welfare surcharge on tobacco products, to C-F Li and
W-J Wu) and Kaohsiung Medical University
(KMU-TP103G01, KMU-TP103G00, KMU-TP103G04,
KMU-TP103G05) This study was also supported by
grants from Kaohsiung Medical University “Aim for
the Top Universities (KMU-TP103E19).
Competing Interests
The authors have declared that no competing
interest exists.
References
1. Eble JN, Sauter G, Epstein JI, Sesterhenn IA. World Health Organization
Classification of Tumours. Pathology and Genetics of Tumours of the Urinary
System and Male Genital Organs. Lyon, Fance: International Agency for
Research on Cancer (IARC) press; 2004.
2. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin.
2014;64:9-29.
3. Raman JD, Messer J, Sielatycki JA, Hollenbeak CS. Incidence and survival of
patients with carcinoma of the ureter and renal pelvis in the USA, 1973-2005.
BJU Int. 2011;107:1059-64.
4. Chou YH, Huang CH. Unusual clinical presentation of upper urothelial
carcinoma in Taiwan. Cancer. 1999;85:1342-4.
5. Yang MH, Chen KK, Yen CC, et al. Unusually high incidence of upper urinary
tract urothelial carcinoma in Taiwan. Urology. 2002;59:681-7.
6. Tan LB, Chen KT, Guo HR. Clinical and epidemiological features of patients
with genitourinary tract tumour in a blackfoot disease endemic area of
Taiwan. BJU Int. 2008;102:48-54.
7. McLaughlin JK, Silverman DT, Hsing AW, et al. Cigarette smoking and
cancers of the renal pelvis and ureter. Cancer Res. 1992;52:254-7.
8. Pommer W, Bronder E, Klimpel A, Helmert U, Greiser E, Molzahn M.
Urothelial cancer at different tumour sites: role of smoking and habitual intake
of analgesics and laxatives. Results of the Berlin Urothelial Cancer Study.
Nephrol Dial Transplant. 1999;14:2892-7.
9. Shinka T, Miyai M, Sawada Y, Inagaki T, Okawa T. Factors affecting the
occurrence of urothelial tumors in dye workers exposed to aromatic amines.
Int J Urol. 1995;2:243-8.
10. Stewart JH, Hobbs JB, McCredie MR. Morphologic evidence that
analgesic-induced kidney pathology contributes to the progression of tumors
of the renal pelvis. Cancer. 1999;86:1576-82.
11. Debelle FD, Vanherweghem JL, Nortier JL. Aristolochic acid nephropathy: a
worldwide problem. Kidney Int. 2008;74:158-69.
12. Laing C, Hamour S, Sheaff M, Miller R, Woolfson R. Chinese herbal uropathy
and nephropathy. Lancet. 2006;368:338.
13. Dragicevic D, Djokic M, Pekmezovic T, et al. Survival of patients with
transitional cell carcinoma of the ureter and renal pelvis in Balkan endemic
nephropathy and non-endemic areas of Serbia. BJU Int. 2007;99:1357-62.
14. Catto JW, Yates DR, Rehman I, et al. Behavior of urothelial carcinoma with
respect to anatomical location. J Urol. 2007;177:1715-20.
15. Zhang Z, Furge KA, Yang XJ, Teh BT, Hansel DE. Comparative gene
expression profiling analysis of urothelial carcinoma of the renal pelvis and
bladder. BMC Med Genomics. 2010;3:58.
16. Croce CM. Oncogenes and cancer. N Engl J Med. 2008;358:502-11.
17. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57-70.
18. Barker PA, Salehi A. The MAGE proteins: emerging roles in cell cycle
progression, apoptosis, and neurogenetic disease. J Neurosci Res.
2002;67:705-12.
19. Maruyama K, Usami M, Aizawa T, Yoshikawa K. A novel brain-specific
mRNA encoding nuclear protein (necdin) expressed in neurally differentiated
embryonal carcinoma cells. Biochem Biophys Res Commun. 1991;178:291-6.
20. Hayashi Y, Matsuyama K, Takagi K, Sugiura H, Yoshikawa K. Arrest of cell
growth by necdin, a nuclear protein expressed in postmitotic neurons.
Biochem Biophys Res Commun. 1995;213:317-24.
21. Chapman EJ, Kelly G, Knowles MA. Genes involved in differentiation, stem
cell renewal, and tumorigenesis are modulated in telomerase-immortalized
human urothelial cells. Mol Cancer Res. 2008;6:1154-68.
22. Huang WW, Huang HY, Liao AC, et al. Primary urothelial carcinoma of the
upper tract: important clinicopathological factors predicting bladder
recurrence after surgical resection. Pathol Int. 2009;59:642-9.
23. Wu LC, Chen LT, Tsai YJ, et al. Alpha-methylacyl coenzyme A racemase
overexpression in gallbladder carcinoma confers an independent prognostic
indicator. J Clin Pathol. 2012;65:309-14.
24. Budwit-Novotny DA, McCarty KS, Cox EB, et al. Immunohistochemical
analyses of estrogen receptor in endometrial adenocarcinoma using a
monoclonal antibody. Cancer Res. 1986;46:5419-25.
Journal of Cancer 2016, Vol. 7
http://www.jcancer.org
313
25. McClelland RA, Finlay P, Walker KJ, et al. Automated quantitation of
immunocytochemically localized estrogen receptors in human breast cancer.
Cancer Res. 1990;50:3545-50.
26. Liang PI, Wang YH, Wu TF, et al. IGFBP-5 overexpression as a poor prognostic
factor in patients with urothelial carcinomas of upper urinary tracts and
urinary bladder. J Clin Pathol. 2013;66:573-82.
27. Rodriguez-Alonso A, Pita-Fernandez S, Gonzalez-Carrero J, Nogueira-March
JL. Multivariate analysis of survival, recurrence, progression and developmen t
of mestastasis in T1 and T2a transitional cell bladder carcinoma. Cancer.
2002;94:1677-84.
28. Townsend CM JR, Evers BM, Mattox KL. Sabiston Textbook of Surgery: The
Biological Basis of Modern Surgical Practice. 16th ed. Philadelphia, USA:
Saunders; 2002.
29. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell.
2011;144:646-74.
30. Jay P, Rougeulle C, Massacrier A, et al. The human necdin gene, NDN, is
maternally imprinted and located in the Prader-Willi syndrome chromosomal
region. Nat Genet. 1997;17:357-61.
31. Taniura H, Matsumoto K, Yoshikawa K. Physical and functional interactions
of neuronal growth suppressor necdin with p53. J Biol Chem.
1999;274:16242-8.
32. Taniura H, Taniguchi N, Hara M, Yoshikawa K. Necdin, a postmitotic
neuron-specific growth suppressor, interacts with viral transforming proteins
and cellular transcription factor E2F1. J Biol Chem. 1998;273:720-8.
33. Hasegawa K, Yoshikawa K. Necdin regulates p53 acetylation via Sirtuin1 to
modulate DNA damage response in cortical neurons. J Neurosci.
2008;28:8772-84.
34. Lafontaine J, Rodier F, Ouellet V, Mes-Masson AM. Necdin, a p53-target gene,
is an inhibitor of p53-mediated growth arrest. PLoS One. 2012;7:e31916.
35. Aizawa T, Maruyama K, Kondo H, Yoshikawa K. Expression of necdin, an
embryonal carcinoma-derived nuclear protein, in developing mouse brain.
Brain Res Dev Brain Res. 1992;68:265-74.
36. Hoek K, Rimm DL, Williams KR, et al. Expression profiling reveals novel
pathways in the transformation of melanocytes to melanomas. Cancer Res.
2004;64:5270-82.
37. Tan AC, Jimeno A, Lin SH, et al. Characterizing DNA methylation patterns in
pancreatic cancer genome. Mol Oncol. 2009;3:425-38.
38. Wang L, Liu HL, Li Y, Yuan P. Proteomic analysis of pancreatic intraepithelial
neoplasia and pancreatic carcinoma in rat models. World J Gastroenterol.
2011;17:1434-41.
39. Chang IW, Liang PI, Li CC, et al. HAS3 underexpression as an indicator of
poor prognosis in patients with urothelial carcinoma of the upper urinary tract
and urinary bladder. Tumour Biol. 2015;36:5441-50.
40. Chang IW, Lin VC, He HL, et al. CDCA5 overexpression is an indicator of
poor prognosis in patients with urothelial carcinomas of the upper urinary
tract and urinary bladder. Am J Transl Res. 2015;7:710-22.
41. Li CF, Wu WJ, Wu WR et al. The cAMP responsive element binding protein 1
transactivates epithelial membrane protein 2, a potential tumor suppressor in
the urinary bladder urothelial carcinoma. Oncotarget 2015;6:9220-39.
... BCAT1 has been identified as being highly expressed in a variety of cancers, such as gliomas, gastric cancer, and hepatocellular carcinoma, and has been frequently reported as a promoter in the occurrence and development of these cancers. [6][7][8][9][10] Moreover, in glioma, 11 prostate cancer, 12 and urothelial cancer, 13 it has been demonstrated that the prognosis of tumor patients with upregulated BCAT1 expression was poor. Nevertheless, the comprehensive potential mechanism and clinical significance of BCAT1 in pan-cancer have not previously been reported, and more efforts are needed to address this lack. ...
... BCAT1 expression showed conspicuous clinical significance in cancers. Previously, several studies have revealed the risk role of high expression of BCAT1 in the prognosis of patients with glioma, 11 prostate cancer, 12 urothelial cancer, 13 and HNSCC. 42 This scenario was observed in ACC, BLCA, BRCA, KIRC, LGG, LIHC, PAAD, and UVM via analysis of relevance between BCAT1 expression and OST. ...
Article
Full-text available
Background: Although branched chain amino acid transaminase 1 (BCAT1) has been identified to play an essential role in multiple tumors, no studies on its role in pan-cancer have been consulted before. Methods: The study comprehensively analyzes the expression, potential mechanisms, and clinical significance of BCAT1 in pan-cancer through utilizing 16,847 samples, providing novel clues for the treatment of cancers. A Kruskal-Wallis test and the Wilcoxon rank-sum and signed-rank tests were applied to investigate diverse BCAT1 expression between various groups (e.g., cancer tissues versus normal tissues). Spearman's rank correlation coefficient was used in all correlation analyses in the study. Cox analyses and Kaplan-Meier curves were utilized to identify the prognosis significance of BCAT1 expression in cancers. The significance of BCAT1 expression in differentiating cancer and non-cancer tissues was explored via the area under the receiver operating characteristic curves (AUC). Results: The differential expression of BCAT1 was detected in various cancers (p < 0.05), which is relevant to some DNA methyltransferases expression. BCAT1 expression was associated with mismatch repair gene expression, immune checkpoint inhibitors expression, microsatellite instability, and tumor mutational burden in some cancers, indicating its potential in immunotherapy. BCAT1 expression showed prognosis significance and played a risk role in multiple cancers (hazard ratio > 0, p < 0.05). BCAT1 expression also demonstrated conspicuous ability to distinguish some cancers tissues from their normal tissues (AUC > 0.7), indicating its potential to detect cancers. Further analyses on head and neck squamous cell carcinoma certified upregulated BCAT1 expression at both mRNA and protein levels in this disease based on in-house tissue microarrays and multicenter datasets. Conclusions: For the first time, the research comprehensively demonstrates the overexpression of BCAT1 in pan-cancer, which improves the understanding of the pathogenesis of BCAT1 in pan-cancer. Upregulated BCAT1 expression represented a poor prognosis for cancers patients, and it serves as a potential marker for cancer immunotherapy.
... The branched-chain amino acids (BCAAs) leucine, isoleucine, and valine are essential amino acids that are converted to their respective ketoacids (BCKAs) via transamination of α-ketoglutarate (α-KG) to produce glutamate, catalyzed by Branched Chain Aminotransferase, which exists as cytosolic (BCAT1) and mitochondrial (BCAT2) isoforms ( Figure 1A). Upregulation of BCAT1 has been observed in various cancers, [1][2][3][4][5][6][7][8][9][10][11][12][13] where in some cases it has been linked to increased cell proliferation and invasion. However, this is dependent on tissue of origin, for example, BCAT1 has been shown to be required for tumor formation in non-small-cell lung cancer but not in pancreatic ductal adenocarcinoma. ...
Article
Full-text available
Background Branched-chain aminotransferase 1 (BCAT1) has been proposed to drive proliferation and invasion of isocitrate dehydrogenase (IDH) wild-type glioblastoma cells. However, the Cancer Genome Atlas (TCGA) dataset shows considerable variation in the expression of this enzyme in glioblastoma. The aim of this study was to determine the role of BCAT1 in driving the proliferation and invasion of glioblastoma cells and xenografts that have widely differing levels of BCAT1 expression and the mechanism responsible. Methods The activity of BCAT1 was modulated in IDH wild-type patient-derived glioblastoma cell lines, and in orthotopically implanted tumors derived from these cells, to examine the effects of BCAT1 expression on tumor phenotype. Results In cells with constitutively high BCAT1 expression and a glycolytic metabolic phenotype, inducible shRNA knockdown of the enzyme resulted in reduced proliferation and invasion by increasing the concentration of α-ketoglutarate, leading to reduced DNA methylation, HIF-1α destabilization, and reduced expression of the transcription factor Forkhead box protein M1 (FOXM1). Conversely, overexpression of the enzyme increased HIF-1α expression and promoted proliferation and invasion. However, in cells with an oxidative phenotype and very low constitutive expression of BCAT1 increased expression of the enzyme had no effect on invasion and reduced cell proliferation. This occurred despite an increase in HIF-1α levels and could be explained by decreased TCA cycle flux. Conclusions There is a wide variation in BCAT1 expression in glioblastoma and its role in proliferation and invasion is dependent on tumor subtype.
... Studies have reported that BCAT1 was overexpressed in UC and has implications for the regulation of amino acid metabolism in urinary bladder urothelial cancer (UBUC), which is extremely important for the proliferation, spread and invasion of cancer cells. This indicates that BCAT1 are important in the tumor progression of upper tract urothelial cancer (UTUC) and UBUC (48). ...
Article
Full-text available
It is well known that the enzyme catalyzes the first step of branched-chain amino acid (BCAA) catabolism is branched-chain amino transferase (BCAT), which is involved in the synthesis and degradation of leucine, isoleucine and valine. There are two main subtypes of human branched chain amino transferase (hBCAT), including cytoplasmic BCAT (BCAT1) and mitochondrial BCAT (BCAT2). In recent years, the role of BCAT in tumors has attracted the attention of scientists, and there have been continuous research reports that BCAT plays a role in the tumor, Alzheimer’s disease, myeloid leukaemia and other diseases. It plays a significant role in the growth and development of diseases, and new discoveries about this gene in some diseases are made every year. BCAT usually promotes cancer proliferation and invasion by activating the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin pathway and activating Wnt/β-catenin signal transduction. This article reviews the role and mechanism of BCAT in different diseases, as well as the recent biomedical research progress. This review aims to make a comprehensive summary of the role and mechanism of BCAT in different diseases and to provide new research ideas for the treatment, prognosis and prevention of certain diseases.
... After a meal Fasting Starvation Severe starvation Level of BCAA oxidation numerous cancers (Fig. 3) and correlates with poor tumor outcomes [48][49][50][51]. BCAT1 expression in glioblastoma, an aggressive type of cancer that can occur in the brain or spinal cord, is specific to those carrying wild-type isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2) [52]. ...
Article
Full-text available
Branched-chain amino acids (BCAAs), isoleucine, leucine and valine, are essential amino acids with vital roles in protein synthesis and energy production. We reviewed the fundamentals of BCAA metabolism in advanced cancer patients. BCAAs and various catabolic products act as signalling molecules, which activate mechanisms ranging from protein synthesis to insulin secretion. Recently, BCAA metabolism has been suggested to contribute to cancer progression. Of particular interest is the modulation of the mTOR activity by BCAAs. There are likely multiple pathways involved in BCAA metabolism implicated in carcinogenesis. Understanding the mechanism(s) underlying altered BCAAs metabolism will significantly advance the current understanding of nutrient involvement in carcinogenesis and direct future studies to unravel the significance of BCCA metabolites in tumor development and progression.
... Recent data have demonstrated that BCAT1 is overexpressed in a broad range of malignancies. The malignancies that carry overexpressed BCAT1 include glioblastoma carrying wildtype isocitrate dehydrogenase 1 [4], non-small cell lung carcinoma [12], epithelial ovarian cancer [5], myeloid leukemia [6], urothelial carcinomas [18], hepatocellular carcinoma [19], and gastric cancer [9] among others. A caveat is that not all cancers carry overexpressed BCAT1. ...
Article
Malignant melanoma has a high mutational rate. As a result, resistance to current therapies is common. Consequently, there is an unmet medical need to develop novel therapies. Recent data suggest that branched-chain amino acid transaminase 1 (BCAT1) is overexpressed in multiple cancers, and such overexpressed BCAT1 is necessary for individual cancer progression. Therefore, BCAT1 appears to be a good target in cancer treatment. Additionally, because its expression in healthy tissues is highly restricted in adults and is limited to the brain, ovary, and placenta, BCAT1 is especially an ideal target in cancer therapies. Currently, the function of BCAT1 in malignant melanoma has not been demonstrated. Therefore, we investigated the role of BCAT1 in the proliferation and migration of malignant melanomas using human samples and mouse malignant B16 melanoma cell line. Our data showed that BCAT1 was overexpressed in malignant melanoma tissues both in humans and mice. Besides, BCAT1 knockdown suppressed melanoma cell proliferation and migration, which was associated with reduced oxidative phosphorylation. Collectively, our data indicate that BCAT1 is a promising therapeutic target for the treatment of malignant melanomas.
... Numerous studies have revealed that BCAT1 is overexpressed in several malignancies; more importantly, it is in involved in various malignant phenotypes, including changes in cell proliferation, cell cycle progression, differentiation, and apoptosis . Here, we verified the higher expression of BCAT1 in GC tumor tissues compared with normal tissues in both TCGA data and clinical samples, similar to the results of previous studies (Zhou et al., 2013;Chang et al., 2016). In addition, the bioinformatics analysis, including gene expression profiling as well as the associated GO and KEGG analyses and GSEA, showed that BCAT1 might affect invasion-and proliferation-related pathways, including ECM-receptor interaction, cytokinecytokine receptor interaction, focal adhesion, and the PI3K-Akt signaling pathway, which all have critical roles in mediating survival signals in cancer (Martini et al., 2014;Bao et al., 2019). ...
Article
Full-text available
Background Focusing on antiangiogenesis may provide promising choices for treatment of gastric cancer (GC). This study aimed to investigate the mechanistic role of BCAT1 in the pathogenesis of GC, particularly in angiogenesis.Methods Bioinformatics and clinical samples analysis were used to investigate the expression and potential mechanism of BCAT1 in GC. BGC823 cells with BCAT1 overexpression or silencing were induced by lentiviral transduction. Cell phenotypes and angiogenesis were evaluated. The relevant proteins were quantized by Western blotting, immunohistochemistry, or immunofluorescence. Xenograft models were constructed to confirm the role of BCAT1 in vivo.ResultsBCAT1 was overexpressed in GC patients and associated with lower survival. BCAT1 expression was correlated with proliferation-, invasion-, or angiogenesis-related markers expression and pathways. Silencing BCAT1 expression suppressed cell viability, colony formation, cycle progression, invasion, and angiogenesis of BGC823 cells, as well as the tumor growth of xenograft models, whereas overexpressing BCAT1 had the opposite results both in vitro and in vivo. Bioinformatics analysis and Western blotting demonstrated that BCAT1 activated the PI3K/AKT/mTOR pathway. The addition of LY294002 reversed the tumor growth induced by BCAT1 overexpression, further verifying this mechanism.ConclusionBCAT1 might act as an oncogene by facilitating proliferation, invasion, and angiogenesis through activation of the PI3K/AKT/mTOR pathway. This finding could aid the optimization of antiangiogenesis strategies.
Preprint
Full-text available
Objective Long noncoding RNAs (lncRNAs) are significant regulators in gastric cancer(GC); However, studies of their mechanisms of action are needed to determine their clinical value. In this study, we investigated the effects and mechanism of action of THUMPD3-AS1 in GC. Methods Candidate lncRNAs and mRNAs were getted from The Cancer Genome Atlas, revealing the differential expression and prognostic significance of THUMPD3-AS1-BCAT1 in GC. qRT-PCR was performed to detect THUMPD3-AS1 levels in GC samples and cell lines. CCK8, scratch wound healing, and Transwell assays as well as experiments in vivo were conducted to evaluate the function of THUMPD3-AS1 in GC. Related genes were analysed to detect interactions between THUMPD3-AS1, BCAT1, and miR-1297. Results THUMPD3-AS1 levels were significantly elevated in GC and were positively correlated with poor prognosis. Functionally, THUMPD3-AS1 promoted GC cell proliferation, migration, invasion, and epithelial–mesenchymal transition (EMT) and induced tumour growth in vivo. THUMPD3-AS1 regulated BCAT1 by competitively binding to miR-1297; further analyses revealed that both THUMPD3-AS1 and miR-1297 can interact with BCAT1. Conclusions These findings demonstrate that THUMPD3-AS1 promotes GC cell invasion and EMT via the miR-1297/BCAT1 pathway, suggesting that THUMPD3-AS1 is a novel biomarker and therapeutic target for GC.
Article
Full-text available
Branched-chain amino acid aminotransferase (BCAT) catalyzes bidirectional transamination in the cell between branched-chain amino acids (BCAAs; valine, leucine, and isoleucine) and branched-chain α-keto acids (BCKAs; α-ketoisovalerate, α-ketoisocaproate, and α-keto-β-methylvalerate). Eukaryotic cells contain two types of paralogous BCATs: mitochondrial BCAT (BCATm) and cytosolic BCAT (BCATc). Both isozymes have identical enzymatic functions, so they have long been considered to perform similar physiological functions in the cells. However, many studies have gradually revealed the differences in physiological functions and regulatory mechanisms between them. In this article, we present overviews of BCATm and BCATc in both yeast and human. We also introduce BCAT variants found natively or constructed artificially, which could have significant implications for research into the relationship between the primary structures and protein functions of BCATs. Key points • BCAT catalyzes bidirectional transamination in the cell between BCAAs and BCKAs. • BCATm and BCATc are different in the metabolic roles and regulatory mechanisms. • BCAT variants offer insight into a relationship between the structure and function.
Article
To make new infectious particles, all viruses must manipulate host cell metabolism to secure sufficient availability of biomolecules and energy — a phenomenon now known as metabolic reprogramming. Numerous observations of this has already been made for a range of viruses with each type of virus seemingly applying its own unique tactics to accomplish this unifying goal. In this light, metabolic reprogramming of the infected cell is largely beneficial to the virus and not to the host. On the other hand, virus-induced metabolic reprogramming represents a transformed self with distorted cellular and extracellular levels of distinct metabolites and metabolic by-products. This review briefly outlines current knowledge of virus-induced metabolic reprogramming, discusses how this could be sensed by the infected host to initiate anti-viral programs, and presents examples of innate anti-viral mechanisms of the host that target the availability of biomolecules to block viral replication.
Article
Full-text available
In this study, we report that EMP2 plays a tumor suppressor role by inducing G2/M cell cycle arrest, suppressing cell viability, proliferation, colony formation/anchorage-independent cell growth via regulation of G2/M checkpoints in distinct urinary bladder urothelial carcinoma (UBUC)-derived cell lines. Genistein treatment or exogenous expression of the cAMP responsive element binding protein 1 (CREB1) gene in different UBUC-derived cell lines induced EMP2 transcription and subsequent translation. Mutagenesis on either or both cAMP-responsive element(s) dramatically decreased the EMP2 promoter activity with, without genistein treatment or exogenous CREB1 expression, respectively. Significantly correlation between the EMP2 immunointensity and primary tumor, nodal status, histological grade, vascular invasion and mitotic activity was identified. Multivariate analysis further demonstrated that low EMP2 immunoexpression is an independent prognostic factor for poor disease-specific survival. Genistein treatments, knockdown of EMP2 gene and double knockdown of CREB1 and EMP2 genes significantly inhibited tumor growth and notably downregulated CREB1 and EMP2 protein levels in the mice xenograft models. Therefore, genistein induced CREB1 transcription, translation and upregulated pCREB1(S133) protein level. Afterward, pCREB1(S133) transactivated the tumor suppressor gene, EMP2, in vitro and in vivo. Our study identified a novel transcriptional target, which plays a tumor suppressor role, of CREB1.
Article
Full-text available
Oxidative stress is believed to be one of the important etiologies in carcinogenesis that has not been systemically investigated in urothelial carcinoma (UC). Through data mining from a published transcriptomic database of UC of urinary bladders (UBUCs) (GSE31684), glutathione peroxidase 2 (GPX2) was identified as the most significant downregulated gene among those response to oxidative stress (GO:0006979). We therefore analyze GPX2 transcript and protein expressions and its clinicopathological significance. Real-time RT-PCR assay was used to detect GPX2 mRNA level in 20 fresh UBUC specimens. Immunohistochemistry was used to determine GPX2 protein expression in 340 urothelial carcinomas of upper tracts (UTUCs) and 295 UBUCs with mean/median follow-up of 44.7/38.9 and 30.8/23.1 months, respectively. Its expression status was further correlated with clinicopathological features and evaluated for its impact on disease-specific survival and metastasis-free survival (MeFS). Decrease in GPX2 transcript level was associated with both higher pT and positive nodal status in 20 UBUCs (all p < 0.05). GPX2 protein underexpression was also significantly associated with advanced pT status, nodal metastasis, high histological grade, vascular invasion, and frequent mitoses in both groups of UCs (all p < 0.05). GPX2 underexpression not only predicted dismal DDS and MeFS at univariate analysis, but also implicated worse DDS (UTUC, p = 0.002; UBUC, p = 0.029) and MeFS (UTUC, p = 0.001; UBUC, p = 0.032) in multivariate analysis. GPX2 underexpression is associated with advanced tumor status and implicated unfavorable clinical outcome of UCs, suggesting its role in tumor progression and may serve as a theranostic biomarker of UCs.
Article
Full-text available
Here we show that glioblastoma express high levels of branched-chain amino acid transaminase 1 (BCAT1), the enzyme that initiates the catabolism of branched-chain amino acids (BCAAs). Expression of BCAT1 was exclusive to tumors carrying wild-type isocitrate dehydrogenase 1 (IDH1) and IDH2 genes and was highly correlated with methylation patterns in the BCAT1 promoter region. BCAT1 expression was dependent on the concentration of α-ketoglutarate substrate in glioma cell lines and could be suppressed by ectopic overexpression of mutant IDH1 in immortalized human astrocytes, providing a link between IDH1 function and BCAT1 expression. Suppression of BCAT1 in glioma cell lines blocked the excretion of glutamate and led to reduced proliferation and invasiveness in vitro, as well as significant decreases in tumor growth in a glioblastoma xenograft model. These findings suggest a central role for BCAT1 in glioma pathogenesis, making BCAT1 and BCAA metabolism attractive targets for the development of targeted therapeutic approaches to treat patients with glioblastoma.
Article
Aristolochic acid nephropathy (AAN), a progressive renal interstitial fibrosis frequently associated with urothelial malignancies, was initially reported in a Belgian cohort of more than 100 patients after the intake of slimming pills containing a Chinese herb, Aristolochia fangchi. Although botanicals known or suspected to contain aristolochic acid (AA) were no longer permitted in many countries, several AAN cases were regularly observed all around the world. The incidence of AAN is probably much higher than initially thought, especially in Asia and the Balkans. In Asian countries, where traditional medicines are very popular, the complexity of the pharmacopoeia represents a high risk for AAN because of the frequent substitution of the botanical products by AA-containing herbs. In the Balkan regions, the exposure to AA found in flour obtained from wheat contaminated with seeds of Aristolochia clematitis could be responsible for the so-called Balkan-endemic nephropathy. Finally, despite the Food and Drug Administration's warnings concerning the safety of botanical remedies containing AA, these herbs are still sold via the Internet.
Article
Aims: Urothelial carcinoma (UC) is the most common tumor involving upper urinary tract (UTUC) and urinary bladder (UBUC) whose molecular survival determinants remains obscured. By computerizing a public transcriptomic database of UBUCs (GSE32894), we identified cell division cycle associated 5 (CDCA5) as the most significantly upregulated gene among those associated with G1-S transition of the mitotic cell cycle (GO:0000082). We therefore analyzed the clinicoptaological significance of CDCA5 expression in our well-characterized UC cohort. Methods and results: Quantigene assay was used to detect CDCA5 transcript levels in 36 UTUCs and 30 UBUCs. We used immunohistochemistry evaluated by H-scores to determine CDCA5 protein expression in 295 UBUCs and 340 UTUCs, respectively. CDCA5 expression was further correlated with clinicopathological features and diseasespecific survival (DSS) and metastasis-free survival (MeFS). For both groups of UCs, increments of CDCA5 transcript levels were associated with higher pT status, CDCA5 protein overexpression was also significantly associated with advanced pT status, nodal metastasis, high histological grade, vascular invasion, and frequent mitoses. CDCA5 overexpression was predictive for worse DSS and MeFS in univariate and multivariate analysis. Conclusions: CDCA5 overexpression is associated with advanced clinical features of UC, suggesting its potential value as a prognostic biomarker and a novel therapeutic target.
Article
Locally advanced rectal cancers are currently treated with neoadjuvant concurrent chemoradiotherapy (CCRT) followed by surgery, but risk stratification and final outcomes remain suboptimal. In this study, we identify and validate targetable metabolic drivers relevant to the prognosis of patients with rectal cancer treated with CCRT. Using a published transcriptome of rectal cancers, we found that asparagine synthetase (ASNS) gene significantly predicted the response to CCRT. From 172 patients with rectal cancer, the expression levels of ASNS, using immunohistochemistry assays, were further evaluated in tumor specimens initially obtained by using colonoscopy. Expression levels of ASNS were further correlated with major clinicopathological features and clinical survivals in this valid cohort. ASNS deficiency was significantly related to advanced posttreatment tumor (T3, T4; P = .015) and nodal status (N1, N2; P = .004) and inferior tumor regression grade (P < .001). In survival analyses, ASNS deficiency was significantly associated with shorter local recurrence-free survival (LRFS; P = .0039), metastasis-free survival (MeFS; P = .0001), and disease-specific survival (DSS; P = .0006). Furthermore, ASNS deficiency was independently predictive of worse outcomes for MeFS (P = .012, hazard ratio = 3.691) and DSS (P = .022, hazard ratio = 2.845), using multivariate analysis. ASNS deficiency is correlated with poor therapeutic response and worse survivals in patients with rectal cancer receiving neoadjuvant CCRT. These findings indicate that ASNS is a prognostic factor with therapeutic potential for treating rectal cancer.
Article
The risk stratification and final outcomes in patients with nasopharyngeal carcinomas (NPC) still remain suboptimal. Our principal goals were to identify and validate targetable metabolic drivers relevant to pathogenesis of NPC using a published transcriptome. One prominently downregulated gene regulating amino acid metabolism was found to be argininosuccinate synthetase (ASS1). Attributable to epigenetic DNA methylation, ASS1 deficiency may link to the therapeutic sensitivity to the arginine-depriving agents and promote tumor aggressiveness through its newly identified tumor suppressor function. ASS1 immunohistochemistry was therefore examined in a well-defined cohort of 124 NPC biopsy specimens and in the neck lymph node metastases of another ten independent cases. For the latter, bisulphite pyrosequencing was performed to evaluate the extent of ASS1 gene methylation. ASS1 protein deficiency was identified in 64 of 124 cases (51.6 %), significantly related to T3-T4 status (p = 0.006), and univariately associated with inferior local recurrence-free survival (p = 0.0427), distant metastasis-free survival (DMFS; p = 0.0036), and disease-specific survival (DSS; p = 0.0069). Together with advanced AJCC stages III-IV, ASS1 protein deficiency was also independently predictive of worse outcomes for the DFMS (p = 0.010, hazard ratio = 2.241) and DSS (p = 0.020, hazard ratio = 1.900). ASS1 promoter hypermethylation was detected in eight of ten neck nodal metastatic lesions by bisulphite pyrosequencing and associated with ASS1 protein deficiency (p < 0.001). In summary, ASS1 protein deficiency was seen in approximately a half of NPCs and associated with advanced T classification, DNA methylation, and clinical aggressiveness, consistent with its tumor suppressor role. This aberration may render pegylated arginine deiminase as a promising strategy for ASS1-deficient NPCs.