ArticlePDF Available

KRASG12D-mediated oncogenic transformation of thyroid follicular cells requires long-term TSH stimulation and is regulated by SPRY1

Authors:

Abstract and Figures

KRAS(G12D) can cause lung cancer rapidly, but is not sufficient to induce thyroid cancer. It is not clear whether long-term serum thyroid stimulating hormone (TSH) stimulation can promote KRAS(G12D)-mediated thyroid follicular cell transformation. In the present study, we investigated the effect of long-term TSH stimulation in KRAS(G12D) knock-in mice and the role of Sprouty1 (SPRY1) in KRAS(G12D)-mediated signaling. We used TPO-KRAS(G12D) mice for thyroid-specific expression of KRAS(G12D) under the endogenous KRAS promoter. Twenty TPO-KRAS(G12D) mice were given anti-thyroid drug propylthiouracil (PTU, 0.1% w/v) in drinking water to induce serum TSH and 20 mice were without PTU treatment. Equal number of wild-type littermates (TPO-KRAS(WT)) was given the same treatment. The expression of SPRY1, a negative regulator of receptor tyrosine kinase (RTK) signaling, was analyzed in both KRAS(G12D)-and BRAF(V600E)-induced thyroid cancers. Without PTU treatment, only mild thyroid enlargement and hyperplasia were observed in TPO-KRAS(G12D) mice. With PTU treatment, significant thyroid enlargement and hyperplasia occurred in both TPO-KRAS(G12D) and TPO-KRAS(WT) littermates. Thyroids from TPO-KRAS(G12D) mice were six times larger than TPO-KRAS(WT) littermates. Distinct thyroid histology was found between TPO-KRAS(G12D) and TPO-KRAS(WT) mice: thyroid from TPO-KRAS(G12D) mice showed hyperplasia with well-maintained follicular architecture whereas in TPO-KRAS(WT) mice this structure was replaced by papillary hyperplasia. Among 10 TPO-KRAS(G12D) mice monitored for 14 months, two developed follicular thyroid cancer (FTC), one with pulmonary metastasis. Differential SPRY1 expression was demonstrated: increased in FTC and reduced in papillary thyroid cancer (PTC). The increased SPRY1 expression in FTC promoted TSH-RAS signaling through PI3K/AKT pathway whereas downregulation of SPRY1 by BRAF(V600E) in PTC resulted in both MAPK and PI3K/AKT activation. We conclude that chronic TSH stimulation can enhance KRAS(G12D)-mediated oncogenesis, leading to FTC. SPRY1 may function as a molecular switch to control MAPK signaling and its downregulation by BRAF(V600E) favors PTC development.Laboratory Investigation advance online publication, 6 July 2015; doi:10.1038/labinvest.2015.90.
Content may be subject to copyright.
KRAS
G12D
-mediated oncogenic transformation of
thyroid follicular cells requires long-term TSH stimulation
and is regulated by SPRY1
Minjing Zou
1
, Essa Y Baitei
1
, Roua A Al-Rijjal
1
, Ranjit S Parhar
2
, Futwan A Al-Mohanna
3
, Shioko Kimura
4
,
Catrin Pritchard
5
, Huda BinEssa
1
, Azizah A Alanazi
3
, Ali S Alzahrani
3
, Mohammed Akhtar
6
, Abdullah M Assiri
7
,
Brian F Meyer
1
and Yufei Shi
1
KRAS
G12D
can cause lung cancer rapidly, but is not sufficient to induce thyroid cancer. It is not clear whether long-term
serum thyroid stimulating hormone (TSH) stimulation can promote KRAS
G12D
-mediated thyroid follicular cell
transformation. In the present study, we investigated the effect of long-term TSH stimulation in KRAS
G12D
knock-in mice
and the role of Sprouty1 (SPRY1)inKRAS
G12D
-mediated signaling. We used TPO-KRAS
G12D
mice for thyroid-specific
expression of KRAS
G12D
under the endogenous KRAS promoter. Twenty TPO-KRAS
G12D
mice were given anti-thyroid drug
propylthiouracil (PTU, 0.1% w/v) in drinking water to induce serum TSH and 20 mice were without PTU treatment. Equal
number of wild-type littermates (TPO-KRAS
WT
) was given the same treatment. The expression of SPRY1, a negative
regulator of receptor tyrosine kinase (RTK) signaling, was analyzed in both KRAS
G12D
-and BRAF
V600E
-induced thyroid
cancers. Without PTU treatment, only mild thyroid enlargement and hyperplasia were observed in TPO-KRAS
G12D
mice.
With PTU treatment, significant thyroid enlargement and hyperplasia occurred in both TPO-KRAS
G12D
and TPO-KRAS
WT
littermates. Thyroids from TPO-KRAS
G12D
mice were six times larger than TPO-KRAS
WT
littermates. Distinct thyroid histology
was found between TPO-KRAS
G12D
and TPO-KRAS
WT
mice: thyroid from TPO-KRAS
G12D
mice showed hyperplasia with well-
maintained follicular architecture whereas in TPO-KRAS
WT
mice this structure was replaced by papillary hyperplasia.
Among 10 TPO-KRAS
G12D
mice monitored for 14 months, two developed follicular thyroid cancer (FTC), one with
pulmonary metastasis. Differential SPRY1 expression was demonstrated: increased in FTC and reduced in papillary thyroid
cancer (PTC). The increased SPRY1 expression in FTC promoted TSH-RAS signaling through PI3K/AKT pathway whereas
downregulation of SPRY1 by BRAF
V600E
in PTC resulted in both MAPK and PI3K/AKT activation. We conclude that chronic
TSH stimulation can enhance KRAS
G12D
-mediated oncogenesis, leading to FTC. SPRY1 may function as a molecular switch
to control MAPK signaling and its downregulation by BRAF
V600E
favors PTC development.
Laboratory Investigation (2015) 95, 12691277; doi:10.1038/labinvest.2015.90; published online 6 July 2015
Thyroid cancer is the most common type of endocrine
malignancies and its incidence is rising rapidly in recently
years, especially among women.1Histologically, it can be
classified into papillary thyroid cancer (PTC), follicular
thyroid cancer (FTC), and anaplastic thyroid cancer (ATC).
PTC is the most common type of differentiated thyroid
carcinomas, accounting for more than 80% of thyroid cancer,
and FTC accounts for 15%.2The BRAF
V600E
is the most com-
mon genetic alterations in PTC with overall rate of 44%,35
and RAS (HRAS,KRAS,orNRAS) mutations are found in
about 50% of FTC and can also been found in benign
adenomas.6,7
In the earlier studies, several transgenic mouse models were
created to study the molecular mechanisms of RAS-mediated
transformation of thyroid follicular cells in vivo.810 Both PTC
and FTC were developed from overexpression of a mutant
RAS gene under TG promoter, which may not reflect the
activity of endogenous mutant RAS gene expressed at the
1
Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia;
2
Department of Cell Biology, King Faisal Specialist Hospital and Research
Centre, Riyadh, Saudi Arabia;
3
Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia;
4
Laboratory of Metabolism, Center
for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA;
5
Department of Biochemistry, University of Leicester, Leicester, UK;
6
Department of Pathology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia and
7
Department of Comparative Medicine, King Faisal Specialist
Hospital and Research Centre, Riyadh, Saudi Arabia
Correspondence: Dr Y Shi, MD, MBC 3, Department of Genetics, King Faisal Specialist Hospital and Research Centre, PO Box 3354, Riyadh 11211, Saudi Arabia.
E-mail: yufei@kfshrc.edu.sa
Received 18 February 2015; accepted 5 May 2015
www.laboratoryinvestigation.org | Laboratory Investigation | Volume 95 November 2015 1269
Laboratory Investigation (2015) 95, 12691277
©
2015 USCAP, Inc All rights reserved 0023-6837/15
physiological level. Overexpression of KRAS and HRAS could
also downregulate expression in a dose-dependent manner of
NKX2-1,TG, and TPO genes required for thyroid hormone
synthesis.11,12 Recent knock-in mouse studies have shown that
endogenous expression of either HRAS
G12V
or KRAS
G12D
is
not sufficient to induce thyroid dysfunction and cell trans-
formation under their native promoters.13,14
It is known that TSH stimulates the growth or development
of thyroid cancer and higher serum TSH is associated with
both thyroid cancer incidence and recurrence.15,16 It is not
clear whether long-term TSH stimulation can induce
KRAS
G12D
-mediated thyroid follicular cell transformation. In
the present study, we investigated the potential of long-term
TSH stimulation on thyroid cancer development in KRAS
G12D
knock-in mice targeted to express its oncoprotein in thyroid at
the physiological level. We also studied the role of SPRY1 in
the regulation of KRAS
G12D
- vs BRAF
V600E
-mediated signaling.
MATERIALS AND METHODS
Experimental Animals
LSL-KRAS
G12D
(obtained from The Jackson Laboratory, ME,
USA), LSL-BRAF
V600E
, and TPO-Cre strains have been
described previously.1719 LSL-KRAS
G12D
and LSL-BRAF
V600E
carry a latent mutant allele of KRAS and BRAF, respectively.
Both LSL-KRAS
G12D
and LSL-BRAF
V600E
mice were kept as
heterozygotes. LSL-KRAS
G12D
or LSL-BRAF
V600E
was crossed
with TPO-Cre to generate TPO-KRAS
G12D
or TPO-BRAF
V600E
strain where KRAS
G12D
or BRAF
V600E
is conditionally expressed
in thyroid follicular cells through Cre-mediated deletion
of a floxed transcriptional stop sequence. The resulting TPO-
KRAS
G12D
or TPO-BRAF
V600E
strain expressed mutant
KRAS
G12D
or BRAF
V600E
transcripts at the physiological level
under its endogenous promoter. The study was approved by
the Animal Care and Usage Committee of the institution and
conducted in compliance with the Public Health Service
Guidelines for the Care and Use of Animals in Research.
Genotyping of TPO-KRAS
G12D
or TPO-BRAF
V600E
Strain
Genotyping of TPO-Cre mediated recombination of LSL-
KRAS
G12D
or LSL-BRAF
V600E
targeted allele has been
described previously.17,19 Briefly, the following primers were
used to detect LSL-KRAS
G12D
recombination in mouse tissue:
primer 1, 5-GTCTTTCCCCAGCACAGTGC-3, primer 2,
CTCTTGCCTACGCCACCAGCTC-3, and primer 3, 5-AG
CTAGCCACCATGGCTTGAGTAAGTCTGCA-3. Primer
1+2 detects wild-type allele yielding a product of 622 bp.
Primer 1+2 also detects Cre-recombined KRAS
G12D
allele
(Lox-KRAS
G12D
) yielding a product of 650 bp. This product is
larger than the wild-type allele due to the presence of LoxP
site that remains after Cre-mediated recombination. Primer 2
+3 detects the LSL-KRAS
G12D
allele yielding a product of
500 bp. Multiplex PCR containing three primers was used:
95 °C for 10 min followed by 35 cycles of amplification (95 °C
for 1 min, 60 °C for 1 min, 72 °C for 1 min with final
extension at 72 °C for 10 min). The following primers were
used to detect LSL-BRAF
V600E
recombination: primer A, 5-
AGTCAATCATCCACAGAGACCT-3, primer B: 5-GCTTG
GCTGGACGTAAACTC-3, and primer C, 5-GCCCAGG
CTCTTTATGAGAA-3. Primer A+C detects the wild-type
allele yielding a product of 466 bp. Primer A+C also detects
Cre-recombined allele (Lox-BRAF
V600E
) yielding a product
of 518 bp. Primer B+C detects the LSL-BRAF
V600E
allele
yielding a product of 140 bp. The PCR conditions are at
95 °C for 5 min followed by 35 cycles of amplification
(95 °C for 1 min, 60 °C for 1 min, 72 °C for 1 min with a
final extension at 72 °C for 10 min).
Anti-Thyroid Drug Treatment
TPO-KRAS
G12D
mice (410 weeks of age) were divided into
two groups and given 0.1% (w/v) anti-thyroid drug
propylthiouracil (PTU, Sigma-Aldrich, MO) in drinking
water ad libitum, changed once weekly, to induce serum
TSH. One group (n=10) was observed for 8 months and the
other group (n=10) was observed for 14 months. Twenty
TPO-KRAS
G12D
mice were also divided into two groups (10
in each group) without PTU treatment, and observed for 8
and 14 months, respectively. Forty wild-type mice (TPO-
KRAS
WT
, 10 in each group) were given the same treatment as
TPO-KRAS
G12D
mice.
Thyroid Hormone Measurements
Blood was collected by cardiac puncture. Serum TSH was
measured using MILLIPLEX MAP Mouse Pituitary Magnetic
Bead Panel following the manufacturers instruction (EMD
Millipore Corporation, Billerica, MA, USA). Serum total T4
was measured using MILLIPLEX MAP Steroid/Thyroid
Hormone Magnetic Bead Panel (EMD Millipore Corporation).
Tumor Cell Culture
A PTC tumor from a 4-month-old TPO-BRAF
V600E
mouse
was collected aseptically using blunt dissection and mechani-
cally dissociated by mincing and passaged through a 40-μM
mesh sterile screen, and suspended in DMEM/F12 growth
medium (10% fetal bovine serum, 100 units/ml penicillin,
100 μg/ml streptomycin). Cells were further dissociated by
incubation in the growth medium containing 100 U/ml type I
collagenase (Sigma-Aldrich) and 1.0 U/ml dispase I (Roche
Diagnostics, Indianapolis, IN, USA) at 37 °C rocking water
bath for 60 min. The cell suspension was washed twice and
resuspended in 10 mm culture dish with DMEM/F12 growth
medium containing 2 mU/ml bovine TSH (Sigma-Aldrich) to
establish a BVE cell line (BRAF
V600E
-induced tumor cell line).
The genetic background was confirmed by genotyping.
Quantitative Real-Time Reverse Transcriptase-PCR
Analysis for SPRY1 Expression
Total RNA was isolated from thyroid of TPO-KRAS
WT
mice
treated with PTU, PTC of TPO-BRAF
V600E
, or FTC of TPO-
KRAS
G12D
mice by the quanidinium thiocyanate-phenol-
chloroform method as described previously.20 The integrity of
TSH mediated RAS transformation of thyroid follicular cells
M Zou et al
1270 Laboratory Investigation | Volume 95 November 2015 | www.laboratoryinvestigation.org
RNA was verified by denaturing gel electrophoresis. In all, 2
μg of each total RNA was reverse-transcribed to cDNA using
the Promega RT system (Promega, Madison, WI, USA).
LightCycler DNA Master SYBR Green 1 kit was used for
quantitative real-time PCR analysis.21 The cDNA mix was
diluted 10-fold, and 2 μl of the dilution was used for real-time
PCR. PCR primers for the 114-bp SPRY1 cDNA fragment are
5-GCGGAGGCCGAGGATTT-3(forward) and 5-ATCAC
CACTAGCGAAGTGTGGC-3(reverse). The forward primer
spans the junction of exon 1 and exon 2 over 1.7 kb intron 1
so that contaminated genomic DNA will not be amplified.
The SPRY1 cDNA fragment was verified by DNA sequencing.
The mRNA level of housekeeping gene glyceraldehyde-3-
phosphate dehydrogenase (GAPDH) was used as an internal
control, and a 155-bp PCR product was amplified using the
following two primers: 5-ATGTTCCAGTATGACTCCACT
CACG-3(forward) and 5-GAAGACACCAGTAGACT
CCACGACA-3(reverse). The PCR conditions are 95 °C for
30 s followed by 30 cycles of amplification (95 °C for 10 s, 50 °
C for 5 s, and 72 °C for 10 s). The resulting concentration of
SPRY1 PCR products was normalized by comparison with
GAPDH and was used to determine the relative mRNA level
of SPRY1 in thyroid tumors.
Histology and Immunohistochemistry
Histology and immunohistochemical staining was described
previously.22 Briefly, 4-μm-thick formalin-fixed paraffin-
embedded tissue sections were prepared and stained with
hematoxylin and eosin or SPRY1 (1:50 dilution, Abcam,
Cambridge, MA, USA), p-ERK, or p-AKT antibody (1:50
dilution, Cell Signaling Technology, Danvers, MA, USA).
Histological diagnosis was performed by a thyroid pathologist
(MA) blinded to the genotype and the treatment status of the
animal. DAKO LSAB+kit, HRP was used for immunostaining
(DAKO, Carpinteria, CA, USA). The sections were counter-
stained with Mayer's haematoxylin.
Regulation of TSH Signaling by SPRY1
Mouse SPRY1 cDNA in pCMV6 expression vector was
obtained from OriGene (Rockville, MD, USA). BVE cell line
was transfected with either pCMV6 vector or mSPRY1/
pCMV6 using Lipofectamine (Invitrogen, CA). The culture
medium was changed to DMEM/F12 growth medium 16 h
after transfection, and the transfected cells were cultured for
additional 48 h in the presence or absence of 10 mU/ml
bTSH. The expression of SPRY1, phospho-ERK, and
phospho-AKT was determined by western blot analysis.
Western Blot Analysis
BVE cell line was transfected with mouse SPRY1 cDNA under
the control of CMV promoter (Origen). 40 μg of protein
was loaded onto a 12% SDS-polyacrylamide gel. Proteins
were transferred onto a PVDF membrane and subject to
western blot analysis using anti-phospho-ERK 1/2, and
phosphor-AKT antibody (1:1000, Cell Signaling Technology)
or anti-SPRY1 (1:1000, Abcam).
Bioinfomatic Anylysis of Human PTC Samples
The expression profiles of SPRY1,SPRY2,SPRY3, and SPRY4
in the TCGA data set of 572 PTC samples23 were analyzed
using UCSC cancer genomics browser (https://genome-
cancer.soe.ucsc.edu/proj/site/hgHeatmap/).
Statistical Analysis
Unpaired Studentst-test (two-tailed) was used. A P-value of
0.05 or less was considered as significant.
RESULTS
Without PTU treatment, only mild thyroid enlargement and
hyperplasia were observed in TPO-KRAS
G12D
mice (Figure 1b).
The thyroids were about two times larger as compared with
those from the TPO-KRAS
WT
mice. Thyroid cancer develop-
ment was not observed among 10 mice monitored for up to
14 months. With PTU treatment, significant thyroid enlarge-
ment with multinodular goiters and hyperplasia occurred in
both TPO-KRAS
G12D
and TPO-KRAS
WT
mice (Figure 2a).
Massive thyroid enlargement was found in TPO-KRAS
G12D
mice: thyroids of TPO-KRAS
G12D
mice were about six times
larger than TPO-KRAS
WT
mice (118.75 ±13.15 mg vs
21.11 ±6.33 mg, Po0.001, Figure 2b), indicating that TSH
can cooperate with KRAS
G12D
to promote thyroid growth and
hyperplasia. Interestingly, thyroid histology of TPO-
KRAS
G12D
mice showed nodular hyperplasia with well-
maintained follicular architecture whereas in TPO-KRAS
WT
mice such follicular architecture was replaced by papillary
hyperplasia (Figure 2c). However, PTC was not diagnosed due
to lack of characteristic nuclear features such as nuclear
grooves, intranuclear cytoplasmic inclusion, fine chromatin
texture, and nuclear envelope irregularity. As shown in
Figure 2d, serum TSH was significantly elevated in PTU-
treated mice as compared with the control (24 370.8 ±12 942.5
vs 426.4 ±9.65 pg/ml, Po0.01). Serum T4 was significantly
reduced in PTU-treated mice as compared with the control
(5.21 ±1.31 vs 22.68 ±6.23 ng/ml, Po0.05). Among 10 TPO-
KRAS
G12D
mice monitored for 14 months, two developed
follicular thyroid cancer (FTC, Figure 3a) and one with
pulmonary metastasis (Figure 3b). Since the cellular features
were indistinguishable between follicular hyperplasia and
cancer, the diagnosis of FTC was based on the invasion of
surrounding tissues such as blood vessel and neck muscle
(Figure 3a), and pulmonary metastasis (Figure 3b). Among 10
TPO-KRAS
WT
mice monitored for 14 months under PTU
treatment, no tumor development was observed.
The Sprouty (SPRY) family of proteins is involved in the
negative feedback regulation of growth factor-mediated
MAPK activation.24 To understand why TSH stimulation
caused only FTC, but not PTC, we investigated SPRY1
expression in thyroid tumors from TPO-KRAS
G12D
and TPO-
BRAF
V600E
mice. SPRY1 mRNA level was significantly higher
TSH mediated RAS transformation of thyroid follicular cells
M Zou et al
www.laboratoryinvestigation.org | Laboratory Investigation | Volume 95 November 2015 1271
in KRAS
G12D
-induced FTC (9.23 ±0.56) as compared with
KRAS
WT
control (1.03 ±0.15, Po0.01, Figure 4a). SPRY1
expression was also increased in KRAS
G12D
thyroid without
PTU treatment (2.1 ±0.2, Po0.01, Figure 4a), but the
expression level was much lower than under PTU treatment,
suggesting that the increased SPRY1 expression may be a
compensatory mechanism to counter TSH-mediated growth
stimulation. In contrast, SPRY1 mRNA level was significantly
lower in BRAF
V600E
-induced PTC (0.31 ±0.17) as compared
with BRAF
WT
control (1.00 ±0.12, Po0.05, Figure 4b). The
serum TSH was greater than 50 000 pg/ml (beyond the
detection limit of the kit) and T4 was 3.87±0.68 in TPO-
BRAF
V600E
mice (n=6) as compared with 445.2 ±72.21 in
TSH and 29.03 ±3.62 in T4 from TPO-BRAF
WT
mice (n=6),
consistent with previous report that TPO-BRAF
V600E
mice
have severe hypothyroidism.13 The increased SPRY1 expres-
sion in FTC was confirmed by immunohistochemistry
analysis. As shown in Figure 4c, increased SPRY1 and
p-AKT expression was observed in FTC (b and e) with
normal p-ERK expression (h). In PTC, both p-ERK and
p-AKT expression were increased (f and i). Although down-
regulation of SPRY1 expression in PTC was not clearly shown
by immunohistochemistry (Figure 4cC), it was confirmed by
western blot analysis, showing inverse correlation between
SPRY1 expression and p-ERK activation (Figure 4d).
To further investigate the role of SPRY1 in the regulation of
TSH-mediated signaling, we used BVE cell line (derived from
BRAF
V600E
-induced PTC tumor) as a model, for it has low
SPRY1 expression. The cell line was transfected with mouse
SPRY1 cDNA and treated with or without TSH. As shown in
Figure 5, SPRY1 expression reduced p-ERK expression, and
TSH stimulation only activated p-ATK when SPRY1
expression was increased. When SPRY1 expression was
reduced, TSH stimulation increased both p-ERK and p-ATK
expression (Figure 5). These data suggest that SPRY1 may act
as a molecular switch to control thyroid follicular cells to go
through either MAP kinase (RAF-MEK-ERK) or PI3 kinase
(PI3K/AKT) signaling pathway. When SPRY1 is upregulated
by KRAS
G12D
, MAPK pathway is inhibited and only PI3K/AKT
pathway is utilized, resulting in FTC. When SPRY1 is
downregulated by BRAF
V600E
, thyroid follicular cells may go
through both MAPK and PI3K/AKT pathways to initiate PTC.
Finally, we performed bioinformatic mining of the recent
TCGA data set of 572 human PTC samples to compare SPRY
gene (SPRY1,SPRY2,SPRY3, and SPRY4) expression between
classic PTC (CPTC) and follicular variant PTC (FVPTC).
FVPTC shares many molecular and pathological features with
FTC. RAS mutation is frequently detected in FVPTC as
compared with frequent BRAF mutation in CPTC. As shown
in Figure 6, the SPRY1 and SPRY4 expression is reduced in
normal thyroids. The expression of SPRY2 and SPRY3 tends
to increase in normal thyroids and is variable in both CPTC
and FVPTC. The SPRY1 expression is also variable in CPTC
and FVPTC. In CPTC, increased SPRY1 expression is present
in 36.6% (162/409) samples, decreased expression in 40.6%
(166/409) and unchanged in 19.8% (81/409) samples. In
FVPTC, 40.6% (43/106) samples have increased expression,
36.8% (39/106) have decreased expression, and 22.6%
(24/106) unchanged. The expression of SPRY4 is consistently
increased in FVPTC: 66.1% (72/109) have increased expres-
sion and 13.8% (15/109) have decreased expression. These
data suggest that the SPRY family of proteins is involved in
the regulation of thyroid tumorigenesis. The exact role of each
protein remains to be determined.
Figure 1 Activation of KRAS
G12D
in thyroid induces mild thyroid enlargement and hyperplasia. (a) Genotyping of TPO-Cre mediated recombination of
LSL-KRAS
G12D
(A) and LSL-BRAF
V600E
(B). The PCR product was run on a 1.5% agarose gel. The activation of KRAS
G12D
and BRAF
V600E
occurred only in the
thyroid as a result of Cre-mediated deletion of a floxed transcriptional stop sequence. (b) Mild thyroid enlargement and hyperplasia were observed in
TPO-KRAS
G12D
mice. Thyroids of a TPO-KRAS
WT
mouse (A) and a TPO-KRAS
G12D
mouse (B) at the age of 8 months were shown at the top panel. Histology
of thyroids from the same TPO-KRAS
WT
mice (C, × 20) and TPO-KRAS
G12D
mice (D, × 20) was shown at the bottom panel.
TSH mediated RAS transformation of thyroid follicular cells
M Zou et al
1272 Laboratory Investigation | Volume 95 November 2015 | www.laboratoryinvestigation.org
Figure 3 TSH cooperates with KRAS
G12D
to induce transformation of thyroid follicular cells into FTC. (a) Thyroid of a TPO-KRAS
G12D
mouse with FTC
following 14-month PTU treatment (A). FTC with neck muscle invasion as indicated by arrows (B, × 10) and blood vessel invasion (indicated by arrows,
C, × 20; D, × 40). (b) Pulmonary metastasis of FTC from the same mice. Multiple foci of metastasis can be seen from gross examination of the lung (A)
and microscopic examination (B, × 10; C, × 20, D, × 40).
Figure 2 TSH cooperates with KRAS
G12D
to induce massive thyroid hyperplasia. (a) Thyroids of a TPO-KRAS
wt
mouse (A) and a TPO-KRAS
G12D
mouse (B) at
16 months old were shown. The mice were treated with PTU in drinking water for 14 months. Goiters were present in both mice and significant thyroid
enlargement was observed in TPO-KRAS
G12D
mice. (b) Thyroid weight in PTU-treated mice. Thyroid was collected from 10 TPO-KRAS
G12D
and 10 TPO-KRAS
WT
mice treated with PTU for 14 months (at the end of experiment). Thyroid weight was measured and data were expressed as mean ± s.e.m., *Po0.001. (c)
Histology of thyroids from the same TPO-KRAS
WT
and TPO-KRAS
G12D
mice was shown. Follicular architecture was replaced by papillary structure in
hyperplastic goiters from a TPO-KRAS
WT
mouse (A, × 4; C, × 20) whereas in the goiter of a TPO-KRAS
G12D
mouse, the follicular architecture was intact (B, × 4,
D, × 20). (d) Thyroid hormone levels in PTU-treated mice. Serum was collected from six TPO-KRAS mice treated with PTU for 1 month and from six mice of
same age group without PTU treatment. The samples were assayed individually in triplicates. The data are expressed as mean ± s.em., *Po0.05.
TSH mediated RAS transformation of thyroid follicular cells
M Zou et al
www.laboratoryinvestigation.org | Laboratory Investigation | Volume 95 November 2015 1273
DISCUSSION
In the present study, we have demonstrated that KRAS
G12D
mutation alone is not sufficient to induce oncogenic
transformation of thyroid follicular cells. With long-term
TSH stimulation, KRAS
G12D
can promote thyroid growth and
hyperplasia, and eventually result in oncogenic transforma-
tion into follicular thyroid cancer. This demonstrates the
significant oncogenic role of TSH in thyroid tumorigenesis:
without TSH stimulation, thyroid cancer would not be able to
develop or progress even in the presence of KRAS
G12D
or
BRAF
V600E
.25
The animal model in the current study is very similar to
congenital hypothyroidism in humans, which is caused by
defects in thyroid hormone synthesis.26 High levels of TSH
are often present in patients with congenital hypothyroidism
due to poor management or patientsnon-compliance to
treatment. We and others have reported thyroid cancer
development as a result of long-term TSH stimulation.27,28
Although both PTC and FTC were reported in those cases,
BRAF mutation was found only in patients with PTC.29 RAS
mutations have been reported in both benign thyroid goiters
and cancers,30 indicating that additional factors are required
to initiate malignant transformation. The current study has
demonstrated that chronic TSH stimulation is one of the
factors required for RAS-mediated carcinogenesis.
TSH stimulates growth and differentiation of thyroid
follicular cells through its G protein-coupled receptor. TSH
can activate both cAMP-protein kinase A (PKA) pathway for
differentiation31 and RAS-PI3K pathway for proliferation.32,33
RAS allows growth promoting signals through both MAPK
(RAF-MEK-ERK) and PIK3CA/AKT pathways. Previous
studies have shown that cAMP can inhibit RAS-mediated
signals to MAPK by blocking RAF-1 activation via RAP1.34,35
Under these circumstances, PI3K signaling is the main
mediator of RAS effects. RAP1 is a member of the RAS
family of small G proteins that transmit signals from cAMP-
Figure 4 SPRY1 expression in thyroid cancer. SPRY1 mRNA from KRAS
G12D
-induced FTC (a) and BRAF
V600E
-induced PTC (b) was analyzed by qRT-PCR. RNA
from TPO-KRAS
WT
mouse thyroids and TPO-BRAF
WT
mouse thyroids was used as a control. Data were expressed as mean ± s.e.m. of three separate
experiments. (c) Immunohistochemistry staining of SPRY1, p-AKT, and p-ERK. Normal thyroid (A, D, and G, × 40), FTC from a TPO-KRAS
G12D
mouse
(B, E, and H, × 40), and PTC from a TPO-BRAF
V600E
mouse were stained with SPRY1 antibody (A, B, and C, 1:50 dilution), p-AKT antibody (D, E, and F, 1:50
dilution) and p-ERK antibody (G, H, and I, 1:50 dilution). Increased expression of SPRY1 and p-AKT was seen in FTC. p-ERK was not activated in FTC
whereas both p-AKT and p-ERK were activated in PTC. (d) Western blot analysis of SPRY1 expression in a PTC tumor. Decreased SPRY1 expression is
observed in PTC as compared with normal thyroid.
TSH mediated RAS transformation of thyroid follicular cells
M Zou et al
1274 Laboratory Investigation | Volume 95 November 2015 | www.laboratoryinvestigation.org
PKA pathway to MAPK pathway by interaction with BRAF.36
Without BRAF, GTP-loaded RAP1 blocks RAS activation of
RAF-1, thereby inhibiting MAPK signaling.37 In a mouse
model with constitutive RAP1 activation in thyroid, FTC was
not developed until given 12 months goitrogen treatment,
indicating that TSH signaling is required and is probably via
RAS-PI3K pathway for FTC initiation.38 In two different FTC
mouse models, TSH signaling is constantly activated with
simultaneous activation of either PI3K/AKT39,40 or phospho-
lipase C pathway.41 FTC was developed in all mice with PI3K/
AKT activation, suggesting that PI3K is an important
mediator of RAS effects in TSH-induced FTC. FTC can
develop rapidly without TSH signaling when both RAS and
PI3K/AKT pathways are constitutively activated.42 Although
MAPK is activated in these mice,42 PI3K signaling probably
predominates over MAPK, resulting in FTC phenotype. Since
serum TSH is very low in these mice, MAPK activation may
reflect the loss of cAMP-mediated inhibition of RAS signals to
MAPK.43
TSH signaling promotes the tumorigenesis of PTC,13
indicating a cross-talk between TSH-cAMP-PKA pathway
and MAPK pathway.37 As discussed above, RAP1 may be one
of the molecular switches to control TSH-cAMP signaling
through either RAS-PI3K to initiate FTC or RAS-MAPK to
induce PTC. In the current study, we have identified that
SPRY1 is another molecular switch to regulate RAS-MAPK
signaling. SPRY1 has been reported as a candidate tumor-
suppressor gene in medullary thyroid carcinoma44 and its
expression is decreased in human prostate cancer.45 We also
found reduced SPRY1 expression in human PTC with
BRAF
V600E
(data will be presented elsewhere). Concomitant
Figure 5 Regulation of TSH signaling by SPRY1. BVE cell line was
transfected with mouse SPRY1 cDNA and cultured in the presence or
absence of 10 mU/ml bovine TSH for 48 h. The expression of SPRY1,
phospho-ERK, and phospho-AKT was determined by western blot analysis.
Reduction of p-ERK is observed by increased SPRY1 expression, and TSH
stimulation only activates p-ATK in the presence of increased SPRY1
expression. TSH can increase both p-ERK and p-ATK expression when
SPRY1 expression is reduced.
Figure 6 The gene expression profile of SPRY family of proteins in human thyroid cancer. The TCGA thyroid cancer gene expression data set of 572
thyroid cancer samples was used to examine gene expression profile among normal thyroid, PTC, classic PTC (CPTC), tall cell PTC, and follicular variant
PTC (FVPTC). The gene expression profile is presented as a heatmap of red (upregulated), black (no change), and green (downregulated). The SPRY1 and
SPRY4 expression is downregulated in normal thyroids. The expression of SPRY4 is consistently increased in FVPTC.
TSH mediated RAS transformation of thyroid follicular cells
M Zou et al
www.laboratoryinvestigation.org | Laboratory Investigation | Volume 95 November 2015 1275
loss of SPRY1 and SPRY2 results in hyperactive MAPK
signaling and low-grade prostatic intraepithelial neoplasia.46
However, when SPRY1 and SPRY2 loss-of-function occurs in
the context of loss of one Pten allele, aberrant activation of
AKT and invasive neoplasia occur, suggesting that Sprouty
genes can negatively regulate the PI3K/AKT pathway as well.46
Indeed, PI3K/AKT activation has been reported in PTC
patients with BRAF
V600E
.47 Our data also demonstrate the
activation of both MAPK and PI3K/AKT pathways in
BRAF
V600E
-induced PTC when SPRY1 expression is reduced.
In KRAS
G12D
-induced lung cancer, SPRY2 is upregulated via
negative feedback loop to inhibit MAPK signaling.48 However,
we only found SPRY1 upregulation in KRAS
G12D
-induced FTC
and SPRY2 expression was not changed (data not shown). The
differential SPRY1 expression in TPO-KRAS
G12D
and TPO-
BRAF
V600E
mice indicates that SPRY1 can selectively regulate
TSH-mediated RAS signaling pathways in thyroid: favoring
MAPK pathway and PTC initiation when its expression is
downregulated and PI3K/AKT pathway and FTC initiation
when its expression is upregulated. It has been reported that
supraphysiologic expression of a mutant RAS can result in
PTC or mixed papillaryfollicular features in early transgenic
mice studies,8,10 which appears to be contradictory to our
hypothesis. However, overexpressed mutant RAS beyond
physiological level may escape SPRY1-mediated feedback
inhibition of MAPK signaling and induce PTC.
ACKNOWLEDGMENTS
The study is supported by KACST grants (11-BIO1434-20 and PL-10-0051). We
would like to thank Dr Mario Encinas for critical discussions.
DISCLOSURE/CONFLICT OF INTEREST
The authors declare no conflict of interest.
1. Davies L, Welch HG. Increasing incidence of thyroid cancer in the
United States, 1973-2002. JAMA 2006;295:21642167.
2. Hundahl SA, Fleming ID, Fremgen AM et al. A National Cancer Data
Base report on 53,856 cases of thyroid carcinoma treated in the U.S.,
1985-1995 [see commetns]. Cancer 1998;83:26382648.
3. Kimura ET, Nikiforova MN, Zhu Z et al. High prevalence of BRAF
mutations in thyroid cancer: genetic evidence for constitutive
activation of the RET/PTC-RAS-BRAF signaling pathway in papillary
thyroid carcinoma. Cancer Res 2003;63:14541457.
4. Cohen Y, Xing M, Mambo E et al. BRAF mutation in papillary thyroid
carcinoma. J Natl Cancer Inst 2003;95:625627.
5. Xing M. BRAF mutation in thyroid cancer. Endocr Relat Cancer 2005;12:
245262.
6. Vasko V, Ferrand M, Di Cristofaro J et al. Specific pattern of RAS
oncogene mutations in follicular thyroid tumors. J Clin Endocrinol
Metab 2003;88:27452752.
7. Fukahori M, Yoshida A, Hayashi H et al. The associations between RAS
mutations and clinical characteristics in follicular thyroid tumors: new
insights from a single center and a large patient cohort. Thyroid
2012;22:683689.
8. Rochefort P, Caillou B, Michiels FM et al. Thyroid pathologies in
transgenic mice expressing a human activated Ras gene driven by a
thyroglobulin promoter. Oncogene 1996;12:111118.
9. Santelli G, de Franciscis V, Portella G et al. Production of transgenic
mice expressing the Ki-ras oncogene under the control of a
thyroglobulin promoter. Cancer Res 1993;53:55235527.
10. Vitagliano D, Portella G, Troncone G et al. Thyroid targeting of the
N-ras(Gln61Lys) oncogene in transgenic mice results in follicular
tumors that progress to poorly differentiated carcinomas. Oncogene
2006;25:54675474.
11. Francis-Lang H, Zannini M, De Felice M et al. Multiple mechanisms
of interference between transformation and differentiation in
thyroid cells. Mol Cell Biol 1992;12:57935800.
12. De Vita G, Bauer L, da Costa VM et al. Dose-dependent inhibition of
thyroid differentiation by RAS oncogenes. Mol Endocrinol 2005;19:
7689.
13. Franco AT, Malaguarnera R, Refetoff S et al. Thyrotrophin receptor
signaling dependence of Braf-induced thyroid tumor initiation in mice.
Proc Natl Acad Sci USA 2011;108:16151620.
14. Charles RP, Iezza G, Amendola E et al. Mutationally activated BRAF
(V600E) elicits papillary thyroid cancer in the adult mouse. Cancer Res
2011;71:38633871.
15. Kim HK, Yoon JH, Kim SJ et al. Higher TSH level is a risk factor
for differentiated thyroid cancer. Clin Endocrinol (Oxf) 2013;78:
472477.
16. Boelaert K. The association between serum TSH concentration and
thyroid cancer. Endocr Relat Cancer 2009;16:10651072.
17. Jackson EL, Willis N, Mercer K et al. Analysis of lung tumor initiation
and progression using conditional expression of oncogenic K-ras.
Genes Dev 2001;15:32433248.
18. Kusakabe T, Kawaguchi A, Kawaguchi R et al. Thyrocyte-specific
expression of Cre recombinase in transgenic mice. Genesis 2004;39:
212216.
19. Mercer K, Giblett S, Green S et al. Expression of endogenous oncogenic
V600EB-raf induces proliferation and developmental defects in mice
and transformation of primary fibroblasts. Cancer Res 2005;65:
1149311500.
20. Zou M, Baitei EY, Alzahrani AS et al. Oncogenic activation of MAP
kinase by BRAF pseudogene in thyroid tumors. Neoplasia 2009;11:
5765.
21. Zou M, Famulski KS, Parhar RS et al. Microarray analysis of metastasis-
associated gene expression profiling in a murine model of thyroid
carcinoma pulmonary metastasis: identification of S100A4 (Mts1) gene
overexpression as a poor prognostic marker for thyroid carcinoma.
J Clin Endocrinol Metab 2004;89:61466154.
22. Zou M, Al-Baradie RS, Al-Hindi H et al. S100A4 (Mts1) gene
overexpression is associated with invasion and metastasis of papillary
thyroid carcinoma. Br J Cancer 2005;93:12771284.
23. Cancer Genome Atlas Research Network. Integrated genomic
characterization of papillary thyroid carcinoma. Cell 2014;159:
676690.
24. Hanafusa H, Torii S, Yasunaga T et al. Sprouty1 and Sprouty2 provide a
control mechanism for the Ras/MAPK signalling pathway. Nat Cell Biol
2002;4:850858.
25. Shimamura M, Nakahara M, Orim F et al. Postnatal expression of
BRAFV600E does not induce thyroid cancer in mouse models of
thyroid papillary carcinoma. Endocrinology 2013;154:44234430.
26. Kopp P. Perspective: genetic defects in the etiology of congenital
hypothyroidism. Endocrinology 2002;143:20192024.
27. Alzahrani AS, Baitei EY, Zou M et al. Clinical case seminar: metastatic
follicular thyroid carcinoma arising from congenital goiter as a result of
a novel splice donor site mutation in the thyroglobulin gene. J Clin
Endocrinol Metab 2006;91:740746.
28. Raef H, Al-Rijjal R, Al-Shehri S et al. Biallelic p.R2223H mutation in the
thyroglobulin gene causes thyroglobulin retention and severe
hypothyroidism with subsequent development of thyroid carcinoma.
J Clin Endocrinol Metab 2010;95:10001006.
29. Hishinuma A, Fukata S, Kakudo K et al. High incidence of thyroid
cancer in long-standing goiters with thyroglobulin mutations. Thyroid
2005;15:10791084.
30. Shi YF, Zou MJ, Schmidt H et al. High rates of ras codon 61 mutation in
thyroid tumors in an iodide-deficient area. Cancer Res 1991;51:
26902693.
31. Laugwitz KL, Allgeier A, Offermanns S et al. The human thyrotropin
receptor: a heptahelical receptor capable of stimulating members of all
four G protein families. Proc Natl Acad Sci USA 1996;93:116120.
32. Ciullo I, Diez-Roux G, Di Domenico M et al. cAMP signaling selectively
influences Ras effectors pathways. Oncogene 2001;20:11861192.
33. Suh JM, Song JH, Kim DW et al. Regulation of the phosphatidylinositol
3-kinase, Akt/protein kinase B, FRAP/mammalian target of rapamycin,
and ribosomal S6 kinase 1 signaling pathways by thyroid-stimulating
TSH mediated RAS transformation of thyroid follicular cells
M Zou et al
1276 Laboratory Investigation | Volume 95 November 2015 | www.laboratoryinvestigation.org
hormone (TSH) and stimulating type TSH receptor antibodies in the
thyroid gland. J Biol Chem 2003;278:2196021971.
34. Cook SJ, McCormick F. Inhibition by cAMP of Ras-dependent
activation of Raf. Science 1993;262:10691072.
35. Schmitt JM, Stork PJ. Cyclic AMP-mediated inhibition of cell growth
requires the small G protein Rap1. Mol Cell Biol 2001;21:36713683.
36. Wang Z, Dillon TJ, Pokala V et al. Rap1-mediated activation of
extracellular signal-regulated kinases by cyclic AMP is dependent on
the mode of Rap1 activation. Mol Cell Biol 2006;26:21302145.
37. Stork PJ, Schmitt JM. Crosstalk between cAMP and MAP kinase
signaling in the regulation of cell proliferation. Trends Cell Biol
2002;12:258266.
38. Ribeiro-Neto F, Leon A, Urbani-Brocard J et al. cAMP-dependent
oncogenic action of Rap1b in the thyroid gland. J Biol Chem 2004;279:
4686846875.
39. Suzuki H, Willingham MC, Cheng SY. Mice with a mutation in the
thyroid hormone receptor beta gene spontaneously develop thyroid
carcinoma: a mouse model of thyroid carcinogenesis. Thyroid 2002;12:
963969.
40. Furuya F, Lu C, Willingham MC et al. Inhibition of phosphatidylinositol
3-kinase delays tumor progression and blocks metastatic spread in a
mouse model of thyroid cancer. Carcinogenesis 2007;28:24512458.
41. Ledent C, Denef JF, Cottecchia S et al. Costimulation of adenylyl
cyclase and phospholipase C by a mutant alpha 1B-adrenergic
receptor transgene promotes malignant transformation of thyroid
follicular cells. Endocrinology 1997;138:369378.
42. Miller KA, Yeager N, Baker K et al. Oncogenic Kras requires
simultaneous PI3K signaling to induce ERK activation and transform
thyroid epithelial cells in vivo. Cancer Res 2009;69:36893694.
43. Li Y, Takahashi M, Stork PJ. Ras-mutant cancer cells display B-Raf
binding to Ras that activates extracellular signal-regulated kinase and
is inhibited by protein kinase A phosphorylation. J Biol Chem 2013;288:
2764627657.
44. Macia A, Gallel P, Vaquero M et al. Sprouty1 is a candidate tumor-
suppressor gene in medullary thyroid carcinoma. Oncogene 2012;31:
39613972.
45. Kwabi-Addo B, Wang J, Erdem H et al. The expression of Sprouty1, an
inhibitor of fibroblast growth factor signal transduction, is decreased
in human prostate cancer. Cancer Res 2004;64:47284735.
46. Schutzman JL, Martin GR. Sprouty genes function in suppression
of prostate tumorigenesis. Proc Natl Acad Sci USA 2012;109:
2002320028.
47. Faustino A, Couto JP, Populo H et al. mTOR pathway overactivation in
BRAF mutated papillary thyroid carcinoma. J Clin Endocrinol Metab
2012;97:E1139E1149.
48. Shaw AT, Meissner A, Dowdle JA et al. Sprouty-2 regulates oncogenic
K-ras in lung development and tumorigenesis. Genes Dev 2007;21:
694707.
TSH mediated RAS transformation of thyroid follicular cells
M Zou et al
www.laboratoryinvestigation.org | Laboratory Investigation | Volume 95 November 2015 1277
... However, tumor cells exhibit resistance to anoikis under pathological conditions. In the process of tumor cells detaching from the primary site, passing through the lymphatic and circulatory system, invading and implanting into the secondary site for proliferation and growth, the tolerance to anoikis becomes a prerequisite for tumor metastasis and drug resistance [36]. The resistance of cancer cells to anoikis contributes to the invasion, migration and development of drug resistance in tumors [37]. ...
Article
Full-text available
As a type of programmed cell death, anoikis resistance plays an essential role in tumor metastasis, allowing cancer cells to survive in the systemic circulation and as a key pathway for regulating critical biological processes. We conducted an exploratory analysis to improve risk stratification and optimize adjuvant treatment choices for patients with breast cancer, and identify multigene features in mRNA and lncRNA transcriptome profiles associated with anoikis. First, the variance selection method filters low information content genes in RNA sequence and then extracts the mRNA and lncRNA expression data base on annotation files. Then, the top ten key mRNAs are screened out through the PPI network. Pearson analysis has been employed to identify lncRNAs related to anoikis, and the prognosis-related lncRNAs are selected using Univariate Cox regression and machine learning. Finally, we identified a group of RNAs (including ten mRNAs and six lncRNAs) and integrated the expression data of 16 genes to construct a risk-scoring system for BRCA prognosis and drug sensitivity analysis. The risk score's validity has been evaluated with the ROC curve, Kaplan-Meier survival curve analysis and decision curve analysis (DCA). For the methylation data, we have obtained 169 anoikis-related prognostic methylation sites, integrated these sites with 16 RNA features and further used the deep learning model to evaluate and predict the survival risk of patients. The developed anoikis feature is demonstrated a consistency index (C-index) of 0.778, indicating its potential to predict the survival probability of breast cancer patients using deep learning methods.
... At present, it is unclear whether long-term TSH stimulation can promote KRAS G12D -mediated transformation of thyroid follicular cells. Therefore, Zou et al. studied the effect of long-term TSH stimulation on KRAS G12D knock-in mice and the role of Sprouty1 (SPRY1) in KRAS G12D -mediated signal transduction and found that chronic TSH stimulation can enhance KRAS G1D -mediated tumorigenesis, leading to FTC [90]. ...
Article
Full-text available
Simple Summary Thyroid-stimulating hormone (TSH) plays a role in the regulation of thyroid hormones and is an important indicator for assessing thyroid function. Thyroid cancer (TC) is a common tumor of endocrine system, and its incidence rate is increasing year by year. The role of TSH in the thyroid gland suggests a potential influence on the occurrence and progression of TC, which has attracted the attention of the scientific community. Based on the controversial fact that TSH may affect TC, this article reviews the epidemiological evidence and potential mechanisms of the relationship between TSH and TC, in order to provide valuable references for future research work. Abstract Thyroid Stimulating Hormone (TSH) is a hormone secreted by the pituitary gland and plays a role in regulating the production and secretion of thyroid hormones by the thyroid gland. This precise feedback loop is essential for maintaining a harmonious balance of thyroid hormones in the body, which are vital for numerous physiological processes. Consequently, TSH serves as a significant marker in assessing thyroid function, and deviations from normal TSH levels may indicate the presence of a thyroid disorder. Thyroid cancer (TC) is the malignant tumor within the endocrine system. In recent years, numerous experts have dedicated their efforts to discovering efficacious biomarkers for TC. These biomarkers aim to improve the accurate identification of tumors with a poor prognosis, as well as facilitate active monitoring of tumors with a more favorable prognosis. The role of TSH in the thyroid gland underscores its potential influence on the occurrence and progression of TC, which has garnered attention in the scientific community. However, due to the limited scope of clinical research and the dearth of high-quality foundational studies, the precise impact of TSH on TC remains unclear. Consequently, we present a comprehensive review of this subject, aiming to offer a valuable reference for future research endeavors.
... In contrast, when its expression is downregulated, it promotes PI3K/AKT pathway activation and FTC initiation. The abnormal expression of RAS can escape SPRY1-mediated feedback inhibition of the MAPK signal and induce the occurrence of DTC [92]. ...
Article
Full-text available
Thyroid cancer remains the most prevailing endocrine malignancy, and a progressively increasing incidence rate has been observed in recent years, with 95% of thyroid cancer represented by differentiated thyroid carcinomas. The genetics and epigenetics of thyroid cancer are gradually increasing, and gene mutations and methylation changes play an important roles in its occurrence and development. Although the role of RAS and BRAF mutations in thyroid cancer have been partially clarified,but the pathogenesis and molecular mechanisms of thyroid cancer remain to be elucidated. Epigenetic modification refer to genetic modification that does not change the DNA sequence of a gene but causes heritable phenotypic changes in its expression. Epigenetic modification mainly includes four aspects: DNA methylation, chromatin remodelling, noncoding RNA regulation, and histone modification. This article reviews the importance of thyroid cancer epigenetic modification and BRAF gene mutation in the treatment of thyroid cancer.
... TaqMan assays were performed to examine the expression of AhR exon 2, and Cyp1a1 induction, a downstream biomarker of activated AhR. Recombination of Apc S580 and Kras LSL G12D was determined genetically by modifying a previously described multiplex PCR assay (26)(27)(28). PCR was performed using Hot-Start-Taq Blue Mastermix (Denville Scientific, CB4040-8) according to the manufacturer's instructions for 35 cycles using the Apc and Kras primer sets: Apc (5 0 -GTTCTGTATCATGGAAAGATAGGTGGTC-3 0 , 5 0 -CACTCAAAACGCTTTTGAGGGTTGATTC-3 0 , and 5 0 -GAG-TACGGGGTCTCTGTCTCAGTGAA-3 0 ), and Kras (5 0 -GTCTTTCC-CCAGCACAGTGC-3 0 , 5 0 -GCAGCGTTACCTCTATCGTA-3 0 , and 5 0 -AGCTAGCCACCATGGCTTGAGTAAGTCTGCA-3 0 ). ...
Article
The mutational genetic landscape of colorectal cancer has been extensively characterized; however, the ability of “cooperation response genes” to modulate the function of cancer “driver” genes remains largely unknown. In this study, we investigate the role of aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, in modulating oncogenic cues in the colon. We show that intestinal epithelial cell–targeted AhR knockout (KO) promotes the expansion and clonogenic capacity of colonic stem/progenitor cells harboring ApcS580/+; KrasG12D/+ mutations by upregulating Wnt signaling. The loss of AhR in the gut epithelium increased cell proliferation, reduced mouse survival rate, and promoted cecum and colon tumorigenesis in mice. Mechanistically, the antagonism of Wnt signaling induced by Lgr5 haploinsufficiency attenuated the effects of AhR KO on cecum and colon tumorigenesis. Implications: Our findings reveal that AhR signaling plays a protective role in genetically induced colon tumorigenesis at least by suppressing Wnt signaling and provides rationale for the AhR as a therapeutic target for cancer prevention and treatment.
... Total RNA from peripheral blood leukocytes was extracted as described previously [35]. Two μg of total RNA were reverse-transcribed into cDNA using Promega reverse transcription system (Promega, Madison, WI). ...
Article
Full-text available
Background Hereditary hypophosphatemia is a group of rare renal phosphate wasting disorders. The diagnosis is based on clinical, radiological, and biochemical features, and may require genetic testing to be confirmed. Methodology Clinical features and mutation spectrum were investigated in patients with hereditary hypophosphatemia. Genomic DNA of 23 patients from 15 unrelated families were screened sequentially by PCR-sequencing analysis for mutations in the following genes: PHEX, FGF23, DMP1, ENPP1, CLCN5, SLC34A3 and SLC34A1. CytoScan HD Array was used to identify large deletions. Results Genetic evaluation resulted in the identification of an additional asymptomatic but intermittent hypophosphatemic subject. Mutations were detected in 21 patients and an asymptomatic sibling from 13 families (86.6%, 13/15). PHEX mutations were identified in 20 patients from 12 families. Six of them were novel mutations present in 9 patients: c.983_987dupCTACC, c.1586+2T>G, c.1206delA, c.436+1G>T, c.1217G>T, and g.22,215,887–22,395,767del (179880 bp deletion including exon 16–22 and ZNF645). Six previously reported mutations were found in 11 patients. Among 12 different PHEX mutations, 6 were de novo mutations. Patients with de novo PHEX mutations often had delayed diagnosis and significantly shorter in height than those who had inherited PHEX mutations. Novel compound heterozygous mutations in SLC34A3 were found in one patient and his asymptomatic sister: c.1335+2T>A and c.1639_1652del14. No mutation was detected in two families. Conclusions This is the largest familial study on Turkish patients with hereditary hypophosphatemia. PHEX mutations, including various novel and de novo variants, are the most common genetic defect. More attention should be paid to hypophosphatemia by clinicians since some cases remain undiagnosed both during childhood and adulthood.
... In the same study, expression analysis based on real-time PCR showed that the SPRY1 expression was significantly higher in mutant compared to that of wild-type mice for KRAS in contrast to that of the mutant and wild-type mice for BRAF that showed significantly lower level of SPRY1 expression for the former compared to the latter. Using the outcomes of this study, it was suggested that (i) the initiation of FTC is associated with activation of the PI3K/AKT pathway through the activation of p-AKT and reduced expression of p-ERK by the increased expression of SPRY1 in KRASG12D cells when treated with TSH, (ii) the initiation of PTC (instead of FTC) is associated with the activation of both MAPK/ERK and PI3K/AKT pathways through the increased expression of both p-ERK and p-AKT by the reduced expression of SPRY1 in BRAFV600E cells, and (iii) the SPRY1 acts as a molecular switch to control and regulate the transformation of thyroid follicular cells into malignant state (FTC or PTC) [35]. ...
Article
Full-text available
... In the same study, expression analysis based on real-time PCR showed that the SPRY1 expression was significantly higher in mutant compared to that of wild-type mice for KRAS in contrast to that of the mutant and wild-type mice for BRAF that showed significantly lower level of SPRY1 expression for the former compared to the latter. Using the outcomes of this study, it was suggested that (i) the initiation of FTC is associated with activation of the PI3K/AKT pathway through the activation of p-AKT and reduced expression of p-ERK by the increased expression of SPRY1 in KRASG12D cells when treated with TSH, (ii) the initiation of PTC (instead of FTC) is associated with the activation of both MAPK/ERK and PI3K/AKT pathways through the increased expression of both p-ERK and p-AKT by the reduced expression of SPRY1 in BRAFV600E cells, and (iii) the SPRY1 acts as a molecular switch to control and regulate the transformation of thyroid follicular cells into malignant state (FTC or PTC) [35]. ...
Article
Oncogene-induced senescence (OIS) explains the phenomenon of cellular senescence triggered by the action of oncogenes. It is a mechanism adopted by a cell to inhibit progression of benign tumors into malignancy, occurs in premalignant lesions, and is almost never present in malignant lesions. BRAF mutations occur in about 40–45% of all papillary thyroid carcinomas (PTCs) and of which 99.7% is the BRAFV600E mutation. A unique phenotype of the BRAFV600E mutation is the upregulation of the thyroid-stimulating hormone receptor (TSHR) on thyrocyte membranes. Despite the overexpression of the receptor, BRAFV600E cells undergo cell cycle arrest leading to OIS via a negative feedback signaling mechanism. A simultaneous increase in serum thyroid-stimulating hormone (TSH) in response to hypothyroidism (common in autoimmune diseases such as Hashimoto’s thyroiditis) would cause senescent tumor cells to overcome OIS and proceed towards malignancy, hence showing the importance of TSH/TSHR signaling in the development of PTCs. Increase in TSH/TSHR signaling triggers an increase in levels of downstream enzymes such as manganese superoxide dismutase (MnSOD) and dual-specific phosphatase 6 (DUSP6) which eventually results in the production of oncogenic proteins such as c-Myc. Therefore, the detection of these genetic alterations as effective biomarkers for premalignant lesions of PTC is important in clinical settings and techniques such as polymerase chain reaction-mediated restriction fragment length polymorphism (PCR-RFLP) and real-time PCR can be used to detect the BRAFV600E point mutation and overexpression of TSHR, MnSOD, and DUSP6, respectively.
... Total RNA from peripheral blood leucocytes was extracted as described previously. 24 Two micrograms of total RNA was reverse-transcribed into cDNA using Promega reverse transcription system (Promega, Madison, WI, USA). To determine the effect of a splice site mutation at c.1768+1G>A, RT-PCR was used to amplify PHEX transcripts using the following two primers: 5′-TTGGGGAACAGAATATCCTC-3′ (forward primer located in exon 16) and 5′-CCGCAGGCCTCCATTATCA -3′ (reverse primer located in exon 19). ...
Article
Full-text available
Context: Hypophosphataemic rickets (HR) is a group of rare hereditary renal phosphate wasting disorders caused by mutations in PHEX, FGF23, DMP1, ENPP1, CLCN5, or SLC34A3. Objective: To investigate underlying genetic defects in patients with hypophosphataemic rickets. Methods: We analyzed genomic DNA from 9 unrelated families for mutations in the entire coding region of PHEX, FGF23, DMP1, ENPP1, CLCN5, or SLC34A3 by PCR-sequencing and copy number analysis. Results: A total of 14 patients were studied. PHEX mutations were identified in 12 patients from 7 families. Five of them were novel mutations present in 8 patients: c.154G>T (p.E52*), c.401_402insGCCAAA (p.Q134_K135insPK), c.1600C>T (p.P534S), g.22016715_22056805del (40kb deletion including promoter and exons 1-3) and c.2242_2243delCT (p.L748fs*48). Four patients had previously reported mutations: c.1768+1G>A, and c.1807G>A (p.W602*). Novel CLCN5 (c.1205G>A, p.W402*) and FGF23 (c.526C>G, p.R176G) mutations were found in two patients from the remaining 2 families. Many of the mutations were de novo: c.154G>T and c.2242_2243delCT in PHEX, and c.526C>G in FGF23. Furthermore, we characterized the breakpoint of the novel PHEX g.22016715_22056805del and the c.2242_2243delCT, which is 6 bp from the stop codon, resulting in a frameshift and extension of the reading frame by 42 amino acids. Conclusions: Novel and de novo mutations are frequent and PHEX mutations are still the most common genetic defects in the Turkish population. Gene copy number analysis should be considered in patients with negative results by conventional PCR-based sequencing analysis. The current study further expands the mutation spectrum underlying HR. This article is protected by copyright. All rights reserved.
Chapter
Cancer Genes is a comprehensive list of the most critical genes known to contribute to cancer imitation and progression. The book delves into their location on each chromosome, providing valuable insights into the mechanisms of cancer gene dysregulation and genetic mutations which provide cancer cells with an advantage during each stage of tumorigenesis. The reference will familiarize readers with the location of cancer genes and equip them with the necessary information to identify relevant gene expression targets for research aimed at preventing the disease. The book is divided into two volumes focusing on cancer-causing genes found in chromosome pairs 1-12 (volume 1), and chromosomes 13-23 (volume 2). A key features of the book is a detailed reference list for advanced readers. The compilation is therefore a quick and handy reference on cancer causing genes for researchers, medical professionals, and anyone interested in understanding the genetic basis of cancer.
Article
The genomic simplicity of differentiated cancers derived from thyroid follicular cells offers unique insights into how oncogenic drivers impact tumour phenotype. Essentially, the main oncoproteins in thyroid cancer activate nodes in the receptor tyrosine kinase-RAS-BRAF pathway, which constitutively induces MAPK signalling to varying degrees consistent with their specific biochemical mechanisms of action. The magnitude of the flux through the MAPK signalling pathway determines key elements of thyroid cancer biology, including differentiation state, invasive properties and the cellular composition of the tumour microenvironment. Progression of disease results from genomic lesions that drive immortalization, disrupt chromatin accessibility and cause cell cycle checkpoint dysfunction, in conjunction with a tumour microenvironment characterized by progressive immunosuppression. This Review charts the genomic trajectories of these common endocrine tumours, while connecting them to the biological states that they confer.
Article
Full-text available
Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer. Here, we describe the genomic landscape of 496 PTCs. We observed a low frequency of somatic alterations (relative to other carcinomas) and extended the set of known PTC driver alterations to include EIF1AX, PPM1D, and CHEK2 and diverse gene fusions. These discoveries reduced the fraction of PTC cases with unknown oncogenic driver from 25% to 3.5%. Combined analyses of genomic variants, gene expression, and methylation demonstrated that different driver groups lead to different pathologies with distinct signaling and differentiation characteristics. Similarly, we identified distinct molecular subgroups of BRAF-mutant tumors, and multidimensional analyses highlighted a potential involvement of oncomiRs in less-differentiated subgroups. Our results propose a reclassification of thyroid cancers into molecular subtypes that better reflect their underlying signaling and differentiation properties, which has the potential to improve their pathological classification and better inform the management of the disease.
Article
Full-text available
The small G protein Ras regulates proliferation through activation of the mitogen-activated protein (MAP) kinase (ERK) cascade. The first step of Ras-dependent activation of ERK signaling is Ras binding to members of the Raf family of MAP kinase kinase kinases, C-Raf and B-Raf. Recently, it has been reported that in melanoma cells harboring oncogenic Ras mutations, B-Raf does not bind to Ras and does not contribute to basal ERK activation. For other types of Ras-mutant tumors, the relative contributions of C-Raf and B-Raf are not known. We examined non-melanoma cancer cell lines containing oncogenic Ras mutations and express both C-Raf and B-Raf isoforms, including the lung cancer cell line H1299 cells. Both B-Raf and C-Raf were constitutively bound to oncogenic Ras and contributed to Ras-dependent ERK activation. Ras binding to B-Raf and C-Raf were both subject to inhibition by the cAMP-dependent protein kinase PKA. cAMP inhibited the growth of H1299 cells and Ras-dependent ERK activation via PKA. PKA inhibited the binding of Ras to both C-Raf and B-Raf through phosphorylations of C-Raf at Ser-259 and B-Raf at Ser-365, respectively. These studies demonstrate that in non-melanocytic Ras-mutant cancer cells, Ras signaling to B-Raf is a significant contributor to ERK activation and that the B-Raf pathway, like that of C-Raf, is a target for inhibition by PKA. We suggest that cAMP and hormones coupled to cAMP may prove useful in dampening the effects of oncogenic Ras in non-melanocytic cancer cells through PKA-dependent actions on B-Raf as well as C-Raf. Background: How the cAMP-dependent protein kinase PKA regulates B-Raf binding to Ras is not known. Results: PKA inhibits the binding of B-Raf to active Ras via phosphorylation of serine 365 in B-Raf. Conclusion: B-Raf can participate in the Ras-dependent activation of ERK in Ras-mutant cancers, and this is inhibited by PKA. Significance: PKA can block Ras binding to both Raf isoforms C-Raf and B-Raf.
Article
Full-text available
TVansgenic mice have been generated bearing three fusion genes con sisting of: (a) a 900-base pair rat thyroglobulin promoter followed by a gene coding for a chloroamphenicol acetyl transferase activity; (In the same promoter followed by the complementary DNA of the human acti vated Ki-ras oncogene; (ci a 2000-base pair rat thyroglobulin promoter followed by the complementary DNA of the human activated Ki-ra\. We have shown that the 900-base pair rat thyroglobulin promoter is able to direct the expression of the reporter gene specifically in the thyroid gland of transgenic mice. The mice bearing the two Ki-/-«.\ constructs, which express the transgene in thyroid glands, show thyroid abnormalities, al though at very low incidence. These lesions appear after a long latency and with a benign aspect, thus suggesting that, in agreement with literature data on naturally occurring human thyroid tumors, the action of an activated rax gene is not sufficient to attain a complete malignant conver sion of thyroid glands in vivo. However, ras expression in thyroid follicular cells represents a favorable ground for tumor development, as shown by the fact that goitrogen stimulation experiments increase the occurrence of tumors.
Article
Transformation of the thyroid cell line FRTL-5 results in loss or reduction of differentiation as measured by the expression of thyroglobulin and thyroperoxidase, two proteins whose genes are exclusively expressed in thyroid follicular cells. The biochemical mechanisms leading to this phenomenon were investigated in three cell lines obtained by transformation of FRTL-5 cells with Ki-ras, Ha-ras, and polyomavirus middle-T oncogenes. With the ras oncogenes, transformation leads to undetectable expression of the thyroglobulin and thyroperoxidase genes. However, the mechanisms responsible for the extinction of the differentiated phenotype seem to be different for the two ras oncogenes. In Ki-ras-transformed cells, the mRNA encoding TTF-1, a transcription factor controlling thyroglobulin and thyroperoxidase gene expression, is severely reduced. On the contrary, nearly wild-type levels of TTF-1 mRNA are detected in Ha-ras-transformed cells. Furthermore, overexpression of TTF-1 can activate transcription of the thyroglobulin promoter in Ki-ras-transformed cells, whereas it has no effect on thyroglobulin transcription in the Ha-ras-transformed line. Expression of polyoma middle-T antigen in thyroid cells leads to only a reduction of differentiation and does not severely affect either the activity or the amount of TTF-1. Another thyroid cell-specific transcription factor, TTF-2, is more sensitive to transformation, since it disappears in all three transformed lines, and probably contributes to the reduced expression of the differentiated phenotype.
Article
Proliferation of thyroid follicular cells is controlled by three intracellular cascades [cAMP, inositol 1,4,5-triphosphate (IP3)/Ca2+/diacylglycerol (DAG), and tyrosine kinases] that are activated by distinct extracellular signals and receptors. We had previously generated a transgenic mouse model in which the cAMP cascade was permanently stimulated in thyroid cells by an adenosine A2a receptor (Tg-A2aR model). In the present work, we have generated a transgenic model characterized by the chronic stimulation of both adenylyl cyclase and phospholipase C in thyroid follicular cells. The bovine thyroglobulin gene promoter was used to direct the expression of a constitutively active mutant of theα 1B adrenergic receptor, which is known to couple to both cascades in transfected cell lines. The expression of the transgene resulted, as expected, in the activation of phospholipase C and adenylyl cyclase, as demonstrated by the direct measurement of IP3 and cAMP in thyroid tissue. The phenotype resulting from this...
Article
The mutant BRAF (BRAF(V600E)) is the most common genetic alteration in papillary thyroid carcinomas (PTC). The oncogenicity of this mutation has been shown by some genetically engineered mouse models. However, in these mice, BRAF(V600E) is expressed in all the thyroid cells from the fetal periods, and suppresses thyroid function, thereby leading to TSH elavation, which by itself promotes thyroid tumorigenesis. To overcome these problems, we exploited two different approaches both of which allowed temporally and spatially restricted expression of BRAF(V600E) in the thyroid glands. Firstly, we generated conditional transgenic mice harboring the loxP-neo(R)-loxP-BRAF(V600E)-IRES-GFP sequence [Tg(LNL-BRAF(V600E))]. The double transgenic mice (LNL-BRAF(V600E);TPO-Cre) derived from a high expressor line of Tg(LNL-BRAF(V600E)) mice and TPO-Cre mice, the latter expresses Cre DNA recombinase under the control of thyroid-specific thyroid peroxidase (TPO) promoter, developed PTC-like lesions in early life under normal serum TSH levels due to mosaic recombination. In contrast, injection of adenovirus expressing Cre under the control of another thyroid-specific thyroglobulin (Tg) promoter (Ad-TgP-Cre) into the thyroids of LNL-BRAF(V600E) mice did not induce tumor formation despite detection of BRAF(V600E) and pERK in a small fraction of thyroid cells. Secondly, postnatal expression of BRAF(V600E) in a small number of thyroid cells was also achieved by injecting the lentivirus expressing loxP-GFP-loxP-BRAF(V600E) into the thyroids of TPO-Cre mice; however no tumor development was again observed. These results suggest that BRAF(V600E) does not appear to induce PTC-like lesions when expressed in a fraction of thyroid cells postnatally under normal TSH concentrations.
Article
Expression of Sprouty genes is frequently decreased or absent in human prostate cancer, implicating them as suppressors of tumorigenesis. Here we show they function in prostate tumor suppression in the mouse. Concomitant inactivation of Spry1 and Spry2 in prostate epithelium causes ductal hyperplasia and low-grade prostatic intraepithelial neoplasia (PIN). However, when Spry1 and Spry2 loss-of-function occurs in the context of heterozygosity for a null allele of the tumor suppressor gene Pten, there is a striking increase in PIN and evidence of neoplastic invasion. Conversely, expression of a Spry2 gain-of-function transgene in Pten null prostatic epithelium suppresses the tumorigenic effects of loss of Pten function. We show that Sprouty gene loss-of-function results in hyperactive RAS/ERK1/2 signaling throughout the prostate epithelium and cooperates with heterozygosity for a Pten null allele to promote hyperactive PI3K/AKT signaling. Furthermore, Spry2 gain-of-function can suppress hyperactivation of AKT caused by the absence of PTEN. Together, these results point to a key genetic interaction between Sprouty genes and Pten in prostate tumorigenesis and provide strong evidence that Sprouty genes can function to modulate signaling via the RAS/ERK1/2 and PI3K/AKT pathways. The finding that Sprouty genes suppress tumorigenesis caused by Pten loss-of-function suggests that therapeutic approaches aimed at restoring normal feedback mechanisms triggered by receptor tyrosine kinase signaling, including Sprouty gene expression, may provide an effective strategy to delay or prevent high-grade PIN and invasive prostate cancer.
Article
Objectives: Higher thyroid-stimulating hormone (TSH) levels are associated with differentiated thyroid cancers (DTC). To validate this association, we compared TSH levels obtained from euthyroid patients with DTC with TSH levels from controls in the general population. Design and patients: The case group included 1759 patients with DTC, who underwent thyroid surgery at Chonnam National University Hwasun Hospital. The control group (n = 1548), who had participated in the Thyroid Disease Prevalence Study were used as a healthy control group. The subjects were divided into four groups of similar size according to their TSH levels, with the first quartile used as a reference group. Results: The mean TSH level of the case group was significantly higher than the mean TSH level of the control group (1.95 ± 0.9 mIU/l vs 1.62 ± 0.8 mIU/l, P < 0.001), and was associated with DTC risk. Multiple logistic regression, after controlling for age, gender and the presence of a family history of thyroid cancer, showed that the odds ratios and 95% confidence intervals for the second, third and fourth quartiles of TSH levels were 1.27 (1.03-1.57), 1.55 (1.25-1.92) and 2.21 (1.78-2.74) respectively. No significant differences were observed in mean TSH levels in patients with different tumour stages and tumour sizes. Conclusion: Having a high TSH level within the normal range is an independent risk factor for DTC, and may contribute to the initiation of thyroid carcinogenesis. TSH levels in patients with thyroid nodules may be used as diagnostic adjuncts for the identification of high-risk patients, who require further investigation and/or surgical intervention.
Article
BACKGROUND The National Cancer Data Base (NCDB) represents a national electronic registry system now capturing nearly 60% of incident cancers in the U. S. In combination with other Commission on Cancer programs, the NCDB offers a working example of voluntary, accurate, cost-effective "outcomes management" on a both a local and national scale. In addition, it is of particular value in capturing clinical information concerning rare cancers, such as those of the thyroid.METHODS For the accession years 1985-1995, NCDB captured demographic, patterns-of-care, stage, treatment, and outcome information for a convenience sample of 53,856 thyroid carcinoma cases (1% of total NCDB cases). This article focuses on overall 10-year relative survival and American Joint Committee on Cancer (AJCC) (3rd/4th edition) stage-stratified 5-year relative survival for each histologic type of thyroid carcinoma. Care patterns also are discussed.RESULTSThe 10-year overall relative survival rates for U. S. patients with papillary, follicular, Hürthle cell, medullary, and undifferentiated/anaplastic carcinoma was 93%, 85%, 76%, 75%, and 14%, respectively. For papillary and follicular neoplasms, current AJCC staging failed to discriminate between patients with Stage I and II disease at 5 years. Total thyroidectomy ± lymph node sampling/dissection represented the dominant method of surgical treatment rendered to patients with papillary and follicular neoplasms. Approximately 38% of such patients receive adjuvant iodine-131 ablation/therapy. At 5 years, variation in surgical treatment (i.e., lobectomy vs. more extensive surgery) failed to translate into compelling differences in survival for any subgroup with papillary or follicular carcinoma, but longer follow-up is required to evaluate this. NCDB data appeared to validate the AMES prognostic system, as applied to papillary cases. Younger age appeared to influence prognosis favorably for all thyroid neoplasms, including medullary and undifferentiated/anaplastic carcinoma. NCDB data also revealed that unusual patients diagnosed with undifferentiated/anaplastic carcinoma before age of 45 years have better survival.CONCLUSIONS The NCDB system permits analysis of care patterns and survival for large numbers of contemporaneous U. S. patients with relatively rare neoplasms, such as thyroid carcinoma. In this context, it represents an unsurpassed clinical tool for analyzing care, evaluating prognostic models, generating new hypotheses, and overcoming the volume-related drawbacks inherent in the study of such neoplasms. [See editorial on pages 2434-6, this issue.] Cancer 1998;83:2638-2648. © 1998 American Cancer Society.
Article
Many studies on thyroid follicular tumors have reported the presence of somatic mutations to three forms of RAS: HRAS, KRAS, and NRAS. However, the frequency and clinical significance of these RAS mutations remain unclear, in large part due to the different methodologies being used for mutation analysis and the limited number of cases featured in studies. To clarify the significance of RAS mutations, we examined a large number of follicular adenomas and carcinomas obtained from a single institute using established methods for the analysis of RAS. Tumor samples from 40 follicular adenoma and 58 follicular carcinoma patients treated at the Kanagawa Cancer Center Hospital were analyzed. The three RAS mutations at codons 12 and 61 were assessed with a polymerase chain reaction-based loop-hybrid mobility shift assay followed by confirmation with direct sequencing. The relationships between mutation status and clinicopathological features at the time of the initial operation and the prognosis of the patients were also analyzed. Twelve out of 40 (30%) adenomas harbored RAS mutations. In contrast, 33 out of 58 (57%) follicular carcinomas harbored RAS mutations, and the mutation was predominantly found in the NRAS codon 61 (22/33, 67%, p<0.01). The rate of gene mutations was significantly higher in the carcinomas than in the adenomas (p<0.01). The NRAS codon 61 mutation in follicular carcinomas was positively associated with distant metastases through the entire clinical course of the patients (p<0.05), and RAS mutations were associated with poor overall patient survival (p<0.05). We investigated the frequency of RAS mutations in follicular thyroid tumors from a large number of cases obtained from a single institute. The predominance of NRAS codon 61 mutations as a feature of carcinomas indicates that the diagnosis of adenoma alongside the presence of this mutation should be made cautiously. Our study raises the possibility that follicular adenomas with the RAS mutations have an inherent malignant potential; however, the clinical significance of this finding should be further investigated in more patients and over a longer follow-up period.