ArticlePDF Available

Selenoprotein deficiency and high levels of selenium compounds can effectively inhibit hepatocarcinogenesis in transgenic mice

Authors:

Abstract and Figures

The micronutrient element selenium (Se) has been shown to be effective in reducing the incidence of cancer in animal models and human clinical trials. Selenoproteins and low molecular weight Se compounds were implicated in the chemopreventive effect, but specific mechanisms are not clear. We examined the role of Se and selenoproteins in liver tumor formation in TGFalpha/c-Myc transgenic mice, which are characterized by disrupted redox homeostasis and develop liver cancer by 6 months of age. In these mice, both Se deficiency and high levels of Se compounds suppressed hepatocarcinogenesis. In addition, both treatments induced expression of detoxification genes, increased apoptosis and inhibited cell proliferation. Within low-to-optimal levels of dietary Se, tumor formation correlated with expression of most selenoproteins. These data suggest that changes in selenoprotein expression may either suppress or promote tumorigenesis depending on cell type and genotype. Since dietary Se may have opposing effects on cancer, it is important to identify the subjects who will benefit from Se supplementation as well as those who will not.
Content may be subject to copyright.
Selenoprotein deficiency and high levels of selenium compounds can
effectively inhibit hepatocarcinogenesis in transgenic mice
Sergey V Novoselov
1
, Diego F Calvisi
2
, Vyacheslav M Labunskyy
1
, Valentina M Factor
2
,
Bradley A Carlson
3
, Dmitri E Fomenko
1
, Mohamed E Moustafa
3
, Dolph L Hatfield
3
and
Vadim N Gladyshev*
,1
1
Department of Biochemistry, University of Nebraska, N 151 Beadle Center, 1901 Vine Street, Lincoln, NE 68588, USA;
2
Laboratory
of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
20892, USA;
3
Molecular Biology of Selenium Section, Laboratory of Cancer Prevention, Center for Cancer Research, National
Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
The micronutrient element selenium (Se) has been shown
to be effective in reducing the incidence of cancer in
animal models and human clinical trials. Selenoproteins
and low molecular weight Se compounds were implicated
in the chemopreventive effect, but specific mechanisms are
not clear. We examined the role of Se and selenoproteins
in liver tumor formation in TGFa/c-Myc transgenic mice,
which are characterized by disrupted redox homeostasis
and develop liver cancer by 6 months of age. In these mice,
both Se deficiency and high levels of Se compounds
suppressed hepatocarcinogenesis. In addition, both treat-
ments induced expression of detoxification genes, in-
creased apoptosis and inhibited cell proliferation. Within
low-to-optimal levels of dietary Se, tumor formation
correlated with expression of most selenoproteins. These
data suggest that changes in selenoprotein expression may
either suppress or promote tumorigenesis depending on
cell type and genotype. Since dietary Se may have
opposing effects on cancer, it is important to identify the
subjects who will benefit from Se supplementation as well
as those who will not.
Oncogene (2005) 24, 8003–8011. doi:10.1038/sj.onc.1208940;
published online 19 September 2005
Keywords: selenoprotein; selenocysteine; selenium;
cancer prevention; redox regulation
Introduction
The micronutrient element selenium (Se) has been
shown to be effective in reducing the incidence of cancer
in animal models and human clinical trials (Clark et al.,
1996; Yu et al., 1997; Yoshizawa et al., 1998; Milner
et al., 2001; Ip et al., 2002). Of the more than 100
reported studies that have examined the chemopreven-
tive potential of Se, approximately two-thirds demon-
strated a protective effect and the remaining studies
reported no effect. A double-blind clinical trial showed
that supplementing a diet with Se resulted in a
significant decrease in cancer mortality and overall
cancer incidence (Clark et al., 1996). In this study of
1312 individuals, supplementation of the normal diet
with 200 mg of Se daily resulted in decreases of 63, 58,
and 48% in the incidence of prostate, colon, and lung
cancers, respectively (Clark et al., 1996). However, in the
same study, a chemopreventive effect of this micronu-
trient on the incidence of melanoma was not observed.
The biochemical basis for the protective effect of Se
against cancer is not well understood. A number of
mechanisms have been proposed, which are based on
two major ideas. One considers that the chemopreven-
tive action of dietary Se is mediated by an increased
abundance in Se-containing proteins, while the other
that low molecular weight Se-containing compounds (or
selenocompounds) are responsible for the chemopre-
ventive effect (Ganther, 1999; Whanger, 2004). The term
of low molecular weight selenocompounds refers herein
to Se-containing organic and inorganic compounds that
have potent cancer prevention potential. Among the
best characterized Se compounds that decrease cancer
incidence in various rodent models or inhibit growth of
human cancer cells are methylselenenic acid, methylse-
lenocysteine, and selenomethionine. Whether selenopro-
teins may provide the protection against cancers at an
optimal nutritional concentration of Se, and low
molecular weight Se compounds at higher concentra-
tion, is not known.
The predominant biological form of Se in mammals
at low, suboptimal, and optimal dietary levels of this
element is selenocysteine in proteins (Sunde, 1994).
Under these conditions, selenoproteins contain >90%
of the Se pool in mammals. Selenocysteine is encoded by
the UGA codon and is recognized as the 21st amino acid
in protein (Low and Berry, 1996; Stadtman, 1996; Bock,
2000; Hatfield, 2001; Driscoll and Copeland, 2003).
There are 25 human and 24 mouse selenoproteins
(Kryukov et al., 2003).
In the present report, we used a well-characterized
transgenic mouse model of liver cancer, in which
hepatocyte-specific coexpression of c-Myc oncogene
and transforming growth factor alpha (TGFa) leads to
Received 22 February 2005; revised 20 May 2005; accepted 15 June 2005;
published online 19 September 2005
*Correspondence: VN Gladyshev; E-mail: vgladyshev1@unl.edu
Oncogene (2005) 24, 8003–8011
&
2005 Nature Publishing Group
All rights reserved 0950-9232/05 $30.00
www.nature.com/onc
rapid hepatocellular carcinoma (HCC) development
with high penetrance (Santoni-Rugiu et al., 1996). The
trademark features of liver oncogenesis in TGFa/c-Myc
mice are chronic oxidative stress resulting from
increased production of reactive oxygen species and
genomic instability (Factor et al., 1998, 2000; Hironaka
et al., 2003; Calvisi et al., 2004). In contrast to
expectations, we found that not only supplementation
of the diet with high levels of Se but also Se deficiency
suppressed tumorigenesis in this model. Our study
implicated selenoproteins in tumor formation in
TGFa/c-Myc mice and suggested that some cancers
may be promoted by dietary Se supplementation.
Results
Regulation of selenoprotein expression in TGFa/c-Myc
mice
By performing metabolic labeling of mice with
75
Se, we
analysed the distribution of selenoproteins by SDS–
PAGE in TGFa/c-Myc mice at the early dysplastic stage
(10 weeks) and tumor stage (27 weeks) (Supplementary
Figure S1). There were no significant differences in the
labeling pattern between dysplastic livers at 2 and 6
months of age. However, in the tumors derived from
6-month-old mice, expression of glutathione peroxidase 1
(GPx1; a major 25 kDa
75
Se-labeled band) was signifi-
cantly reduced, whereas expression of thioredoxin
reductase 1 (TR1; a 55 kDa
75
Se-labeled band) appeared
to be slightly elevated (Supplementary Figure S1). These
data were consistent with the possibility that tumorigen-
esis alters selenoprotein expression in these mice. Thus, it
is also possible that changes in selenoprotein levels might
influence the incidence of cancer in TGFa/c-Myc mice.
Se diets
Regulation of selenoprotein expression in TGFa/c-Myc
mice was further analysed by changing dietary levels of
Se. Torula yeast Se-deficient diet (designated as the
0 ppm Se diet) was used. We also supplied this diet with
0.1, 0.4, and 2.25 ppm Se (0.1, 0.4, and 2.25 ppm Se
diets, respectively) (see Materials and methods for the
rationale regarding the use of diets). A fifth diet was the
0 ppm Se diet supplemented with 30 ppm triphenylsele-
nonium (TPS) chloride (designated the TPS diet). This
diet served as a control diet that achieved selenoprotein
deficiency in the presence of high levels of Se in the diet.
Regulation of selenoprotein expression by dietary Se
Expression and/or catalytic activity of several seleno-
proteins were assessed in livers of TGFa/c-Myc mice fed
the five Se diets. Glutathione peroxidase activity was
reduced approximately 25- and 50-fold in mice on the
0 ppm Se diet relative to 0.1 and 0.4 ppm Se diets,
respectively (Figure 1a). The increase in Se above
0.4 ppm (the 2.25 ppm Se diet) did not result in an
additional increase in GPx activity. GPx activity in
transgenic mice maintained on the TPS diet was similar
to that observed in mice fed the 0 ppm Se diet. Although
this enzyme activity assay measured total glutathione
peroxidase activity, GPx1 is by far the most active (and
abundant) glutathione peroxidase. Therefore, this assay
primarily measured GPx1 activity.
The activity of selenoprotein R (SelR), which is a
major methionine-R-sulfoxide reductase in mammals,
was regulated by Se similarly to the GPx activity
(Figure 1b), with a dramatic decrease in 0 ppm Se and
TPS diets and maximal activity in 0.4 and 2.25 ppm
diets. Expression of GPx1 and SelR were also assessed
by immunoblot assays. Both proteins were undetectable
in mice fed 0 ppm Se and TPS diets and were
significantly reduced in the 0.1 ppm Se dietary group
(relative to the 0.4 ppm Se group) (Figure 1d). Two
additional selenoproteins, 15 kDa selenoprotein (Sep15)
and TR1 were less responsive to changes in Se levels in
the diets (Figure 1d, e, respectively). There was little
difference in their expression between 0.1, 0.4, and
2.25 ppm Se diets. However, Se deficiency (0 ppm Se) as
well the TPS diet resulted in a significant reduction in
their expression levels (although less dramatic than that
of GPx1 and SelR). A fifth protein analysed, mitochon-
drial thioredoxin reductase 3 (TR3), did not respond
significantly to Se availability (Figure 1e).
Since selenoprotein deficiency was expected to cause
oxidative stress, we reasoned that certain compensatory
antioxidant pathways could be upregulated in the 0 ppm
Se group. We assayed catalase activity and found that
0 ppm Se and TPS diets resulted in increased catalase
activity (Figure 1c). However, there were no obvious
differences in levels of oxidized glutathione or in the
ratio of oxidized to reduced glutathione in mice
maintained on the five diets (data not shown).
Regulation of the liver/body mass ratio by Se
We observed no significant differences in body weights
between different groups of TGFa/c-Myc mice,
although mice fed 0.4 and 2.25 ppm Se diets were about
7–10% larger (39.470.2 and 38.770.6 g, respectively).
The effect of dietary Se supplementation on liver mass
was more pronounced. Mice, which received 0.1 ppm Se,
displayed a transgene-induced hepatomegaly character-
istic of fast-growing TGFa/c-Myc livers. However, the
liver/body weight ratios progressively decreased with
increase in dietary Se (6.6970.26%, P ¼ 0.013 and
5.9570.26%, P ¼ 0.06 in mice fed 0.4 and 2.25 ppm Se
diets, respectively, as compared to 8.1870.58% in the
0.1 ppm Se group). Similar decrease in liver mass was
found in mice maintained on the TPS diet
(6.0770.59%, P ¼ 0.037 as compared to the 0.1 ppm
Se group), whereas Se deficiency (0 ppm Se) did not
significantly affect liver mass (P ¼ 0.247).
Regulation of hepatic tumor formation in TGFa/c-Myc
mice by Se
TGFa/c-Myc mice develop hepatic tumors by 6–8
months of age. To determine whether and how changes
Mechanisms of cancer prevention by selenium in mice
SV Novoselov et al
8004
Oncogene
in selenoprotein expression and concentration of Se
compounds affected TGFa/c-Myc-mediated hepato-
carcinogenesis, we maintained mice on the 0 ppm Se
(n ¼ 11), 0.1 ppm Se (n ¼ 10), 0.4 ppm Se (n ¼ 12),
2.25 ppm Se (n ¼ 8), and TPS diets (n ¼ 4) starting from
weaning until 27 weeks of age. The highest incidence of
both preneoplastic and neoplastic lesions was found in
mice maintained on the 0.4 and 0.1 ppm Se diets.
Surprisingly, both Se deficiency (0 ppm Se) and high
doses of Se (2.25 ppm Se and TPS) significantly inhibited
all stages of neoplastic development, including foci,
adenomas, and carcinomas, as compared to the 0.4 ppm
Se group (Figure 2a). In addition, the tumor multiplicity
was also significantly reduced in these three groups of
Figure 1 Regulation of selenoprotein expression and antioxidant enzyme activities by dietary selenium in livers of TGFa/c-Myc mice.
Glutathione peroxidase (a), SelR (methionine-R-sulfoxide reductase) (b) and catalase (c) activities were analysed in crude extract
samples (n ¼ 8 for each activity) prepared from mice maintained on five different Se diets. (d) Expression of Sep15 (upper panel), SelR
(second panel) and GPx1 (third panel) was analysed by immunoblot assays. The lower panel represents a control and shows a pattern
of Coomassie Blue-staining for samples used in the upper two panels. Expression is shown for two mice fed the 0 ppm Se diet, two mice
the 0.1 ppm Se diet,two mice the 0.4 ppm Se diet, one mouse the 2.25 Se diet and two mice the TPS diet. Molecular weights of Sep15,
SelR and GPx1 based on SDS–PAGE gel migration are shown on the right. (e) Regulation of expression of TR1 and TR3. Thioredoxin
reductases present in liver homogenates were enriched on ADP-Sepharose columns and their levels were determined by immunoblot
assays. For each dietary group, homogenates from two different mice were combined. (f) Expression of 3b hydroxysteroid
dehydrogenase (3bHSD) and pancreatitis-associated protein (PAP) was assayed by immunoblot assays using the samples shown in
panel (d)
Mechanisms of cancer prevention by selenium in mice
SV Novoselov et al
8005
Oncogene
mice (Figure 2b). Thus, in mice fed the 0 ppm Se diet, on
average, only one tumor per mouse was found
compared to an average of 7.5 tumors in the 0.4 ppm
Se group (P ¼ 0.007). Morphologically, Se supplementa-
tion did not affect the phenotypes of TGFa/c-Myc
tumors. The vast majority of double transgenic mice
developed hepatocellular adenomas and carcinomas
with small/clear cell phenotype.
Hepatic dysplasia was evident in all livers from
TGFa/c-Myc transgenic mice regardless of the diet.
However, the degree of dysplasia was strikingly different
between the five dietary groups. TGFa/c-Myc transgenic
mice maintained on 0.1 and 0.4 ppm Se diets showed
highly proliferative diffuse small cell dysplasia resulting
in a marked disorganization of the lobular architecture
(Figure 3a). In contrast, mice receiving 0 or 2.25 ppm Se
displayed mainly large dysplastic cells undergoing a
frequent apoptotic death (Figure 3b–d). Furthermore,
large areas of necrosis and inflammatory infiltrates
within preneoplastic and neoplastic hepatic lesions were
detected in TGFa/c-Myc mice fed the 2.25 ppm Se diet
(Figure 3d). Transgenes probably caused or contributed
to the observed lesions and infiltrates as inflammation
and neoplasia were not evident in control (wild type)
mice receiving 2.25 ppm Se (data not shown).
To better define the effect of Se treatment on tumor
development, we next determined mitotic and apoptotic
indices in TGFa/c-Myc livers (Figure 2c, d). The highest
rate of cell proliferation was found in mice receiving
0.4 ppm Se when compared with that in either 0 ppm,
2.25 ppm or TPS groups (P ¼ 0.001, 6.25E-05, 0.049,
respectively) (Figure 3a). In contrast, the rate of
apoptosis was significantly higher in mice fed 2.25 ppm
Se, 0 ppm Se, and TPS diets (P ¼ 4.6E-06, 3E-06, 0.0001
vs the 0.4 ppm Se group, respectively) (Figures 2d, 3b).
The resulting mitosis/apoptosis ratios were considerably
reduced in the 2.25 ppm Se, 0 ppm Se and TPS dietary
groups coincidently with decreased tumor growth. Of
importance, the most frequent apoptotic cell death was
found in association with extensive inflammatory
infiltrates found in mice fed 2.25 ppm Se diet suggesting
that high levels of Se might promote immune surveil-
lance (Figure 3d). Taken together, these data indicate
that both low and high levels of dietary Se decrease the
severity of dysplastic lesions and tumor formation in
TGFa/c-Myc transgenic mice. In particular, high levels
of Se might inhibit liver tumor development by inducing
a proapoptotic state and favoring immune response
against the tumor.
Gene expression changes in response to Se deficiency and
high Se in TGFa/c-Myc mice
To gain further insights into the mechanism of
suppressed hepatocarcinogenesis in TGFa/c-Myc mice
maintained on the Se-deficient (0 ppm Se) and high Se
(2.25 ppm. Se) diets, we compared gene expression
profiles using Affymetrix mouse oligonucleotide chips
representing B60% of mouse genes (Supplementary
Figure S2). The analysis was performed at the stage
preceding major tumor formation (21 weeks on the diet)
on two mice from each (0, 0.4 and 2.25 ppm Se) dietary
groups (the full data set is at http://genomics.unl.edu/
CANCER). Changes in gene expression in response to
Figure 2 Selenium-deficient diet and diets with high levels of selenium inhibit hepatocarcinogenesis in TGFa/c-Myc transgenic mice.
(a) Frequencies of foci, adenomas and carcinomas in TGFa/c-Myc mice maintained on different Se diets. All mice fed the 0.4 ppm Se
diet developed foci and adenomas, and 11 out of 12 developed carcinomas. The frequencies of foci, adenomas and carcinomas were
significantly reduced in mice on 0 ppm Se (P ¼ 0.008, 0.001, 0.01), 2.25 ppm Se (P ¼ 7.24E-05, 0.04, 0.0003), and TPS (P ¼ 0.007, 0.003,
0.06) as compared to corresponding lesion frequencies in the 0.4 ppm Se group. (b) Multiplicity of tumors in mice maintained on the
five Se diets. (c) Mitotic indices in TGFa/c-Myc livers. (d) Apoptosis indices in TGFa/c-Myc livers
Mechanisms of cancer prevention by selenium in mice
SV Novoselov et al
8006
Oncogene
dietary Se deficiency (0 ppm Se) or high levels of Se
(2.25 ppm Se) were compared vs the 0.4 ppm Se group.
Subsequently, genes affected in 0 and 2.25 ppm Se
samples were compared against each other to identify
unique and common alterations in gene expression
between these two groups.
As expected, we found that several selenoprotein
genes showed decreased mRNA levels in mice fed the
0 ppm Se diet (Table 1), but these did not change when
0.4 and 2.25 ppm Se samples were compared. Additional
unique responses of 0 and 2.25 ppm Se groups included
genes involved in cell cycle, apoptosis, transcription,
redox regulation, fatty acid metabolism, protein folding
and immune/inflammatory response (Supplementary
Tables S1 and S2).
Surprisingly, we also observed a very significant
coregulation of genes in response to 0 and 2.25 ppm Se
diets (Supplementary Figure S3 and Table S3). Among
the genes with elevated expression in both 0 and
2.25 ppm Se groups, most prominent were genes
responsible for detoxification and repair, as well as the
proteins involved in redox regulation (Table 2). In
particular, a number of glutathione-S-transferases and
cytochrome P450s manifested dramatically elevated
gene expression.
These observations are consistent with the idea
that repair/detoxification systems are upregulated
during both Se deficiency and exposure to high levels
of Se, thereby contributing to suppression of tumor-
igenesis observed in TGFa/c-Myc transgenic mice.
Although a possible contribution of variable amounts
of neoplastic tissue to some of the observed effects
in gene expression cannot be excluded, we also
observed increased expression of certain detoxification
enzymes in other animal models subjected to Se diets
(unpublished data).
To verify the microarray data, we assayed expression
of two proteins, 3b hydroxysteroid dehydrogenase
(3bHSD) and pancreatitis-associated protein (PAP)
(Figure 1f). Expression of 3bHSD and PAP mRNAs
was strongly affected by both 0 and 2.25 ppm Se diets
(relative to the 0.4 ppm Se diet) (Table 2 and Supple-
mentary Table S3), but they responded in an opposite
manner to the Se diets. Consistent with these mRNA
Figure 3 Liver histopathology in TGFa/c-Myc mice fed different Se diets. (a) Widespread small cell liver dysplasia in transgenic
mouse receiving the 0.4 ppm Se diet. Note the presence of frequent mitosis (arrows). (b,c) Large cell dysplasia limited to pericentral
areas of hepatic lobules in mice from the 0 ppm Se (b) and 2.25 ppm Se groups (c). Note the presence of multiple apoptotic bodies in c
(arrows). (d) Diffuse liver necrosis accompanied by inflammatory infiltrate in TGFa/c-Myc mice fed the 2.25 ppm Se diet. Inset,
Eosinophilic focal lesion displaying inflammatory infiltrate leading to extensive apoptosis as indicated by brown ApoTag staining
Mechanisms of cancer prevention by selenium in mice
SV Novoselov et al
8007
Oncogene
changes, 3bHSD expression increased in response to
both the 0 and 2.25 ppm Se diets, whereas expression of
PAP decreased in both diets, relative to the 0.4 ppm Se
diet (Figure 1f).
Discussion
The goal of this study was to examine the cancer
preventive effect of selenoproteins and low molecular
weight Se compounds in the same animal model. The
TGFa/c-Myc double transgenic mice, which exhibit a
high penetrance of liver cancer, were chosen as a well
characterized model relevant to human liver disease
(Santoni-Rugiu et al ., 1996; Factor et al., 1998;
Hironaka et al., 2003). In these mice, constitutive
coexpression of c-Myc and TGFa in the liver generates
an oxidative stress environment prior to tumor devel-
opment and predisposes mice to a 100% incidence of
HCC by 6–8 months of age (Calvisi et al., 2004).
Importantly, vitamin E, a potent free radical scavenging
antioxidant, was able to protect liver tissue against
oxidative stress and suppress hepatic tumor formation in
TGFa/c-Myc mice (Factor et al., 2000).
Admittedly, we initially anticipated that optimally
increased levels of Se (0.4 ppm) would similarly normal-
ize redox homeostasis by elevating expression of certain
selenoproteins and hypothesized that it would result in
suppression of the tumorigenesis process in the TGFa/c-
Myc mice. On the contrary, we found that selenoprotein
deficiency significantly suppressed hepatic tumor forma-
Table 1 Changes in selenoprotein gene expression in response to low and high selenium in livers of TGFa/c-Myc mice
Selenoprotein Fold change (0 vs
0.4 ppm Se diet)
Fold change (2.25 vs
0.4 ppm Se diet)
Glutathione peroxidase 1 (GPx1) 2.2 k No change
Glutathione peroxidase 2 (GPx2) No change No change
Glutathione peroxidase 3 (GPx3) No change No change
Phospholipid hydroperoxide glutathione peroxidase (PHGPX; GPx4) No change No change
Thioredoxin reductase 1 (TR1, TxnRd1, TrxR1) 2.17 k No change
Thioredoxin/glutathione reductase (TGR) No change No change
Thyroid hormone deiodinase 1 (D1) 3.2 k No change
15 kDa selenoprotein (Sep15) No change No change
Selenoprotein M (SelM) No change No change
Selenoprotein K (SelK) No change No change
Selenoprotein R (methionine-R-sulfoxide reductase 1; MsrB1; SelR; SelX) No change No change
Selenoprotein W (SelW) 2.2 k No change
Fold change is shown for mice fed the 0 ppm Se diet relative to the 0.4 ppm Se diet and for mice fed the 2.25 ppm Se diet relative to the 0.4 ppm Se
diet. TGFa/c-Myc mice (24 months old) were used, which were fed Se diets from weaning. Each dietary group had two independent samples and the
average fold change in mRNA levels is shown
Table 2 Changes in expression of repair and detoxification genes in liver in response to low and high selenium in livers of TGFa/c-Myc mice
Accession number 2.25 ppm Se 0 ppm Se Annotation
NM_008295 m46.85 m28.84 3b hydroxysteroid dehydrogenase
NM_010357 m18.7 m29.86 Glutathione S-transferase, alpha 4
NM_053262 m8.28 m10.2 Dehydrogenase/reductase, SDR family
NM_023617 m6.73 m8.28 Aldehyde oxidase 3
NM_007825 m4.14 m3.86 Cytochrome P450, family 7, subfamily b, polypeptide 1
BC018344 m3.19 m4.29 Cytochrome P450, family 2, subfamily d, polypeptide 13
BC026757 m2.93 m2.22 Hydroxysteroid dehydrogenase-2, delta
NM_008182 m2.22 m6.73 Glutathione S-transferase, alpha 2, Yc2
BC010973 m2 m2.83 Cytochrome P450, family 8, subfamily b, polypeptide 1
BC012707 m2 m3.14 Glutathione S-transferase, theta 2
AF276917 m2 m2.55 Glutaredoxin 1, thioltransferase
NM_010000 NC m7.73 Cytochrome P450, family 2, subfamily b, polypeptide 9
NM_007812 NC m3.61 Cytochrome P450, family 2, subfamily a, polypeptide 4
AF128849 NC m6.28 Cytochrome P450, family 2, subfamily b, polypeptide 20
AB041034 NC m2.46 NADPH oxidase 4
BE952632 NC m2.38 2-4-dienoyl-Coenzyme A reductase 2, peroxisomal
BC020001 NC m2.22 Aldehyde dehydrogenase 1 family, member B1
NM_010191 NC m2.17 Farnesyl diphosphate farnesyl transferase 1
NM_134072 NC m2.07 3-alpha-hydroxysteroid dehydrogenase type 1
BC025822 NC m2.07 Cytochrome P450, family 2, subfamily c, polypeptide 70
Fold change is shown for mice fed the 0 ppm Se diet relative to the 0.4 ppm Se diet and for mice fed the 2.25 ppm Se diet relative to the 0.4 ppm Se
diet. TGFa/c-Myc mice (24 months old) were used, which were fed Se diets from weaning. Each dietary group had two independent samples and
the average fold change in mRNA levels is shown
Mechanisms of cancer prevention by selenium in mice
SV Novoselov et al
8008
Oncogene
tion. To our knowledge, this is the first demonstration of
a lower HCC incidence in response to Se deficiency.
The increase in dietary Se levels from 0 to 0.4 ppm of
Se resulted in elevated expression of various selenopro-
teins, as assessed by Affymetrix chips, enzyme assays,
and immunoblot analyses. Antioxidant selenoproteins
GPx1 and SelR were particularly responsive to Se
supplementation. Sep15 and TR1 also responded,
although the changes in expression were less pro-
nounced. Expression of TR3 remained constant regard-
less of the Se diet. At low levels of dietary Se,
selenoproteins account for the major part of Se in the
body. Thus, there was a clear correlation between
overall selenoprotein expression and increased tumor-
igenesis in the TGFa/c-Myc mice.
Although both tumor incidence and tumor multi-
plicity were similarly decreased in mice receiving high
levels of Se in the diet, this decrease did not correlate
with selenoprotein expression, as the 0.4 ppm Se already
saturated selenoprotein expression. These data suggest
that low molecular weight Se compounds are respon-
sible for the chemopreventive effect of higher doses of
dietary Se in the TGFa/c-Myc mice. Thus, both
selenoprotein deficiency and high levels of Se com-
pounds independently inhibited tumorigenesis in TGFa/
c-Myc mice. Such a correlated effect of Se deficiency and
high levels of Se (as compared to optimal Se levels) has
not been previously reported.
Moreover, both Se deficient and high Se diets resulted
in decreases in cell proliferation and increases in
apoptosis. A likely possibility is that high doses of Se
(2.25 ppm Se and TPS) are cytotoxic, particularly for
preneoplastic and cancer cells. It appears that decreased
expression of selenoproteins observed in mice fed the
0 ppm Se diet was also cytotoxic as it promoted
apoptosis and decreased the frequency of mitosis
concomitantly with inhibition of tumorigenesis. In
addition, we observed upregulation of mRNAs for
repair and detoxification proteins, such as glutathione-
S-transferases, cytochrome P450s, and glutaredoxin,
under conditions of both Se deficiency (0 ppm Se)
and high levels of Se (2.25 ppm Se). Although our study
did not employ chemicals to induce tumorigenesis, it
seems possible that the low levels of antioxidant
selenoproteins may have increased the demand for
detoxification of endogenous toxic compounds and for
repair of damaged macromolecules. Thus, cytotoxicity
may be a common feature associated with both Se
deficiency and high levels of Se suggesting that
detoxification and repair mechanisms may contribute
to or be responsible for suppression of tumorigenesis in
this animal model.
Our findings are consistent with the known inhibitory
effect of antioxidant selenoproteins on apoptosis (Fu
et al., 2001; Gouaze et al., 2002). Previous studies
reported elevated expression of detoxification and repair
enzymes in Se-deficient mouse livers (Reiter and
Wendel, 1984; Carlson et al., 2004), as well as in cells
treated with high levels of Se (El-Bayoumy and Sinha,
2004; Zhao et al., 2004). In addition, Se deficiency had a
protective effect against acetaminophen and aflatoxin
injuries in rat livers, presumably due to stimulation
of GSH synthesis and induction of expression of
detoxification enzymes (Burk and Lane, 1983; Hill and
Burk, 1984).
Our data also agree with the recent report that Se
deficiency abrogated peritoneal plasmacytomas in
BALB/c mice, the experimental model of inflamma-
tion-dependent plasma cell transformation (Felix et al.,
2004). Thus, the role of Se and selenoproteins in cancer
and the other findings reported in our study are likely
not limited to strain, organ, and type of tumor that were
examined.
When used in physiological levels, Se is often viewed
as antioxidant that works in concert with vitamin E.
However, vitamin E decreased tumorigenesis in TGFa/
c-Myc mice (Factor et al., 2000), suggesting that at least
in this mouse model, these compounds may have
opposite effects. Besides, different selenoproteins may
influence carcinogenesis in various ways (Gladyshev
et al., 1998). Whereas some, such as GPx1, are
antioxidant proteins with well-defined functions, the
contribution of other selenoproteins may be more
complex. For example, TR1, the enzyme controlling
the redox state of thioredoxin, is implicated in a myriad
of cellular functions (Arner and Holmgren, 2000).
Thioredoxin serves as an antioxidant and antitumor
protein by reducing thioredoxin peroxidase and con-
trolling the redox state of tumor suppressor p53.
However, it also reduces various transcription factors
and provides electrons for deoxyribonucleotide bio-
synthesis. Thus, Se deficiency, by suppressing TR1
expression, may reduce the antioxidant and tumor
suppressor capacity of the thioredoxin system as well
as inhibit growth control functions. It is possible that in
the TGFa/c-Myc model, growth inhibitory functions of
Se deficiency prevail over antioxidant functions thereby
inhibiting carcinogenesis.
Our data have important (although indirect) implica-
tions for the use of dietary Se in cancer prevention in
humans. Optimal levels of dietary Se, which are slightly
above those needed for maximal expression of seleno-
proteins, were thought to provide health benefits,
particularly in reducing cancer incidence (Rayman,
2000; Hatfield, 2001; Burk, 2002). However, our new
findings suggest a possibility that elevated Se might also
promote tumorigenesis in certain genetic backgrounds.
Thus, whereas the overall effect of Se, when assessed in
large population groups (Clark et al., 1996; Yu et al.,
1997), may be positive, there might be individuals, in
which Se supplementation may promote tumor forma-
tion. Although our studies cannot be directly applied to
humans (e.g. severe Se deficiency is unlikely to occur in
humans unless there is a disruption in the pathway of Se
insertion), they establish a precedent for a similar effect
of Se deficiency and high levels of Se on cancer incidence
in the same model system.
Given the promising results of previous clinical trials
involving dietary Se (Clark et al., 1996, Yu et al., 1997),
a number of new trials have been initiated, including the
Se and Vitamin E Cancer Prevention Trial (SELECT)
that involves 32 400 male subjects (Klein et al., 2000).
Mechanisms of cancer prevention by selenium in mice
SV Novoselov et al
8009
Oncogene
Despite the extent of ongoing clinical trials, basic
knowledge of the types of cancer that are prevented by
Se and the mechanism of the chemopreventive effect is
lacking. If the effect of Se on hepatocarcinogenesis that
we observed in the TGFa/c-Myc mice can be seen in
other mouse models or in subsets of the human
population, it would mean that certain human cancers
may be promoted by Se supplementation. Thus, rather
than generally increasing the intake of Se, it may be
important to identify segments of the human population
that can benefit from Se supplementation as well as
those in which Se supplementation can increase cancer
incidence.
Materials and methods
Mice and diets
Double transgenic TGFa/c-Myc mice were generated as
described (Santoni-Rugiu et al., 1996). Five different diets
(Harland TekLad, Madison, WI, USA) were used: 0 ppm Se
(Se-deficient), 0.1 ppm Se, 0.4 ppm Se, 2.25 ppm Se, and
30 ppm TPS. The diets were based on the Torula yeast Se-
deficient diet. Se was provided in the form of sodium selenite
(except for the TPS diet).
Previous reports indicated that B0.1 ppm Se is a minimal
amount of Se that is sufficient for maximal expression of
GPx1. Since maximal expression of plasma GPx is achieved at
B55 mg/day Se in humans (Burk, 2002), the 0.1 ppm Se mouse
diet may be viewed as approximately corresponding to the
human Recommended Dietary Allowance for adults. By
analogy, the 0.4 ppm Se diet would correspond to supplemen-
tation of the human diet with 200 mg Se/day, which is the
common amount of Se used in clinical trials (Clark et al.,
1996). The rationale for the 2.25 ppm Se diet was based on the
average amount of Se that was required for the chemopreven-
tive effect of Se against chemically induced carcinogenesis in
rodents (Ganther, 1999; Ip et al., 2002; Whanger, 2004).
Finally, TPS was recently introduced as a cancer preventive
compound (Ip et al., 1998). Even at very high (30 ppm) levels,
TPS-derived Se was not accessible for insertion into seleno-
proteins, and thus, high levels of low molecular weight Se
compounds were achieved under conditions of selenoprotein
deficiency (Ip et al., 1998). Se-deficient and high Se diets have
been used in numerous previous studies involving rodents.
These diets, as well as the TPS diet, provide excellent tools for
the studies on the mechanisms of cancer prevention by Se in
animal model systems. One limitation of these diets, however,
is that the data can only partially be extrapolated to humans,
as severe Se-deficient and Se-enriched conditions are unlikely
to occur in humans.
Prior to the TGFa/c-Myc dietary study, we tested the Se
diets using wild-type mice, which were housed in groups of
three. After 2 months on the diets, the mice were killed and
GPx1 activity was determined in each dietary group. The mice
fed the 0 ppm Se diet had 3.6% of the GPx1 activity of mice on
the 0.4 ppm Se diet. Likewise, the mice on the TPS diet had
4.5% of the GPx1 activity of the 0.4 ppm Se control. In
addition, the diets were documented by analysing the amount
of Se in the livers of wild-type mice fed 0.1 and 0 ppm Se diets.
We found that the 0 ppm Se livers had 10 times lower total
Se content than the 0.1 ppm Se livers. The TGFa/c-Myc mice
were housed in groups of 2–5 and had free access to food and
water. The dietary interventions started from weaning.
Histology and tumor sampling
The mice were killed at 27 weeks of age by CO
2
inhalation. The
liver and body weights as well as the number and the size of all
grossly visible tumors were recorded. Representative samples
of liver taken from each lobe were fixed in 10% neutral
formalin at 41C overnight or frozen in liquid nitrogen and
stored at 801C. Paraffin sections (5 mm) were stained with
hematoxylin–eosin, and hepatocellular lesions were graded as
foci, adenomas and carcinomas as described (Calvisi et al .,
2004). The data are presented as mean7s.e. For statistical
comparison the Student’s t-test was used.
Mitotic and apoptotic indices
Mitotic and apoptotic indices were scored in TGFa/c-Myc
livers from at least four mice per diet group. Mitotic index was
determined by counting mitotic figures on H&E-stained slides.
For the apoptotic index, sections were stained with the
ApoTag peroxidase in situ apoptosis detection kit (Serologicals
Corporation, Norcross, GA, USA), and positive cells were
counted under a light microscope at a magnification 200.
Apoptotic cells, single, and closely clustered apoptotic bodies
were all scored as single events. Mitotic and apoptotic indices
were represented as a percentage (mean7s.e.) of the total cells
counted (at least 2000 cells per animal).
Metabolic labeling
To label selenoproteins in TGFa/c-Myc mice, animals were
injected with freshly neutralized
75
Se[selenite] (Research
Reactor Facility, University of Missouri, Columbia, MO,
USA), and after 48 h, the mice were killed, tissues removed,
and proteins extracted, electrophoresed, and transferred onto
PVDF membranes. Resulting transblots were analysed with a
PhosphorImager.
Enzyme assays
GPx1 and catalase activities were assayed using BIOXITECH
GPx-340 and BIOXITECH Catalase-520 kits (OxisResearch,
Portland, OR, USA), respectively, according to the manufac-
turer’s protocol. The methionine-R-sulfoxide reductase activ-
ity of SelR was assayed as described previously (Kim and
Gladyshev, 2004). Liver homogenates from eight animals from
each group (except the TPS dietary group that had four
animals) were used and each sample was analysed in triplicate.
Selenoprotein expression analyses
TR1 (cytosolic enzyme) and TR3 (this mitochondrial enzyme
is also called TnxRd2 and TrxR2) expression was assessed by
immunoblot assays with antibodies specific for each isozyme,
after the enzymes were enriched by binding to ADP-Sepharose
as described previously (Moustafa et al., 2001). Expression of
SelR, GPx1, and Sep15 was analysed by immunoblot assays
with antibodies specific for these proteins.
Gene expression analyses
Affymetrix GeneChip System (Affymetrix, Santa Clara, CA,
USA) was used to carry out gene expression analyses. Total
RNA was extracted from two 0 ppm Se, two 0.4 ppm Se, and
two 2.25 ppm Se frozen mouse livers using Trizol Reagent
(Invitrogen, Carlsbad, CA, USA) and RNeasy columns
(Qiagen, Valencia, CA, USA). Double-stranded cDNA synth-
esis and biotinylation was carried out as described in the
Expression Analysis Technical Manual (Affymetrix). Mouse
15K arrays were hybridized and scanned using Affymetrix
Mechanisms of cancer prevention by selenium in mice
SV Novoselov et al
8010
Oncogene
GeneChip Scanner 3000. To compare gene expression, the data
generated from the 0 ppm Se samples were averaged and
analysed against averaged 0.4 ppm Se data. Likewise, the
2.25 ppm Se data were averaged and analysed against the
0.4 ppm Se data. Expression of 3bHSD and PAP were assayed
by immunoblot assays with antibodies specific for each protein
(kindly provided by Dr Ian Mason and purchased from R&D
Systems, Minneapolis, MN, USA, respectively).
Abbreviations
GPx1, glutathione peroxidase 1; HCC, hepatocellular carci-
noma; Se, selenium; SelR, selenoprotein R; Sep15, 15 kDa
selenoprotein; TGFa, transforming growth factor a; TPS,
triphenylselenonium chloride; TR1, thioredoxin reductase 1
(cytosolic enzyme* also called TxnRd1 and TrxR1); TR3,
thioredoxin reductase 3 (mitochondrial enzyme* also called
TxnRd2 and TrxR2).
Acknowledgements
Services of the University of Nebraska-Lincoln genomics
facility, headed by Dr Yuannan Xia, and the bioinformatics
facility, headed by Dr Guoqing Lu, are acknowledged.
We thank Dr Ian Mason for providing hydroxysteroid
dehydrogenase antibodies and Dr Hwa-Young Kim for help
with methionine sulfoxide reductase assays. Supported by
NIH CA080946 and in part by GM061603 and AG021518
(to VNG).
References
Arner ES and Holmgren A. (2000). Eur. J. Biochem., 267,
6102–6109.
Bock A. (2000). Biofactors, 11, 77–78.
Burk RF. (2002). Nutr. Clin. Care, 5, 75–79.
Burk RF and Lane JM. (1983). Fundam. Appl. Toxicol., 3,
218–221.
Calvisi DF, Factor VM, Ladu S, Conner EA and Thorgeirsson
SS. (2004). Gastroenterology, 126, 1374–1386.
Carlson BA, Novoselov SV, Kumaraswamy E, Lee BJ, Anver
MR, Gladyshev VN and Hatfield DL. (2004). J. Biol. Chem.,
279, 8011–8017.
Clark LC, Combs GF, Turnbull BW, Slate EH, Chalker DK,
Chow J, Davis LS, Glover RA, Graham GF, Gross EG,
Krongrad A, Lesher Jr JL, Park HK, Sanders Jr BB,
Smith CL and Taylor JR. (1996). J. Am. Med. Assoc., 276,
1957–1963.
Driscoll DM and Copeland PR. (2003). Annu. Rev. Nutr., 23,
17–40.
El-Bayoumy K and Sinha R. (2004). Mutat. Res., 551, 181–197.
Factor VM, Kiss A, Woitach JT, Wirth PJ and Thorgeirsson
SS. (1998). J. Biol. Chem., 273, 15846–15853.
Factor VM, Laskowska D, Jensen MR, Woitach JT, Popescu
NC and Thorgeirsson SS. (2000). Proc. Natl. Acad. Sci.
USA, 97, 2196–2201.
Felix K, Gerstmeier S, Kyriakopoulos A, Howard OM, Dong
HF, Eckhaus M, Behne D, Bornkamm GW and Janz S.
(2004). Cancer Res., 64, 2910–2917.
Fu Y, Sies H and Lei XG. (2001). J. Biol. Chem., 276,
43004–43009.
Ganther HE. (1999). Carcinogenesis, 20, 1657–1666.
Gladyshev VN, Factor VM, Housseau F and Hatfield DL.
(1998). Biochem. Biophys. Res. Commun., 251, 488–493.
Gouaze V, Andrieu-Abadie N, Cuvillier O, Malagarie-
Cazenave S, Frisach MF, Mirault ME and Levade T.
(2002). J. Biol. Chem., 277, 42867–42874.
Hatfield DL (ed) (2001). Selenium: Its Molecular Biology and
Role in Human Health. Kluwer Academic Publishers:
Norwell, MA.
Hill KE and Burk RF. (1984). Toxicol. Appl. Pharmacol., 72,
32–39.
Hironaka K, Factor VM, Calvisi DF, Conner EA and
Thorgeirsson SS. (2003). Lab. Invest., 83, 643–654.
Ip C, Dong Y and Ganther HE. (2002). Cancer Metastasis
Rev., 21, 281–289.
Ip C, Thompson HJ and Ganther HE. (1998). Anticancer Res.,
18, 9–12.
Kim HY and Gladyshev VN. (2004). Mol. Biol. Cell, 15,
1055–1064.
Klein EA, Thompson IM, Lippman SM, Goodman PJ,
Albanes D, Taylor PR and Coltman C. (2000). Prostate
Cancer Prostatic Dis., 3, 145–151.
Kryukov GV, Castellano S, Novoselov SV, Lobanov AV,
Zehtab O, Guigo R and Gladyshev VN. (2003). Science, 300,
1439–1443.
Low SC and Berry MJ. (1996). Trends Biochem. Sci., 21,
203–208.
Milner JA, McDonald SS, Anderson DE and Greenwald P.
(2001). Nutr. Cancer, 41, 1–16.
Moustafa ME, Carlson BA, El-Saadani MA, Kryukov GV,
Sun QA, Harney JW, Hill KE, Combs GF, Feigenbaum L,
Mansur DB, Burk RF, Berry MJ, Diamond AM, Lee BJ,
Gladyshev VN and Hatfield DL. (2001). tRNA. Mol. Cell.
Biol., 21, 3840–3852.
Rayman MP. (2000). Lancet, 356, 233–241.
Reiter R and Wendel A. (1984). Biochem. Pharmacol., 33,
1923–1928.
Santoni-Rugiu E, Nagy P, Jensen MR, Factor VM and
Thorgeirsson SS. (1996). Am. J. Pathol., 149, 407–428.
Stadtman TC. (1996). Ann. Rev. Biochem., 65, 83–100.
Sunde RA. (1994). Selenium in Biology and Human Health.RF
Burk (ed). Springer-Verlag: New York, pp 45–59.
Whanger PD. (2004). Br. J. Nutr., 91, 11–28.
Yoshizawa K, Willett WC, Morris SJ, Stampfer MJ, Spiegel-
man D, Rimm EB and Giovannucci E. (1998). J. Natl.
Cancer Inst., 90, 1219–1224.
Yu SY, Zhu YJ and Li WG. (1997). Biol. Trace Elements Res.,
56, 117–124.
Zhao H, Whitfield ML, Xu T, Botstein D and Brooks JD.
(2004). Mol. Biol. Cell., 15, 506–519.
Supplementary Information accompanies the paper on Oncogene website (http://www.nature.com/onc)
Mechanisms of cancer prevention by selenium in mice
SV Novoselov et al
8011
Oncogene
... Using immunohistochemistry with caspase-3 antibodies, one study reported that in cancer patients, elevated CASP3 was linked to an increased risk of cancer return and a reduced life expectancy [73]. Interestingly, the triggering of cell death by Se has been found to decrease the development of HCC cells [74][75][76] via activating p53 [77], Bax [78,79], CASP 3 [78][79][80][81][82] and C. CASP3 [83] whereas lowering Bcl2 levels after utilizing Se [77,78,84] and SeNps [85]. Similar effects reported in this study strengthens the hypothesis that synergism between SOR/SeNps has enhanced efficacy in treatment. ...
... The PI3K/AKT/mTOR beside Ras/Raf/MAPK cascades are targeted by SOR, which reduces HCC cell proliferation and invasion [70,86,87]. There is scientific evidence to show that Se can reduce cell growth, invasion and metastasis [21,76,88]. The capacity of liver cancer cells to migrate is boosted during the treatment resistance phase [89]. ...
Article
Aims The first-line treatment for advanced hepatocellular carcinoma (HCC) is the multikinase inhibitor sorafenib (SOR). Sofafenib resistance is linked to protein kinase B/ mammalian target of rapamycin (AKT/mTOR) and nuclear factor kappa B (NF-κB) activation, apoptosis inhibition and oxidative stress. This study investigated selenium nanoparticles (SeNps) to overcome SOR resistance in thioacetamide (TAA) induced HCC in rats. Materials and methods TAA (200 mg/kg/twice weekly, i.p.) was administered for 16 weeks to induce HCC.s. Rats were treated with oral SOR (10 mg/Kg daily), selenium, and SeNps (5 mg/kg three times/week) alone or in combination, for two weeks. Apoptosis, proliferation, angiogenesis, metastasis and drug resistance were assessed. Cleaved caspase 3 (C. CASP3), mTOR, and NF-κB were determined by western blotting. Expression of p53 gene and long-noncoding RNA-AF085935 was determined by qRT-PCR. Expression of B- Cell Leukemia/Lymphoma 2 (Bcl2), Bcl associated X protein (Bax)and glypican 3 (GPC3) was determined by enzyme-linked immunosorbent assay. Liver functions, antioxidant capacity, histopathology and CD34 immunohistochemistry were performed. Key findings SOR/SeNps reversed TAA-induced HCC in rats, through reduction of oxidative stress, activation of p53, Bax and CASP3, and inhibition of Bcl2. SOR/SeNps ameliorated the HCC-induced effect on cell proliferation and drug resistance by targeting mTOR and NF-κB pathways. SOR/SeNps decreased CD34 immunostaining indicating a decrease in angiogenesis and metastasis. SOR/SeNps regulated HCC epigenetically through the lncRNA-AF085935/GPC3 axis. Significance SOR/SeNps are a promising combination for tumor suppression and overcoming sorafenib resistance in HCC by modulating apoptosis, AKT/mTOR and NF-κB pathways, as well as CD34 and lncRNA-AF085935/GPC3 axis.
... Селен также обладает стимулирующим влиянием в отношении Bifi dobacterium [49]. Однако, несмотря на иную локализацию, стоит отметить, что телята, содержащиеся на рационе из обогащенной селеном люцерны, также характеризуются более высоким биоразнообразием назофарингеальной микрофлоры [50], что может обусловливать снижение заболеваемости респираторными инфекциями и частично нивелировать их негативные последствия для организма [51]. Таким образом, обмен селена в организме оказывает значительное влияние на микробиом различных биотопов, в том числе кишечника, что может по крайней мере частично опосредовать часть биологических эффектов селена на продуктивные качества крупного рогатого скота. ...
Article
Full-text available
Selenium is one of the most important essential elements for immune function and general health of cattle. The role of selenium in maintaining animal health is primarily based on the functions of selenocysteine-containing proteins, many of which have antioxidant activity. Role of selenium in Animal Health is primarily based on the functions of selenocysteine-containing proteins, many of which possess antioxidant activity. Insufficient selenium intake can be accompanied by subclinical diseases, which leads to a decrease in productivity. It was determined that the inclusion of additional sources of selenium has a significant effect on live weight gain, quality characteristics of meat, as well as the intestinal microbiome in cattle and other types of farm animals (pigs, poultry). However, despite the rather high level of knowledge of the selenium role in the implementation of the functional traits in farm animals, there is still no consensus in the scientific community on the optimal dosages and methods of feeding selenium, which reduces the effectiveness of the use of selenium-containing preparations in practical animal husbandry.
... Moreover, Sep15 expression is induced when misfolded proteins are accumulated in the ER [34]. The expression of Sep15 is dependent on dietary selenium; however, under selenium deficiency, the decrease in Sep15 in the brain and the testis is not a very marked one [8,35]. Sep15 has also been included in the stress-related selenoproteins group. ...
Article
Full-text available
Selenium (Se) is a metalloid that is recognized as one of the vital trace elements in our body and plays multiple biological roles, largely mediated by proteins containing selenium-selenoproteins. Selenoproteins mainly have oxidoreductase functions but are also involved in many different molecular signaling pathways, physiological roles, and complex pathogenic processes (including, for example, teratogenesis, neurodegenerative, immuno-inflammatory, and obesity development). All of the selenoproteins contain one selenocysteine (Sec) residue, with only one notable exception, the selenoprotein P (SELENOP), which has 10 Sec residues. Although these mechanisms have been studied intensely and in detail, the characteristics and functions of many selenoproteins remain unknown. This review is dedicated to the recent data describing the identity and the functions of several selenoproteins that are less known than glutathione peroxidases (Gpxs), iodothyronine deiodinases (DIO), thioredoxin reductases (TRxRs), and methionine sulfoxide reductases (Msrs) and which are named after alphabetical letters (i.e., F, H, I, K, M, N, O, P, R, S, T, V, W). These "alphabet" selenoproteins are involved in a wide range of physiological and pathogenetic processes such as antioxidant defense, anti-inflammation, anti-apoptosis, regulation of immune response, regulation of oxidative stress, endoplasmic reticulum (ER) stress, immune and inflammatory response, and toxin antagonism. In selenium deficiency, the "alphabet" selenoproteins are affected hierarchically, both with respect to the particular selenoprotein and the tissue of expression, as the brain or endocrine glands are hardly affected by Se deficiency due to their equipment with LRP2 or LRP8.
... Furthermore, selenoprotein dysregulation has been linked to the etiology of many cancers, with corresponding glutathione peroxidase (GPX) and thioredoxin reductases (TRXR) having important roles regulating ROS levels and mitochondrial physiology [16,19,61,70]. Selenoproteins contain the 21st amino acid selenocysteine, which is incorporated using a form of specialized translation termed 'stop-codon recoding' [71][72][73]. There is no dedicated codon for selenocysteine; thus, an internal stop codon is recoded with the help of an epitranscriptomic mark written on tRNA sec by ALKBH8 to promote the synthesis of selenoproteins ( Figure 3D). ...
Article
Many cancers hijack translation to increase the synthesis of tumor-driving proteins, the messenger mRNAs of which have specific codon usage patterns. Termed 'codon-biased translation' and originally identified in stress response regulation, this mechanism is supported by diverse studies demonstrating how the 50 RNA modifications of the epitranscriptome, specific tRNAs, and codon-biased mRNAs are used by oncogenic programs to promote proliferation and chemoresistance. The epitranscriptome writers METTL1-WDR4, Elongator complex protein (ELP)1-6, CTU1-2, and ALKBH8-TRM112 illustrate the principal mechanism of codon-biased translation, with gene amplifications, increased RNA modifications, and enhanced tRNA stability promoting cancer proliferation. Furthermore, systems-level analyses of 34 tRNA writers and 493 tRNA genes highlight the theme of tRNA epitranscriptome dysregulation in many cancers and identify candidate tRNA writers, tRNA modifications, and tRNA molecules as drivers of pathological codon-biased translation.
... The internal UGA stop codon in mRNA is used to encode Sec when tRNA Sec is modified at uridine wobble base position 34. The presence of methoxycarbonylmethyluridine (mcm 5 U) and 5-methoxycarbonylmethyl-2′-O-methyluridine (mcm 5 Um) modifications improves the anticodon-codon interactions required to decode selenocysteine-containing proteins [196]. Similarly, the modification of C to m 5 C in S. cerevisiae by Trm4 at the wobble position of tRNA Leu increases when exposed to H2O2, which consequently stimulates the translation of mRNAs (MoTTs) derived from 38 genes in yeast in which 90% or more of the leucines are encoded by UUG [92]. ...
Article
Full-text available
The evolutional development of the RNA translation process that leads to protein synthesis based on naturally occurring amino acids has its continuation via synthetic biology, the so-called rational bioengineering. Genetic code expansion (GCE) explores beyond the natural translational processes to further enhance the structural properties and augment the functionality of a wide range of proteins. Prokaryotic and eukaryotic ribosomal machinery have been proven to accept engineered tRNAs from orthogonal organisms to efficiently incorporate noncanonical amino acids (ncAAs) with rationally designed side chains. These side chains can be reactive or functional groups, which can be extensively utilized in biochemical, biophysical, and cellular studies. Genetic code extension offers the contingency of introducing more than one ncAA into protein through frameshift suppression, multi-site-specific incorporation of ncAAs, thereby increasing the vast number of possible applications. However, different mediating factors reduce the yield and efficiency of ncAA incorporation into synthetic proteins. In this review, we comment on the recent advancements in genetic code expansion to signify the relevance of systems biology in improving ncAA incorporation efficiency. We discuss the emerging impact of tRNA modifications and metabolism in protein design. We also provide examples of the latest successful accomplishments in synthetic protein therapeutics and show how codon expansion has been employed in various scientific and biotechnological applications.
Article
Full-text available
Numerous natural compounds are considered as potential therapeutic agents against alcohol-induced liver disease (ALD). Research shows that selenium (Se) has a variety of bioactivities, including liver protecting ability. The present study based on in vitro cell culture models and in vivo mouse models was aimed at examining the contribution of selenomethionine (SeMet)-dominated Se-enriched peanut protein (SePP) to liver protection. SeMet and especially SePP reversed cell viability and cell death, inhibited ethanol induced CYP2E1 activation, decreased reactive oxygen species level, and restored GSH level. Hence, SeMet-dominated SePP alleviates alcohol-induced AML-12 cytotoxicity by suppressing oxidative stress. The p38-dependent mechanism was found to be responsible for SePP-induced Nrf-2 activation. Furthermore, supplementation with SePP and SeMet regulated lipid metabolism and reduced oxidative stress, minimizing liver damage in mice. Selenomethionine-dominated SePP possesses potential therapeutic properties and can be used to treat ALD through the suppression of oxidative stress.
Article
Full-text available
The essential micronutrient Selenium (Se) is co-translationally incorporated as selenocysteine into proteins. Selenoproteins contain one or more selenocysteines and are vital for optimum immunity. Interestingly, many pathogenic bacteria utilize Se for various biological processes suggesting that Se may play a role in bacterial pathogenesis. A previous study had speculated that Francisella tularensis, a facultative intracellular bacterium and the causative agent of tularemia, sequesters Se by upregulating Se-metabolism genes in type II alveolar epithelial cells. Therefore, we investigated the contribution of host vs. pathogen-associated selenoproteins in bacterial disease using F. tularensis as a model organism. We found that F. tularensis was devoid of any Se utilization traits, neither incorporated elemental Se, nor exhibited Se-dependent growth. However, 100% of Se-deficient mice (0.01 ppm Se), which express low levels of selenoproteins, succumbed to F. tularensis-live vaccine strain pulmonary challenge, whereas 50% of mice on Se-supplemented (0.4 ppm Se) and 25% of mice on Se-adequate (0.1 ppm Se) diet succumbed to infection. Median survival time for Se-deficient mice was 8 days post-infection while Se-supplemented and -adequate mice was 11.5 and >14 days post-infection, respectively. Se-deficient macrophages permitted significantly higher intracellular bacterial replication than Se-supplemented macrophages ex vivo, corroborating in vivo observations. Since Francisella replicates in alveolar macrophages during the acute phase of pneumonic infection, we hypothesized that macrophage-specific host selenoproteins may restrict replication and systemic spread of bacteria. F. tularensis infection led to an increased expression of several macrophage selenoproteins, suggesting their key role in limiting bacterial replication. Upon challenge with F. tularensis, mice lacking selenoproteins in macrophages (TrspM) displayed lower survival and increased bacterial burden in the lung and systemic tissues in comparison to WT littermate controls. Furthermore, macrophages from TrspM mice were unable to restrict bacterial replication ex vivo in comparison to macrophages from littermate controls. We herein describe a novel function of host macrophage-specific selenoproteins in restriction of intracellular bacterial replication. These data suggest that host selenoproteins may be considered as novel targets for modulating immune response to control a bacterial infection.
Article
Full-text available
We have previously shown that co-expression of c-myc and transforming growth factor (TGF)-alpha as transgenes in mouse liver results in major enhancement of neoplastic development in this organ as compared with expression of either of these transgenes alone. In this report we describe in detail the progression from liver cell dysplasia to hepatocellular carcinomas (HCCs) occurring in the liver of c-myc/TGF-alpha and c-myc transgenic mice. Despite morphological similarities in the sequence of events between the two transgenic lines, the dramatic acceleration, extent, and severity of hepatic lesions in c-myc/TGF-alpha mice clearly demonstrated the synergistic effects of this transgenic combination. Although c-myc/TGF-alpha and c-myc females displayed longer latency and lower tumor incidence, the pathological changes were the same as those seen in the male mice, including the formation of HCCs, which are absent in TGF-alpha single-transgenic females. Tumors in single- and double-transgenic mice showed induction of the endogenous c-myc and TGF-alpha and, most frequently, unchanged or decreased epidermal growth factor receptor, further indicating the collaborative role of c-myc and TGF-alpha in providing a selective growth advantage to tumor cells independently of the epidermal growth factor receptor levels. To identify possible tumor precursors, we focused particularly on the dysplastic changes preceding and accompanying the appearance of preneoplastic and neoplastic lesions in the double-transgenic mice. Early on, these changes were characterized by the appearance of large dysplastic hepatocytes, mostly pericentrally, expressing high levels of TGF-alpha and uPA, as well as TGF-beta 1, particularly in apoptotic cells. After a short period of replication and expansion into the liver parenchyma, as well as penetration into the central veins, these cells underwent apoptotic cell death while preneoplastic and neoplastic lesions were forming. The peritumorous tissues also contained small dysplastic hepatocytes and oval-like cells, similar to those found in the tumors. Transplantation of the transgenic liver tissues harboring only dysplasia with or without vascular lesions onto nude mice was able to yield HCCs composed of small diploid cells, suggesting that initiated cells are generated during the early dysplastic phase and can progress to HCC. It is therefore likely that large dysplastic hepatocytes undergo apoptosis, which may be closely associated with the up-regulation of TGF-beta 1 and uPA, whereas other cells evolve into the precursor population for HCC. Due to the simultaneous presence of c-myc, TGF-alpha, and dysplasia in premalignant human liver diseases, our transgenic mouse system appears to be an appropriate model for studying human hepatocarcinogenesis.
Article
Full-text available
We have previously shown that chronic activation of mitogenic signaling induced by over-expression of c-myc and transforming growth factor-alpha (TGFalpha) transgenes in mouse liver induces a state of oxidative stress. We therefore proposed that increased reactive oxygen species (ROS) generation might be responsible for the extensive chromosomal damage and acceleration of hepatocarcinogenesis characteristic for TGFalpha/c-myc mice. In this study, we show that vitamin E (VE), a potent free radical scavenging antioxidant, is able to protect liver tissue against oxidative stress and suppress tumorigenic potential of c-myc oncogene. Dietary supplementation with VE, starting from weaning, decreased ROS generation coincident with a marked inhibition of hepatocyte proliferation while increasing the chromosomal as well as mtDNA stability in the liver. Similarly, dietary VE reduced liver dysplasia and increased viability of hepatocytes. At 6 mo of age, VE treatment decreased the incidence of adenomas by 65% and prevented malignant conversion. These results indicate that ROS generated by over-expression of c-myc and TGFalpha in the liver are the primary carcinogenic agents in this animal model. Furthermore, the data demonstrate that dietary supplementation of VE can effectively inhibit liver cancer development.
Book
As discussed in this book, a large body of evidence indicates that selenium is a cancer chemopreventive agent. Further evidence points to a role of this element in reducing viral expression, in preventing heart disease, and other cardiovascular and muscle disorders, and in delaying the progression of AIDS in HIV infected patients. Selenium may also have a role in mammalian development, in male fertility, in immune function and in slowing the aging process. The mechanism by which selenium exerts its beneficial effects on health may be through selenium-containing proteins. Selenium is incorporated into protein as the amino acid selenocysteine. Selenocysteine utilizes a specific tRNA, a specific elongation factor, a specific set of signals, and the codeword, UGA, for its cotranslational insertion into protein. It is indeed the 21st naturally occurring amino acid to be incorporated into protein and marks the first and only expansion of the genetic code since the code was deciphered in the mid 1960s.
Chapter
Glutathione peroxidase (glutathione:H202 oxidoreductase E.C. 1.11.1.9) was discovered by Mills (1) in 1957 in his search for the factors that function in the protection of erythrocytes against oxidative hemolysis. Similarly, John Rotruck, working in Professor Hoekstra’s laboratory, focused on GPX in his search for an enzymatic function that made selenium (Se) an antioxidant (2). The unraveling of the Se/GPX story has fascinated a generation of scientists, and it has led to the use of GPX activity as one of the best indicators of Se status as well as to its use as the index of choice for the determination of Se requirements. The form of Se in GPX and the mechanism of Se insertion into the enzyme have become, together, an academic subject matter of their own; the study of this enzyme has now moved into the exciting world of molecular biology, and these investigations are helping us learn much more about the regulation of GPX and Se. This research suggests that the important function of classical GPX in some tissues is to regulate Se metabolism and Se flux to selenoproteins.
Article
Objective. —To determine whether a nutritional supplement of selenium will decrease the incidence of cancer.Design. —A multicenter, double-blind, randomized, placebo-controlled cancer prevention trial.Setting. —Seven dermatology clinics in the eastern United States.Patients. —A total of 1312 patients (mean age, 63 years; range, 18-80 years) with a history of basal cell or squamous cell carcinomas of the skin were randomized from 1983 through 1991. Patients were treated for a mean (SD) of 4.5 (2.8) years and had a total follow-up of 6.4 (2.0) years.Interventions. —Oral administration of 200 μg of selenium per day or placebo.Main Outcome Measures. —The primary end points for the trial were the incidences of basal and squamous cell carcinomas of the skin. The secondary end points, established in 1990, were all-cause mortality and total cancer mortality, total cancer incidence, and the incidences of lung, prostate, and colorectal cancers.Results. —After a total follow-up of 8271 person-years, selenium treatment did not significantly affect the incidence of basal cell or squamous cell skin cancer. There were 377 new cases of basal cell skin cancer among patients in the selenium group and 350 cases among the control group (relative risk [RR], 1.10; 95% confidence interval [CI], 0.95-1.28), and 218 new squamous cell skin cancers in the selenium group and 190 cases among the controls (RR, 1.14; 95% CI, 0.93-1.39). Analysis of secondary end points revealed that, compared with controls, patients treated with selenium had a nonsignificant reduction in all-cause mortality (108 deaths in the selenium group and 129 deaths in the control group [RR, 0.83; 95% CI, 0.63-1.08]) and significant reductions in total cancer mortality (29 deaths in the selenium treatment group and 57 deaths in controls [RR, 0.50; 95% CI, 0.31-0.80]), total cancer incidence (77 cancers in the selenium group and 119 in controls [RR, 0.63; 95% CI, 0.47-0.85]), and incidences of lung, colorectal, and prostate cancers. Primarily because of the apparent reductions in total cancer mortality and total cancer incidence in the selenium group, the blinded phase of the trial was stopped early. No cases of selenium toxicity occurred.Conclusions. —Selenium treatment did not protect against development of basal or squamous cell carcinomas of the skin. However, results from secondary end-point analyses support the hypothesis that supplemental selenium may reduce the incidence of, and mortality from, carcinomas of several sites. These effects of selenium require confirmation in an independent trial of appropriate design before new public health recommendations regarding selenium supplementation can be made.
Article
The role of the micronutrient, selenium, in human cancers associated with chronic inflammations and persistent infections is poorly understood. Peritoneal plasmacytomas (PCTs) in strain BALB/c (C), the premier experimental model of inflammation-dependent plasma cell transformation in mice, may afford an opportunity to gain additional insights into the significance of selenium in neoplastic development. Here, we report that selenium-depleted C mice (n = 32) maintained on a torula-based low-selenium diet (5–8 μg of selenium/kg) were totally refractory to pristane induction of PCT. In contrast, 11 of 26 (42.3%) control mice maintained on a selenium adequate torula diet (300 μg of selenium/kg) and 15 of 40 (37.5%) control mice fed standard Purina chow (440 μg of selenium/kg) developed PCT by 275 days postpristane. Abrogation of PCT was caused in part by the striking inhibition of the formation of the inflammatory tissue in which PCT develop (pristane granuloma). This was associated with the reduced responsiveness of selenium-deficient inflammatory cells (monocytes and neutrophils) to chemoattractants, such as thioredoxin and chemokines. Selenium-deficient C mice exhibited little evidence of disturbed redox homeostasis and increased mutant frequency of a transgenic lacZ reporter gene in vivo. These findings implicate selenium, via the selenoproteins, in the promotion of inflammation-induced PCT and suggest that small drug inhibitors of selenoproteins might be useful for preventing human cancers linked with chronic inflammations and persistent infections.
Article
Isolated hepatocytes from selenium-deficient, vitamin E-deficient, and control rats were treated with cumene hydroperoxide (CuOOH), phorone (diisopropylene acetone), acetaminophen, and diquat. The effect of these chemicals on cell viability, glutathione synthesis and release, and lipid peroxidation as measured by thiobarbituric acid (TBA)-reactive substances was determined during a 4-hr incubation in a complete medium under 95% O2:5% CO2 at 37°C. CuOOH-treated (0.5 mm) selenium-deficient and vitamin E-deficient hepatocytes lost viability sooner than control hepatocytes. Thus, loss of selenium or vitamin E from the hepatocyte resulted in a cell more susceptible to damage by CuOOH. Phorone treatment (1.65 mm) resulted in depletion of intracellular glutathione in all three groups to approximately 20% of that in untreated hepatocytes. Cell viability and TBA-reactive substances were the same in treated and untreated hepatocytes. Thus, lowering of intracellular glutathione did not result in the spontaneous loss of cell viability or increased lipid peroxidation in selenium-deficient or in vitamin E-deficient hepatocytes. Acetaminophen appeared to be less toxic to selenium-deficient hepatocytes than to controls. This finding is in agreement with whole animal studies reported previously showing that selenium deficiency protects rats against acetaminophen hepatotoxicity. A potential explanation of this result is stimulation of glutathione synthesis by selenium deficiency. Severely vitamin E-deficient hepatocytes were protected from cell death by 12.5 and 25.0 mm acetaminophe, apparently by its antioxidant properties, while 50.0 mm acetaminophen was toxic to them. At all concentrations used, acetaminophen decreased the TBA-reactive substances present in the hepatocyte suspensions. Diquat (0.1 mm) caused more rapid cell death and higher levels of TBA-reactive substances in selenium-deficient hepatocytes than in control hepatocytes. Diquat toxicity in selenium-deficient isolated hepatocytes was not as severe as its toxicity in selenium-deficient whole animals, however.
Article
Selenium deficiency causes a number of hepatic metabolic alterations in the rat which could lead to changes in chemical toxicity. It causes a decrease in glutathione peroxidase activity, an increase in glutathione S-transferase activity, and an increase in the rate of glutathione synthesis. The hepatotoxicities of three compounds which bind to glutathione S-transferase; iodipamide, acetaminophen, and aflatoxin B1, are decreased by selenium deficiency. The toxicity of redox cycling compounds is generally increased by selenium deficiency and is accompanied by evidence of lipid peroxidation. Thus, nitrofurantoin (100 mg/kg) causes renal tubular necrosis in selenium-deficient rats but not in controls. Selenium-deficient rats are much more sensitive to diquat toxicity than are controls. Lethality of diquat in selenium-deficient rats appears to be causally linked to lipid peroxidation. Lethality of diquat in control rats is not linked to lipid peroxidation. The effect of selenium does not appear to be mediated by glutathione peroxidase, however, indicating that selenium has another oxidant defense function. Another interesting observation made was that increases in inspired O2 tension decreased ethane production (lipid peroxidation) in selenium-deficient and in control rats given diquat. Thus, O2 appears to prevent diquat-induced lipid peroxidation.