ArticlePDF Available

Abstract and Figures

Overnutrition and genetic predisposition are major risk factors for various metabolic disorders. Stearoyl-CoA desaturase-1 (SCD1) plays a key role in these conditions by synthesizing unsaturated fatty acids (FAs), thereby promoting fat storage and alleviating lipotoxicity. Expression of SCD1 is influenced by various saturated and cis-unsaturated FAs, but the possible role of dietary trans FAs (TFAs) and SCD1 promoter polymorphisms in its regulations has not been addressed. Therefore, we aimed to investigate the impact of the two main TFAs, vaccenate and elaidate, and four common promoter polymorphisms (rs1054411, rs670213, rs2275657, rs2275656) on SCD1 expression in HEK293T and HepG2 cell cultures using luciferase reporter assay, qPCR and immunoblotting. We found that SCD1 protein and mRNA levels as well as SCD1 promoter activity are markedly elevated by elaidate, but not altered by vaccenate. The promoter polymorphisms did not affect the basal transcriptional activity of SCD1. However, the minor allele of rs1054411 increased SCD1 expression in the presence of various FAs. Moreover, this variant was predicted in silico and verified in vitro to reduce the binding of ETS1 transcription factor to SCD1 promoter. Although we could not confirm an association with type 2 diabetes mellitus, the FA-dependent and ETS1-mediated effect of rs1054411 polymorphism deserves further investigation as it may modulate the development of lipid metabolism-related conditions.
This content is subject to copyright. Terms and conditions apply.
1
Vol.:(0123456789)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports






*







*




SCD1



SCD1

SCD1SCD1

SCD1



Fatty acids (FAs) are the main building blocks of the structurally very heterogeneous lipid compounds. e
diverse functions of these lipids include membrane formation, energy storage and signal transduction1. Excessive
FA overload leads to abnormal lipid accumulation, cell dysfunction, or even cell death in both adipose and non-
adipose tissues24, a phenomenon known as lipotoxicity. Several studies in various experimental settings suggest
that the saturated (SFAs) and the unsaturated FAs (UFAs) contribute dierently to lipotoxicity. e deleterious
eects of palmitate, which can be mitigated in the presence of oleate, have already been widely demonstrated58.
In contrast, very little is known about the cellular eects of dietary trans fatty acids (TFAs), despite their implica-
tion in type 2 diabetes mellitus (T2DM) and cardiovascular diseases9, systemic inammation10, dyslipidemia11,
endothelial dysfunction12, dierent types of cancer13 and neurodegenerative disorders14. TFAs are UFAs that
contain at least one double bond in trans conguration. e vast majority of TFAs are produced in industrial
processes (industrial or iTFAs). Elaidate is the primary iTFA, oen found in partially hydrogenated vegetable
oils. Ruminants’ milk and meat also contain small amounts of naturally occurring TFAs (ruminant or rTFAs),
mainly the trans isomer of vaccenate9.
Human de novo FA synthesis yields palmitate, which can be extended by two-carbon units to longer saturated
chains, thus, the balanced production of saturated (SFAs) and mono- (MUFAs) or endogenous polyunsaturated
FAs (PUFAs) is reliant on desaturation. Although cis double bonds can be formed at dierent positions up to 9,
the rst one must be created at Δ9 position by stearoyl-CoA desaturase-1 (SCD1), which makes the activity of
SCD1 crucial for the overall desaturation process15.
e desaturase activity currently available to the cell is dependent on the level of the SCD1 enzyme, which,
in turn, is determined by (i) the transcriptional and post-transcriptional regulation of the synthesis and (ii)

1Department of Molecular Biology, Semmelweis University, 1085 Budapest, Hungary. 2These authors contributed
equally: Kinga Tibori and Veronika Zámbó. *email: zambo.veronika@med.semmelweis-univ.hu; kereszturi.eva@
semmelweis.hu
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
Vol:.(1234567890)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
the regulation of the protein degradation. e gene contains an alternative polyadenylation site, which results
in two dierent 3’ untranslated regions (3’ UTRs) and consequently dierent mRNA transcripts with distinct
stability, which may allow a rapid and ecient regulation of protein levels16. SCD1 transcription is also con-
trolled by several activating (insulin, growth factors, glucose, sucrose and cholesterol) and inhibitory (leptin,
glucagon, docosahexaenoic acid and arachidonic acid) agents acting through various transcription factors (TFs)
(LXR, SREBP, PPAR, C/EBP, TR)17. It is evident that intracellular FA supply and composition also modulates
SCD1 expression as the transcription of SCD1 gene is eciently induced by saturated FAs such as palmitate or
stearate, whereas it is repressed by cis-MUFAs (e.g., oleate)18. In addition, a highly conserved PUFA-sensitive
region (PUFARE) has also been identied in the upstream regulatory region of SCD1, which signicantly down-
regulates the intracellular SCD1 mRNA pool in the presence of linoleate1820. SCD1 is an enzyme with a short
half-life and its intracellular abundance is ne-tuned in the short term by the rapid degradation of the protein.
Its N-terminus contains a PEST degradation domain that is presumably involved in targeting the protein to
the proteasome during the ERAD process21. e stability of SCD1 protein in lung cancer cells is increased by
tyrosine phosphorylation22, and a UFA-induced degradation of stearoyl-CoA desaturase has been reported in
Drosophila, although the latter phenomenon has not been demonstrated in the case of the human orthologue23.
Natural genetic variations in SCD1 may also alter the above-described molecular mechanisms underly-
ing the control of SCD1 expression. A common missense single nucleotide polymorphism (SNP) (rs2234970,
M224L) was found to increase protein and mRNA stability, which could be further enhanced by dierent FAs,
and resulted in elevated intracellular UFA levels24. Furthermore, the GG haplotype of two intronic SCD1 vari-
ants (rs55710213 and rs56334587) signicantly reduced SCD1 expression by disrupting HNF4A TF binding25.
Conversely, a SNP (rs41290540) located in the 3’ UTR increased SCD1 expression in a luciferase reporter assay
by truncating a miR-498 target sequence26.
Changes in the intracellular level of SCD1 may represent risk factors for the development of various diseases.
However, despite their obvious health impact, the possible modulatory eects of either TFAs or natural human
polymorphisms in the SCD1 promoter have not been investigated. In the present study, we aimed to investigate
the eect of dierent saturated, cis- and trans-unsaturated fatty acids on the expression of SCD1 invitro at
mRNA and protein level. In addition, we planned to address the potential role of FAs, in particular elaidate and
vaccenate, and that of selected polymorphisms in the 5’ region of SCD1 in modulating promoter activity, both
separately and in combination, in a luciferase reporter system. We also aimed to analyze the potential impact
of functional promoter variants in silico and invitro on TF binding site modication and their correlation with
T2DM in an association study.


e modulating eect of the dietary SFAs, cis-MUFAs and PUFAs on the expression of the main desaturase
enzyme, SCD1 has been well characterized1820, however the possible regulatory impact of the two major dietary
TFAs, i.e., elaidate (18:1 trans-Δ9) of industrial origin and the naturally occurring vaccenate (18:1 trans-Δ11)
remains to be elucidated. To compare the eects of TFAs with those of other FAs on the cellular level of the
SCD1 protein, HEK293T and HepG2 cells were treated with BSA-conjugated oleate, palmitate, stearate, linoleate,
vaccenate and elaidate at a nal concentration of 100µM for 24h and their SCD1 content was assessed by
immunoblotting (Fig.1A,C) then evaluated by densitometry (Fig.1B,D). Consistent with the literature, mono-
unsaturated oleate and polyunsaturated linoleate treatment resulted in signicantly lower intracellular protein
levels in HEK293T cells (Fig.1A). Oleate reduced the amount of the desaturase enzyme to less than a h of the
control level in untreated cells, and SCD1 level was barely detectable in the cells treated with linoleate (Fig.1B).
Consistent with previous studies, a slight increase in SCD1 protein levels was observed in response to saturated
palmitate and stearate, although the change was only statistically signicant in the latter (Fig.1B). Most impor-
tantly, both TFAs tested (elaidate and vaccenate) markedly aected SCD1 protein amounts in HEK293T cells,
but in opposite directions. Since SCD1 levels were approximately halved by vaccinate and almost doubled by
elaidate, the opposite impact of the two TFAs resulted in a signicant dierence of more than fourfold between
the SCD1 protein content detected in cells treated with the two TFAs (Fig.1A,B).
e impact of the same FAs on SCD1 protein levels was also tested in HepG2 cells, which are of hepatocyte
origin and thus more relevant for lipid metabolism. Although intracellular SCD1 content was not signicantly
altered upon administration of oleate or SFAs, the repression by linoleate, the induction by elaidate and the signif-
icant dierence between the two TFAs could also be demonstrated in HepG2 cells at the protein level (Fig.1C,D).
SCD1
e observed impact of the FAs tested on intracellular SCD1 protein levels may be attributed to changes in tran-
scriptional activity, mRNA stability or protein stability. To learn more about the underlying mechanism, SCD1
expression was also studied at mRNA levels in HEK293T and HepG2 cells. Aer FA treatment, the gDNA-free
total RNA extracts of the cells were reverse transcribed into cDNA and mRNA expression was assessed by qPCR
as described in Materials and Methods section. Consistent with the changes in protein expression, the SCD1
mRNA content showed a very similar pattern aer administration of dietary FAs (Fig.2A,B). In the HEK293T
cell line, oleate, linoleate and vaccenate signicantly decreased the expression of SCD1 mRNA, whereas palmitate,
stearate and elaidate did not cause any measurable change. Again, the two TFAs resulted in remarkably dier-
ent expression levels, i.e., the amount of SCD1 mRNA was approximately twice as high in elaidate-treated cells
than in vaccenate-treated ones (Fig.2A). e expression pattern of SCD1 mRNA in HepG2 cells also faithfully
reected the protein levels detected by immunoblotting, so both the down- and up-regulating eects of linoleate
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
Vol.:(0123456789)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
AC
B
D
SCD1
Actin
SCD1
Actin
40
40
55
kDa
40
40
55
kDa
OPSLV
100 µM for 24 hours
Ctrl EOPSLV
100 µM for 24 hours
Ctrl E
HEK293T HepG2
Relative density (SCD1 / Actin)
Relative density (SCD1 / Actin)
0
50
100
150
200
Ctrl OPSLVE
**
***
0
50
100
150
200
250
Ctrl OPSLVE
*
***
*
**
###
###
Figure1. Eect of various dietary FAs on the expression of SCD1 in HEK293T and HepG2 cells. Cells were
treated with BSA-conjugated oleate (O), palmitate (P), stearate (S), linoleate (L), vaccenate (V) or elaidate (E) at
a nal concentration of 100μM for 24h. Immunoblot analysis of cell lysates (20µg protein per lane) was carried
out using anti-SCD1 and anti-Actin antibodies. Representative results of four (HEK293T) or ve (HepG2)
independent experiments are shown (A, C). Uncropped versions of all parallel blot images are available in
the Supplementary Information le. e band intensities were determined by densitometry and SCD1/Actin
ratios are shown as bar graphs (B, D). Statistical analysis was performed with the Tukey–Kramer Multiple
Comparisons Test. Data are shown as mean values ± SD. Ctrl: control; *p < 0.05; **p < 0.01; *** and ###p < 0.001.
0
50
100
150
Ctrl OPSLVE
0
50
100
150
200
Ctrl OPSL
VE
AB
HEK293
TH
epG2
Relative mRNA expression
(SCD1 / GAPDH)
Relative mRNA expression
(SCD1 / GAPDH)
100 µM for 24 hours100 µM for 24 hours
******
***
###***
***
###
Figure2. SCD1 mRNA expression in FA-treated HEK293T and HepG2 cells. e levels of endogenous SCD1
mRNA were measured in HEK293T (A) and HepG2 (B) cells treated with dierent FAs. FA treatment and
sample preparation were performed as described in Materials and Methods. qPCR was carried out using SCD1
and GAPDH sequence specic primers as indicated in Materials and Methods. e diagram presented depicts
the results of six independent measurements. Statistical analysis was performed with the Tukey–Kramer
Multiple Comparisons Test. Data are shown as mean values ± S.D. *** or ###p < 0.001.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
Vol:.(1234567890)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
and elaidate, respectively, were seen on SCD1 mRNA quantities. e mRNA levels in the elaidate-treated samples
exceeded those in the vaccenate-treated ones to a similar extent as observed in HEK293T cells (Fig.2B).
SCD1
It became evident that, similarly to oleate, linoleate, palmitate and stearate1820, TFAs also remarkably aect the
expression of SCD1. We wanted to investigate whether this eect is based on transcriptional regulation and/or
RNA stabilization. To this end, the 1094 base pair long section of the 5’ regulatory region of SCD1 (Fig.4A) was
cloned into pGL3-Basic vector and used in a luciferase reporter system to assess the SCD1 promoter-dependent
AB
HEK293
TH
epG2
Relative luciferase activity
Relative luciferase activity
0
50
100
150
200
250
pGL3BCtrl OPSLVE
0
50
100
150
200
pGL3B Ctrl OPSL
VE
SCD1 promoter
pGL3B
OPSLVE
Ctrl
100 µM for 24 hours
SCD1 promoter
pGL3B
OPSL
VE
Ctrl
100 µM for 24 hours
***
***
*
**
****
*
***
###
##
Figure3. Eect of dietary FAs on SCD1 promoter activity. Transient transfection and FA treatment of
HEK293T (A) and HepG2 (B) cells were performed as described in Materials and Methods. pCMV-β-gal vector
served as transfection control. Luciferase and β-galactosidase enzyme activities were measured as indicated
in Materials and Methods and their relative ratios are shown as bar graphs. e diagram depicts the results of
three (HEK293T) or six (HepG2) independent measurements normalized to pGL3-SCD1 promoter vector.
Data are shown as mean values ± S.D. Statistical analysis was performed by using the Tukey–Kramer Multiple
Comparisons Test. Ctrl: control; O: oleate; P: palmitate; S: stearate; L: linoleate; V: vaccenate; E: elaidate;
*p < 0.05; **p < 0.01; *** and ###p < 0.001.
C
B
A
1094 bp
5’ 3’‒1089 +5
Translation
SCD1 promoter
rs2275656 G>C
rs2275657 G>C
rs670213 C>T rs1054411 C>G
0
50
100
150
0
50
100
150
Relative luciferase activity
HEK293
TH
epG2
Relative luciferase activity
Figure4. Position (A) and eect of SCD1 promoter SNPs on relative luciferase activity in HEK293T (B)
and HepG2 (C) cells. e subcloned region of SCD1 promoter, the translational start site and the location,
allelic options and ID number of the four selected polymorphisms are presented. Transfection was performed
as described in Materials and Methods. pCMV-β-gal vector served as transfection control. Luciferase and
β-galactosidase enzyme activities were measured as indicated in Materials and Methods and their relative ratios
are shown as bar graphs. e diagram depicts the results of three independent measurements normalized to the
wild type SCD1 promoter. Data are shown as mean values ± S.D. Statistical analysis was performed by using the
Tukey–Kramer Multiple Comparisons Test.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
Vol.:(0123456789)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
transcriptional eects. HEK293T and HepG2 cells transfected with pGL3-SCD1 promoter construct were treated
with BSA-conjugated oleate, palmitate, stearate, linoleate, vaccenate or elaidate at 100µM concentration for 24h.
Aer harvesting the cells, relative luciferase and β-galactosidase activities were determined. e subcloned SCD1
5’ regulatory region worked as a potent promoter, increasing the relative luciferase activity in both cell lines by
approximately 20-fold compared to pGL3-B (Fig.3). As expected, a signicant suppression by linoleate on lucif-
erase activity was seen in both cell lines, but surprisingly, no such phenomenon was observed for oleate. e two
SFAs signicantly enhanced the relative luciferase activity in both HEK293T (Fig.3A) and HepG2 (Fig.3B) cells.
Of the two TFAs, elaidate caused an approximately 50% increase in SCD1 promoter activity, whereas vaccenate
was not eective in this assessment, thus the signicant dierence between the two TFAs was also revealed at
the transcriptional regulation of SCD1.
SCD1
Regulation of the transcription can be signicantly aected by genetic variations of the promoter region. ere-
fore, we tested the known SNPs in the SCD1 promoter with minor allele frequencies (MAFs) above 5% for their
potential impact on FA-sensitivity of SCD1 expression. Using the NCBI dbSNP and Ensembl databases, we
selected four polymorphisms that met the above criterion. Identication number, position, allelic variants and
MAF value of these variants are presented in Supplementary TableS1 and Fig.4A. All four SNPs were agged
as modiers by the VEP prediction program, indicating their potential functionality (Supplementary TableS1),
yet neither of them has been experimentally investigated. We generated both alleles of these genetic variants
in the pGL3-SCD1 promoter vector by site-directed mutagenesis and analyzed them by a luciferase reporter
assay in HEK293T and HepG2 cells aer transient transfection. is invitro approach revealed no signicant
modulation of basal promoter activity by any of the four polymorphisms, i.e., none of the minor allelic variants
altered the relative luciferase activity compared to the major variant (hereaer referred to as wild type) in either
of the two human cell lines (Fig.4B.C).
To further analyze the potential eect of these polymorphisms, the promoter activity of the allelic variants
was also tested in the presence of oleate, palmitate, stearate, linoleate, vaccenate or elaidate. HEK293T cells were
treated with 100µM BSA-conjugated FAs 5h aer transient transfection for 24h, and the relative luciferase
enzyme activities were measured in the collected samples as described in Materials and Methods. e G allele of
the rs1054411 SNP, which was shown to have the same basal promoter activity as the wild type (see Fig.4B,C),
resulted in a signicantly higher promoter activity than the wild type when exposed to any of the six FAs (Fig.5).
It is noteworthy that the most pronounced, almost threefold increase in the relative luciferase activity compared
to the wild type was observed upon elaidate treatment (Fig.5F). When the same allelic variant was tested in
HepG2 cells, the allele-specic impact of some other FAs was also observed, i.e., the relative luciferase activity
with the rs1054411_G promoter was increased not only by elaidate but also by linoleate and vaccenate in the
reporter system (Supplementary Fig.S1).
Beside the general FA-dependent enhancement of rs1054411_G promoter activity, it is also intriguing that
the rs2275656_C variant apparently lost its FA-sensitivity, as it did not respond to any FA treatments investigated
(Fig.5). e other two tested SCD1 promoter variants (rs670213_T and rs2275657_C) showed rather diverse
responsiveness to FAs. While neither of them modied the eect of the cis-unsaturated FAs (oleate and linoleate)
compared to the wild type (Fig.5A,D), rs2275657_C showed a signicantly increased promoter activity both
in the presence of SFAs (palmitate and stearate) (Fig.5B,C) and TFAs (vaccenate and elaidate) (Fig.5E,F), and
such modulation was only observed with palmitate in case of rs670213_T (Fig.5B).
SCD1
e possible impact of the four SNPs on TF binding sites in the promoter of the SCD1 gene was analyzed in silico
using the JASPAR transcription factor binding site prediction program. Specically, we addressed the question
whether the alteration of the four nucleotides aected by the polymorphisms could cause a predictable change
in the binding probability of any TFs in this region. e in silico TF selection protocol has been published
previously27. Briey, for each SNP, the two 41-nucleotide long DNA segments were compared, in which the poly-
morphic nucleotide was located at position 21. Hits were selected with a relative TF binding score greater than
80% for at least one allele and a relative score dierence of at least 15% between the two alleles. Five predicted
TF binding sites for rs1054411, two for rs670213 and rs2275657, and seven for rs2275656 met the above criteria,
their IDs and relative binding probabilities are summarized in Table1 and Supplementary TableS2. e predicted
eect of the rs1054411 polymorphism on ETS1 TF binding appeared to be the most probable hit in the screen.
ETS1 had the highest binding probability score of all hits for the wild type sequence (rs1054411_C: 98.14%),
and also the largest dierence between the two alleles, with the minor allele (rs1054411_G) 21.6% less likely to
form an ETS1 TF binding site (Table1). e predicted large dierence between the two alleles of this SNP is not
surprising, considering that the polymorphic nucleotide is located at the h, highly conserved position of the
six-nucleotide long consensus sequence of ETS1 response element (Fig.6A).
SCD1
In light of the FA sensitivity of the rs1054411_G allele variant observed in our previous experiments (see Fig.5),
and the highly diverse regulatory mechanism of ETS128, the question was raised whether the ETS1 expression
is also FA sensitive. To address this question, HEK293T cells were treated for 24h with 100µM BSA-conjugated
oleate, palmitate, stearate, linoleate, vaccenate or elaidate, and the endogenous gene expression of ETS1 was
assessed in the collected samples, but the ETS1 mRNA levels were not modied by any of the FAs tested (Sup-
plementary Fig.S2). e possible FA-dependent modication of endogenous ETS1 protein levels could not be
examined because the low sensitivity of the commercially available ETS1 antibodies did not allow for proper
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
Vol:.(1234567890)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
detection of the protein without overexpression. e amount of ETS1 protein overexpressed in transfected cells
was not aected by the presence of FAs (Supplementary Fig.S2).
Since the ETS1 binding was predicted to be aected by the rs1054411 polymorphism, the allele-specic
eect of this TF was tested in a luciferase reporter system invitro (Fig.6). HEK293T cells were transiently co-
transfected with the pGL3-SCD1 promoter plasmid containing either the wild type or the rs1054411_G variant,
together with dierent amounts of ETS1 expression vector. 24h aer transfection, the cells were harvested, and
the relative luciferase activities were measured. e samples of the co-transfected cells were compared to the
corresponding wild type or rs1054411_G promoter activities without ETS1 overexpression. ETS1 overexpres-
sion resulted in enhanced activities of both versions of the promoter, and the eect was growing in parallel
with the increasing amounts of ETS1 expression construct applied (Fig.6B) and with the increasing amount of
ETS1 protein, as veried by immunoblotting (Fig.6C). However, there was a marked dierence between the
0
100
200
300
0
100
200
300
0
100
200
300
Relative luciferase activity
Relative luciferase activity
Relative luciferase activity
0
100
200
300
0
100
200
300
0
100
200
300
400
Relative luciferase activity
Relative luciferase activity
Relative luciferase activity
OleatePalmitate Stearate
Linoleate Vaccenat
eE
laidate
*
*** ***
** ** *
*** ***
***
*
ABC
DE
F
*
Figure5. Modulating eect of promoter polymorphisms on SCD1 promoter activity in the presence of various
FAs in HEK293T cells. Transient transfection and FA treatment of HEK293T cells were performed as described
in Materials and Methods. pCMV-β-gal vector served as transfection control. Luciferase and β-galactosidase
enzyme activities were measured as indicated in Materials and Methods and their relative ratios are shown
as bar graphs. e diagram depicts the results of at least three independent measurements normalized to the
relative luciferase activity of oleate- (A), palmitate- (B), stearate- (C), linoleate- (D), vaccenate- (E) or elaidate-
treated (F) wild type SCD1 promoter containing reporter vector. Data are shown as mean values ± S.D. Statistical
analysis was performed by using the Tukey–Kramer Multiple Comparisons Test. O: oleate; P: palmitate; S:
stearate; L: linoleate; V: vaccenate; E: elaidate; *p < 0.05; **p < 0.01; *** p < 0.001.
Table 1. List of transcription factors that are aected by rs1054411 polymorphism. Positive or negative values
of the relative score dierences indicate that the minor allele increases or decreases the probability of TF
binding, respectively.
Name TF ID Strand
Relative score (%)
C allele G allele Dierence
NFATC3 MA0625.2 + 61.45 80.39 18.94
SOX18 MA1563.1 65.39 81.67 16.28
SPI1 MA0080.1 81.85 66.40 15.45
ETV5 MA0765.1 85.96 69.13 16.83
ETS1 MA0098.1 + 98.14 76.54 21.60
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
Vol.:(0123456789)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
ETS1
Actin
55
40
55
kDa
40
0
50
100
150
200
250
300
Ctrl +10ng+25 ng +50ng+100ng+200ng
Relative luciferase activity
B
C
ETS1
Wild type
rs1054411_G
***
***
***
***
!!!
!!!
!!!
##
###
#
#
A
CATCCG
CATCGG
2
1.5
1
0.5
0
Bits
nt position
Wild type
rs1054411_G
5’ ‒
5’ ‒
‒ 3’
‒ 3’
Figure6. Eect of rs1054411 SNP on the ETS1-mediated stimulation of SCD1 promoter activity in a luciferase
reporter system. (A) Structure of the ETS1 TF binding sequence modied by rs1054411_G is illustrated. e
polymorphic site is highlighted in bold and red. (B) Transient co-transfection of HEK293T cells was performed
as described in Materials and Methods. pCMV-β-gal vector served as transfection control. Luciferase and
β-galactosidase enzyme activities were measured as indicated in Materials and Methods and their relative
ratios are shown as bar graphs. e diagram depicts the results of three to twelve independent measurements
normalized to ETS1-free wild type or rs1054411_G pGL3-SCD1 promoter vector, respectively. Data are shown
as mean values ± S.D. Statistical analysis was performed by using the Tukey–Kramer Multiple Comparisons
Test. Ctrl: control; #p < 0.05; ##p < 0.01; ***, ### or !!!p < 0.001. C e increasing amount of ETS1 protein expressed
in HEK293T cells co-transfected with an increasing amount of ETS1 plasmid (10, 25, 50, 100 or 200ng) was
veried by immunoblotting. Immunoblot analysis of cell lysates (20µg protein per lane) was carried out using
anti-ETS1 and anti-Actin antibodies as described in Materials and Methods. Uncropped versions of all parallel
blot images are available in the Supplementary Information le. Ctrl: control; ” indicates non-specic band on
ETS1 immunoblot; #p < 0.05; ##p < 0.01; ***, ### or !!!p < 0.001.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
Vol:.(1234567890)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
extent of the ETS1-dependent enhancement between the two promoter versions tested. In case of the wild type
SCD1 promoter, even the small amount of ETS1 protein yielded by 25ng plasmid signicantly increased the
relative luciferase activity by almost one and a half times (Fig.6B). Furthermore, the largest amount of ETS1
plasmid applied (200ng) resulted in more than a twofold increase in the activity of the wild type SCD1 promoter.
Although the activity of the promoter holding the G allele of rs1054411 SNP showed an increasing trend with
increasing ETS1 levels, it remained below that of the wild type in all experimental conditions (Fig.6B), which
nding is in agreement with the in silico predicted reduction of the TF binding capacity (see Table1). e dif-
ference between the activity of the two alleles of rs1054411 aer co-transfection with 25, 50, 100 or 200ng ETS1
plasmid was signicant in favor of the wild type sequence (Fig.6B).
To further analyze the possible interaction between ETS1 TF and the presence of FAs, a combination of co-
transfection and FA treatment was performed. For this experiment elaidate was selected, as the rs1054411_G
allele in its presence was the most potent in enhancing SCD1 promoter activity. HEK293T cells were transiently
transfected with wild type or rs1054411_G allele-containing SCD1 promoter construct with or without 100ng
ETS1 expression vector and/or in the presence or absence of 100µM BSA-conjugated elaidate (Supplementary
Fig.S3). e appropriate amount of ETS1 vector and the optimal concentration of elaidate were chosen based
on our experiments shown in Figs.5 and 6. Samples were collected 24h aer FA treatment and relative lucif-
erase activities were measured. As expected, ETS1 inhibited and elaidate enhanced SCD1 promoter activity in
the presence of the rs1054411_G allele. When the two agents were administered together, however, the negative
eect of ETS1 could neither reverse nor neutralize the enhancing eect of elaidate on rs1054411_G allele-specic
promoter activity, but merely reduced it by about half (73% vs 37%, Supplementary Fig.S3).

e possible association between the rs1054411 polymorphism and T2DM was investigated in a case–control
setup. e results are summarized in Table2. e observed genotype distribution in the control group was in
agreement with the expected values based on the Hardy–Weinberg equilibrium (χ2-test p = 0.911). Allele frequen-
cies were in line with the European population data available in 1000Genomes (MAF: 41 vs. 40%), however, in
our control group the minor allele was slightly overrepresented compared to both ALFA (MAF: 35%) and global
frequencies (MAF: 28%). Association analyses were performed using both allele- and genotype-based approaches,
including the dominant model (i.e., genotype combination). As shown in Table2, the frequency of the G allele
was slightly but not signicantly lower in the T2DM group in all comparisons. Due to the limited number of
samples that could be included in the study, the power was as low as 35.6% suggesting that the lack of statistically
signicant result does not exclude the putative role of the SNP in the genetic risk of T2DM.

e role of SFAs and cis-unsaturated FAs in regulating the expression of SCD1, one of the key enzymes of lipid
metabolism, is a much-researched topic in the literature. SFAs, not surprisingly and in line with our results
(Figs.1, 2, 3), tend to increase the amount of SCD1 available in the cell, thus enhancing their own conversion to
unsaturated FAs, which favors their utilization in lipid synthesis and hence mitigates their own lipotoxic eects
throughout the body29,30. e attenuating eect of cis-unsaturated FAs on SCD1 expression is also well known20,31,
however, the mechanism of action of monounsaturated oleate and polyunsaturated linoleate may slightly dier.
Linoleate is thought to interfere with desaturation through the regulation of transcription, and it clearly repressed
SCD1 expression at all three levels we examined (promoter activity, mRNA and protein levels) in both cell lines.
is is in agreement with the fact that a PUFA-responsive element has been described and characterized in the
Table 2. Comparison of allele, genotype, and genotype combination frequencies of rs1054411 polymorphism
in control and T2DM groups.
Control
(N = 370) T2DM
(N = 282)
N % N %
Allele
C 437 59 351 62
G 303 41 213 38
χ2p = 0.2447
Genotype
C/C 127 34 107 38
C/G 183 49 137 49
G/G 60 16 38 13
χ2p = 0.4943
Genotype combination
C + 310 84 244 87
C60 16 38 13
χ2p = 0.3319
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
Vol.:(0123456789)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
upstream regulatory region of both human and mouse SCD1 genes1820. While several studies have demonstrated
the SCD1-repressing eect of oleate in a variety of ways32, the exact mechanism is still unknown. Although oleate
clearly decreases the desaturase the level of the enzyme and thus the desaturase activity, there are inconsistent
ndings with respect to the eect at the mRNA level, and even more so with respect to the promoter activity,
suggesting that oleate acts through mRNA and/or protein stabilization rather than reducing transcription33. It
should be noted that the well-characterized reducing eect of oleate on SCD1 was not seen at all three regulatory
levels we examined in HepG2 cells, and the eect of other FAs was also rather modest in this cell line, probably
due to the relatively high FA tolerance of HepG2 cells34.
Human studies have reported a positive correlation between TFA intake and the development of lipid metab-
olism-related conditions such as the metabolic syndrome, T2DM, cardiovascular disease and cancer9, however,
the potential role of TFAs in the regulation of FA desaturation has been so far neglected. It has been reported
that TFAs may have similar protective eects against palmitate toxicity as cis-unsaturated oleate in cell cultures6.
ey promote inammation and ER stress to a much lesser extent than the most lipotoxicity-inducing SFAs,
so they are currently considered less harmful3537. TFAs were also reported to stimulate cholesterol synthesisin
vitro38 and enhance hepatic fat accumulationin vivo39. It is therefore clear that the TFAs have special eects
on the metabolism and basic physiological processes of the human body, that are dierent from other dietary
FAs, and there is an ongoing scientic debate about the similarities and dierences between iTFAs and rTFAs in
terms of their health impacts. Several human studies have reported that the adverse eects of TFAs are limited to
iTFAs40,41, while rTFAs have been shown to be harmless or even benecial for metabolic health42,43. In contrast,
other epidemiological and clinical studies have shown that rTFAs are as culpable as iTFAs in the development
of metabolic and cardiovascular diseases44,45. In the light of the controversial data, we considered it important to
investigate and compare the eect of iTFAs and rTFAs on the expression of SCD1, as modulating the amount of
this key enzyme of lipid metabolism would provide an obvious means for the TFAs to inuence lipid homeostasis
in the human body. It was previously published that the administration of elaidate increased the desaturation
index in HASMC, HUVEC and HepG2 cells31,46, as well as the SCD1 mRNA expression in trophoblast and
HASMC cells31,47, while vaccenate did not seem to alter FA desaturation31,46,48. To rene the overall picture, we
systematically examined the eect of elaidate and vaccenate on SCD1 at the protein and mRNA levels, as well as
on the promoter activity in a luciferase reporter system in HEK293T and HepG2 cell lines. Consistent with the
limited scientic data summarized above, a signicant dierence was detected between the two types of TFAs, as
a marked inducing eect of elaidate was detected in all the three investigated parameters and in both cell lines,
which contrasted with the neutral nature of vaccenate (Figs.1, 2, 3).
Studying FA-dependent regulation of SCD1 expression is of particular interest since consequential alterations
in lipid desaturation have been implicated in a variety of diseases. Elevated cellular activity of SCD1, an enzyme
catalyzing the rate-determining step of FA desaturation, which in turn is essential for major synthetic pathways
of lipid metabolism, signicantly increases the likelihood of developing obesity and related conditions such as
the metabolic syndrome, diabetes, insulin resistance and hepatic steatosis17. SCD1 has also been identied as an
important modulator of cancer cell survival and progression49, and its expression is associated with poor progno-
sis in several cancer types50. Genetic polymorphisms may also regulate the intracellular availability of a gene or
protein in the context of gene-environment interactions, independently of, or possibly in combination with, the
eects of FAs51. As SNPs in the upstream regulatory region of SCD1 have not yet been functionally investigated,
and only the rs670213 polymorphism has been analyzed and found to be unrelated to metabolic risk52,53, in the
present study, we tested the promoter polymorphisms invitro in a luciferase reporter system both in the absence
(Fig.4) and presence (Fig.5, Supplementary Fig.S1) of various dietary FAs. e observed allele-specic inducing
properties of the FAs are not without precedent, as the elevated expression of the only common missense SCD1
variant (rs2234970) is also attributed partly to a FA-mediated and sequence-dependent protein stabilization24.
Although the rs1054411 SNP, which was found to be functional in the presence of FAs (Fig.5), did not show
signicant association with T2DM in our study (Table2), its role in the development of metabolic conditions
cannot be ruled out completely. In light of the results of our in silico analysis and invitro experiments, its possible
correlation with diabetes should be assessed in larger samples, complemented with other phenotypic and clinical
data (e.g., dietary intake composition and serum FA prole). e NCBI LDmatrix tool indicates complete linkage
disequilibrium for rs2275656 and rs2275657 SNPs, while the other two loci are not or only partially linked. In
line with this, the NCBI LDhap predicts the presence of ve haplotypes out of 16 possible combinations of the
four SNPs in the European population. is suggests that the rs670213 and rs1054411 SNPs are evolutionally
younger, their polymorphic alleles are likely to have arisen and combined with the GG haplotype of rs2275656
and rs2275657, whereas the CC haplotype of rs2275656 and rs2275657 is only found with the ancestral alleles
(both C) of rs670213 and rs1054411. Taking these together, it may be worthwhile to analyze the haplotypes of
the four SCD1 promoter SNPs from both a functional and an association perspective in the future.
ETS1, a member of the ETS protein family of TFs, regulates the expression of a diverse set of proteins through
its interaction with specic consensus sequences upstream of target genes. Increased expression of ETS1 has
been detected in a wide variety of cancers and associated particularly with tumor progression and invasion, and
there is also increasing interest in its role in basic metabolic processes54, as it has been revealed to up-regulate
key enzymes in FA metabolism55. Although the highly diversied transcriptional, post-transcriptional and post-
translational control of ETS1 has been thoroughly characterized28, the possible role of FAs in this regulation has
not been investigated. Although ETS1 expression itself was not found to be FA-sensitive in our experimental
setup (Supplementary Fig.S2), our in silico analysis identied ETS1 as a TF with allele-specic binding to the
SCD1 promoter region carrying the rs1054411 SNP (Table1). Moreover, the predicted allele-specic binding of
ETS1 was also veried invitro (Fig.6).
In summary, our results indicate that the two most common TFAs, industrial elaidate and natural vaccenate,
have signicantly dierent eects on SCD1 expression, as the induction by elaidate manifested invitro not only
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
Vol:.(1234567890)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
at the protein and mRNA levels of the endogenous expression but also at the promoter activity assessed in a
reporter gene model. Among the investigated promoter polymorphisms, the rs1054411, which did not modify
basal SCD1 expression, largely aected SCD1 promoter activity in the presence of dierent dietary FAs or under
the inuence of ETS1 TF, as measured by using a luciferase reporter assay.
Elevation of SCD1 expression in various health conditions is of utmost importance, whether as a cause or a
consequence17,32. e enzyme is a promising target for the treatment of metabolic diseases, and eorts have been
made to develop liver-targeted SCD1 inhibitors56. Since genetic variations have a major impact on the ecacy of
therapy57,58, SCD1 variants, including functional promoter polymorphisms, such as rs1054411, are likely to alter
the eectiveness or even the need for medical treatment with SCD1 inhibitors. e development of individual-
ized therapeutic protocols based on genetic proling seems a reasonable future goal in the treatment of lipid
metabolism-related diseases. However, this goal can only be achieved if the pathomechanisms are understood at
the level of gene-environment interaction, which requires both detailed functional characterization of disease-
associated gene polymorphisms and thorough mapping of environmental risk factors.
e main strength of this work lies in its diversity, as the opposing eects of the two trans-monounsaturated
FAs on SCD1 have been successfully demonstrated at multiple levels and in dierent cell lines. Furthermore, a
unique FA-dependent transcriptional modulation mechanism of the rs1054411 SNP in the SCD1 promoter has
been identied, which may be further ne-tuned in an allele-specic manner by the ETS1 proto-oncogene TF.
However, the association study performed is of limited value as it has very low statistical power due to the rather
small sample size. In addition, functional analysis of the four promoter polymorphisms in haplotypes and exten-
sion of the invitro studies to animal models could further increase the reliability of the present work in the future.


Culture medium and supplements were purchased from ermo Fisher Scientic (Waltham, MA, USA). Oleate,
palmitate, stearate, linoleate, elaidate, vaccenate, bovine serum albumin, HEK293T and HepG2 cells were pur-
chased from Sigma-Aldrich (St. Louis, MO, USA). All chemicals used in the study were of analytical grade. All
experiments and measurements were performed using Millipore ultrapure water.

Based on the NCBI and Ensembl databases, SCD1 promoter SNPs with MAF above 5% and heterozygosity
above 0.095 were selected. e JASPAR (http:// jaspar. gener eg. net/, accessed on 30 June 2022) open-access, non-
redundant TF biding prole database was used to predict the potential eect of rs1054411, rs670213, rs2275657
and rs2275656 polymorphisms on TF binding to the SCD1 promoter59. e allele-specic eect on TF binding
was analyzed as previously described27. Briey, both allelic variants of each SNP were compared pairwise. TFs
showing a score dierence of at least 15% between the two variations of the given polymorphism, and a rela-
tive score above 80% for at least one of the alleles, were retained for further analysis. e impact of the selected
sequence variants was predicted in silico using the Variant Eect Predictor (https:// www. ensem bl. org/ Homo_
sapie ns/ Tools/ VEP/, accessed on 12 January 2023)60.

A 1094 base pair fragment of the upstream regulatory region of SCD1 was amplied from human genomic DNA
template by iProof™ High-Fidelity DNA Polymerase (Bio-Rad, Hercules, CA, USA) and cloned into the pGL3-
Basic plasmid (pGL3B, Promega, Madison, WI, USA) between the Xho I and Hind III restriction endonuclease
recognition sites with 5’AAA TTT CTC GAG CAA AAC ATC CCG CAC GCA T–3’ sense and 5’AAA TTT
AAG CTT GGC ATC TTG GCT CTC GGA TG –3’ antisense primers. Bold letters indicate the recognition sites
of the two endonucleases, respectively. Aer purication and restriction endonuclease (ermo Fisher Scientic,
Waltham, MA, USA) digestion, the amplicons were ligated (T4 Ligase, ermo Fisher Scientic, Waltham, MA,
USA) into pGL3B vector (Promega, Madison, WI, USA) upstream the luciferase reporter gene. e natural
variants were generated using Q5® Site-Directed Mutagenesis Kit (New England BioLabs, Ipswich, MA, USA)
following the manufacturer’s instruction. Mutagenic primers were designed using the online NEB primer design
soware, NEBaseChanger™. Aer digestion of the original non-mutated and methylated plasmid by KLD reac-
tion, an aliquot of the constructs was transformed into XL10-Gold® Ultracompetent Cells (Agilent, Santa Clara,
CA, USA), which were then screened for positive colonies by PCR. e cloning and mutagenic primers are listed
in Supplementary TableS3. e ETS1 expression plasmid was purchased from BioCat (Heidelberg, Germany)
with pcDNA3.1() vectorial background. All constructs were veried by Sanger sequencing.

Human embryonic kidney (HEK293T) and hepatocellular carcinoma (HepG2) cells were cultured in 12-well
plates (1 × 106 cells per well) in Dulbecco’s modied Eagle medium (DMEM) supplemented with 10% fetal bovine
serum and 1% penicillin/streptomycin solution at 37°C in a humidied atmosphere containing 5% CO2. Cells
were transfected with 0.5μg pGL3B-SCD1 promoter constructs using 3 µL Lipofectamine 3000 that was sup-
plemented with 2 µL P3000 (Invitrogen, Carlsbad, CA, USA) in 1mL DMEM. As a transfection control, 0.5µg
pCMV-β-gal plasmid was co-transfected. Cells were harvested and processed 24–30h aer transfection.

Oleate, palmitate, stearate, linoleate, elaidate, and vaccenate were diluted in ethanol (Molar Chemicals, Halász-
telek, Hungary) to a nal concentration of 50mM and conjugated with 20% FA-free BSA in 1:4 ratio at 50°C
for 1h. e working solution for FA treatments was prepared freshly in FBS-free and antibiotic-free medium at
Content courtesy of Springer Nature, terms of use apply. Rights reserved
11
Vol.:(0123456789)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
100µM nal concentration. e FA treatment was carried out for 24h in 12-well plates. For luciferase assay, the
culture medium was replaced 5h aer transfection and the cells were incubated for a further 24h.

Cell lysates were prepared for immunoblot analysis by removing the medium and washing the cells twice with
PBS. 100 µL RIPA lysis buer (0.1% SDS, 5mM EDTA, 150mM NaCl, 50mM Tris, 1% Tween 20, 1mM Na3VO4,
1mM PMSF, 10mM benzamidine, 20mM NaF, 1mM pNPP, and protease inhibitor cocktail) was added to each
well and the cells were scraped and briey vortexed. Aer 15min incubation at room temperature, the lysates
were centrifuged for 5min at maximum speed in a benchtop centrifuge at 4°C to remove cell debris. Protein
concentration of the supernatant was measured with Pierce® BCA Protein Assay Kit (ermo Fisher Scientic,
Waltham, MA, USA) and the samples were stored at 20°C until further analysis.
For the luciferase reporter assay, cells were washed twice with PBS and then scraped in 100 µL reporter lysis
buer (Promega, Madison, WI, USA) and vortexed briey. A single freeze–thaw cycle was followed by centrifug-
ing in a benchtop centrifuge (5min, max speed, 4°C). Supernatants were used for enzyme activity determination.
For total RNA isolation, cells were washed twice with PBS and collected in 350 µL RLT buer (Qiagen, Hilden,
Germany) supplemented with 1% β-mercaptoethanol according to manufacturer’s protocol. Samples were stored
at 80°C until further analysis.

Aliquots of cell lysates (20µg protein per lane) were analyzed by SDS-PAGE on 12% Tris–glycine minigels, and
transferred onto Immobilon-P membranes (Millipore, Billerica, MA, USA). Primary and secondary antibodies
were applied overnight at 4°C and for 1h at room temperature, respectively. Horseradish peroxidase (HRP)-
conjugated goat polyclonal anti-Actin (Cell Signaling, Danvers, MA, USA, sc-1616) antibodies were used at
1:2000 dilution. SCD1 was detected with a rabbit polyclonal antibody (Cell Signaling, Danvers, MA, USA,
2438S), used at a dilution of 1:2000, followed by HRP-conjugated goat polyclonal anti-rabbit IgG (Cell Signaling,
Danvers, MA, USA, 7074S) at a dilution of 1:2000. ETS1 was detected with a goat polyclonal antibody (Bethyl
Laboratories, A190-110A), used at a dilution of 1:2000, followed by HRP conjugated mouse monoclonal anti-goat
IgG (Cell Signaling, Danvers, MA, USA, sc-2354) at a dilution of 1:2000. HRP was detected by C-DiGit® Blot
Scanner (LI-COR, Lincoln, NE, USA) using the SuperSignalWest Pico Chemiluminescent Substrate (ermo
Fisher Scientic,Waltham, MA, USA). As the edges of the membranes can blend into the background due to
digital imaging, a protein marker is run on each side of the sample sets to clearly dene them. Uncropped ver-
sions of all parallel blot images are available in the Supplementary Information le.

Luciferase activity was detected using the Luciferase Assay System kit (Promega, Madison, WI, USA) by adding
15 µL Luciferin reagent to 5 µL of al cell extracts. β-galactosidase activity of 20 µL cell lysates was measured by
determining the o-nitrophenyl-β-D-galactopyranoside (at a nal concentration of 3mM) cleavage rate. Lumi-
nescence was detected using a Varioskan multi-well plate reader (ermo Fisher Scientic, Waltham, Massa-
chusetts, USA). Values for luciferase activity were normalized to β-galactosidase activity (measured by standard
protocol using the same Varioskan plate reader in photometry mode). Each experiment was repeated three times
independently, and each sample was analyzed in triplicate.

Total RNA was puried from transfected HEK293T and HepG2 cells by using RNeasy Plus Mini Kit (Qiagen,
Germantown, MD, USA) following the manufacturer’s instruction. Concentrations were measured using Nan-
oDrop1000 spectrophotometer. To assess the integrity and purity of the isolated total mRNA samples, the ratios
of their absorbance at 260/280 and 260/220nm were determined, and they were also analyzed by agarose gel
electrophoresis to visualize bands corresponding to 28S and 18S rRNAs, respectively. Possible DNA contamina-
tion was removed by DNase I treatment using RNAqueous®-4PCR Kit (Invitrogen, Carlsbad, CA, USA). cDNA
samples were produced by reverse transcription of 0.5µg DNA-free RNA, using the SensiFAST™ cDNA Synthesis
Kit (Meridian Bioscience, Memphis, TN, USA).
qPCR
Quantitative PCR assay was performed in 20 µL nal volume containing 5 µL 20 × diluted cDNA, 1 × PowerUp™
SYBR™ Green Master Mix, and 0.5µM forward and reverse primers using QuantStudio 12K Flex Real-Time PCR
System (ermo Fisher Scientic, Waltham, Massachusetts, USA). SCD1 and ETS1 sequences were amplied by
5’ CTG GCC TAT GAC CGG AAG AAA 3’ / 5’ GAC CCC AAA CTC ATT CCA TAG G 3’ and 5’ – AGA
TGA GGT GGC CAG GAG AT 3’ / 5’ – CTG CAG GTC ACA CAC AAA GC – 3’ primer pairs, respectively.
GAPDH cDNA was also amplied as an endogenous control using 5’ GTC CAC TGG CGT CTT CAC CA
3’ / 5’ GTG GCA GTG ATG GCA TGG AC 3’ primer pair. e rst step of the thermocycle was an initial
denaturation and enzyme activation at 95°C for 2min. It was followed by 40 cycles of 95°C for 15s, 55°C for
15s, and 72°C for 1min; measurement of the uorescent signal was carried out during annealing. Reactions were
performed in triplicates, and a reaction mixture with RNase-free water instead of template cDNA was employed
as non-template control. Relative expression levels were calculated as 2ΔCT, where ΔCT values corresponded to
the dierence of the CT-values of the endogenous control and target genes.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
12
Vol:.(1234567890)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
Subjects
282 patients diagnosed with T2DM in the 2nd Department of Internal Medicine, Semmelweis University (51.2%
female, 48.8% male, disease onset at the age of 62.4 ± 12.6 y) were recruited in the study. e control group con-
sisted of 370 volunteers with no medical history of any metabolic disease (61.4% female, 38.6% male, mean age:
33.1 ± 21.6 y). e diagnosis of diabetes was made based on fasting blood sugar values, oral glucose tolerance
test (OGTT), and HbA1C value according to WHO regulations. Individuals with autoimmune, infectious, or
metabolic disorders other than type 2 diabetes were excluded from the study. Genetic analysis of the participants
was approved by the Local Ethical Committee (ETTTUKEB ad.328/KO/2005, ad.323–86/2005-1018EKU from
the Scientic and Research Ethics Committee of the Medical Research Council). e study was conducted in
accordance with the principles of the Declaration of Helsinki. Participants signed a written informed consent
before sample collection for genetic analysis. To avoid the risk of spurious association caused by population
stratication, subjects of Hungarian origin were exclusively included to ensure the comparison of homogenous
populations. Buccal epithelial cells were collected by swabs. e rst step of DNA isolation was an incubation
of the buccal samples at 56°C overnight in 0.2mg/mL Proteinase K cell lysis buer. Subsequently, proteins were
denatured using a saturated NaCl solution. DNA was then precipitated by isopropanol and 70% ethanol. DNA
pellet was resuspended in 100 µL 0.5 × TE (1 × TE: 10mM Tris pH = 8.0; 1mM EDTA) buer. Concentration of
the samples was measured by NanoDrop1000 spectrophotometer.
Genotyping
Rs1054411 promoter polymorphism of the SCD1 gene was genotyped using pre-designed TaqMan assay
(C_34192814_10, ermo Fisher Scientic, Waltham, MA, USA). qPCR assay was performed in 5 µL nal vol-
ume containing approximately 4ng genomic DNA, 1 × TaqPath™ ProAmp™ Master Mix, and 1 × TaqMan® SNP
Genotyping Assay using QuantStudio 12K Flex Real-Time PCR System (ermo Fisher Scientic, Waltham,
MA, USA). ermocycle was started by activating the hot start DNA polymerase and denaturing genomic DNA
at 95°C for 10min. is was followed by 40 cycles of denaturation at 95°C for 15s, and combined annealing
and extension at 60°C for 1min. Real-time detection was carried out during the latter step to verify the results
of the subsequent post-PCR plate reads and automatic genotype calls.
Statistical analysis
Immunoblots were evaluated by densitometry using the Image Studio® 5.2 soware (LI-COR Biotechnology,
Lincoln, NE, USA), and are shown as relative band densities normalized to Actin as a reference. Relative band
densities, luciferase activities and mRNA levels are presented in the diagrams as mean values ± S.D. and were
compared by ANOVA with the Tukey’s multiple comparison post hoc test, using the GraphPad Prism 6.0 soware
(GraphPad Soware, Boston, MA, USA). Dierences with a p < 0.05 value were considered to be statistically
signicant. Genotype–phenotype association was assessed by χ2-test comparing the genotype distribution of the
patient and the control groups (i.e., additive model). Power of the genetic association study was assessed by the
GAS Power Calculator on line tool (https:// csg. sph. umich. edu/ abeca sis/ cats/ gas_ power_ calcu lator/) using the
additive disease model (prevalence of T2DM is 6.28%, genotype relative risk was 1.2.).

All data are available in the main text or in the supplementary material. e raw data and uncropped blot images
underlying the above presented results, as well as all Supplementary Figures and Tables are enclosed in the Sup-
plementary Information le. Any additional data from this study is available from the corresponding authors
(zambo.veronika@med.semmelweis-univ.hu and kereszturi.eva@semmelweis.hu) upon reasonable request.
Received: 28 August 2023; Accepted: 23 December 2023

1. de Carvalho, C. & Caramujo, M. J. e various roles of fatty acids. Molecules 23, 2583 (2018).
2. Shimabukuro, M., Zhou, Y. T., Levi, M. & Unger, R. H. Fatty acid-induced beta cell apoptosis: a link between obesity and diabetes.
Proc. Natl. Acad. Sci. U.S.A. 95, 2498–2502 (1998).
3. Han, J. & Kaufman, R. J. e role of ER stress in lipid metabolism and lipotoxicity. J. Lipid Res. 57, 1329–1338 (2016).
4. Yazıcı, D. & Sezer, H. Insulin resistance, obesity and lipotoxicity. Adv. Exp. Med. Biol. 960, 277–304 (2017).
5. Colvin, B. N., Longtine, M. S., Chen, B., Costa, M. L. & Nelson, D. M. Oleate attenuates palmitate-induced endoplasmic reticulum
stress and apoptosis in placental trophoblasts. Reproduction (Cambridge, England) 153, 369–380 (2017).
6. Sarnyai, F. et al. Eect of cis- and trans-Monounsaturated Fatty Acids on Palmitate Toxicity and on Palmitate-induced Accumula-
tion of Ceramides and Diglycerides. Int. J. Mol. Sci. 21, 2626 (2020).
7. Maedler, K. et al. Distinct eects of saturated and monounsaturated fatty acids on beta-cell turnover and function. Diabetes 50,
69–76 (2001).
8. Chen, X. et al. Oleic acid protects saturated fatty acid mediated lipotoxicity in hepatocytes and rat of non-alcoholic steatohepatitis.
Life Sci. 203, 291–304 (2018).
9. Islam, M. A. et al. Trans fatty acids and lipid prole: A serious risk factor to cardiovascular disease, cancer and diabetes. Diabetes
Metabol. Syndr. 13, 1643–1647 (2019).
10. Okamura, T. et al. Trans fatty acid intake induces intestinal inammation and impaired glucose tolerance. Front. Immunol. 12,
669672 (2021).
11. Schoeneck, M. & Iggman, D. e eects of foods on LDL cholesterol levels: A systematic review of the accumulated evidence from
systematic reviews and meta-analyses of randomized controlled trials. Nutr. Metabol. Cardiovasc. Diseases 31, 1325–1338 (2021).
12. Harvey, K. A. et al. Trans-fatty acids induce pro-inammatory responses and endothelial cell dysfunction. Brit. J. Nutr. 99, 723–731
(2008).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
13
Vol.:(0123456789)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
13. Michels, N., Specht, I. O., Heitmann, B. L., Chajès, V. & Huybrechts, I. Dietary trans-fatty acid intake in relation to cancer risk: a
systematic review and meta-analysis. Nutr. Rev. 79, 758–776 (2021).
14. Ginter, E. & Simko, V. New data on harmful eects of trans-fatty acids. Bratislavske lekarske listy 117, 251–253 (2016).
15. Enoch, H. G., Catalá, A. & Strittmatter, P. Mechanism of rat liver microsomal stearyl-CoA desaturase. Studies of the substrate
specicity, enzyme-substrate interactions, and the function of lipid. J. Biol. Chem. 251, 5095–5103 (1976).
16. Paton, C. M. & Ntambi, J. M. Biochemical and physiological function of stearoyl-CoA desaturase. Am. J. Physiol. Endocrinol.
Metabol. 297, 28–37 (2009).
17. Mauvoisin, D. & Mounier, C. Hormonal and nutritional regulation of SCD1 gene expression. Biochimie 93, 78–86 (2011).
18. Zhang, L., Ge, L., Tran, T., Stenn, K. & Prouty, S. M. Isolation and characterization of the human stearoyl-CoA desaturase gene
promoter: Requirement of a conserved CCAAT cis-element. Biochem. J. 357, 183–193 (2001).
19. Zulkii, R. M., Parr, T., Salter, A. M. & Brameld, J. M. Regulation of ovine and porcine stearoyl coenzyme A desaturase gene
promoters by fatty acids and sterols. J. Animal Sci. 88, 2565–2575 (2010).
20. Yao, D. W. et al. Characterization of the liver X receptor-dependent regulatory mechanism of goat stearoyl-coenzyme A desaturase
1 gene by linoleic acid. J. Dairy Sci. 99, 3945–3957 (2016).
21. Kato, H., Sakaki, K. & Mihara, K. Ubiquitin-proteasome-dependent degradation of mammalian ER stearoyl-CoA desaturase. J.
Cell Sci. 119, 2342–2353 (2006).
22. Zhang, J. et al. EGFR modulates monounsaturated fatty acid synthesis through phosphorylation of SCD1 in lung cancer. Mole.
Cancer 16, 127 (2017).
23. Murakami, A., Nagao, K., Juni, N., Hara, Y. & Umeda, M. An N-terminal di-proline motif is essential for fatty acid-dependent
degradation of Δ9-desaturase in Drosophila. J. Biol. Chem. 292, 19976–19986 (2017).
24. Tibori, K. et al. Molecular mechanisms underlying the elevated expression of a potentially type 2 diabetes mellitus associated SCD1
variant. Int. J. Mole. Sci. 23, 6621 (2022).
25. Pan, G., Cavalli, M. & Wadelius, C. Polymorphisms rs55710213 and rs56334587 regulate SCD1 expression by modulating HNF4A
binding. Biochimica et Biophysica Acta Gene Regulatory Mechanisms 1864, 194724 (2021).
26. Liu, Z. et al. SCD rs41290540 single-nucleotide polymorphism modies miR-498 binding and is associated with a decreased risk
of coronary artery disease. Mole. Genet. Genom. Med. 8, e1136 (2020).
27. Zámbó, V. et al. A single nucleotide polymorphism (rs3811792) aecting human SCD5 promoter activity is associated with diabetes
mellitus. Genes 13, 1784 (2022).
28. Dittmer, J. e role of the transcription factor Ets1 in carcinoma. Seminars Cancer Biol. 35, 20–38 (2015).
29. Dalla Valle, A. et al. Induction of stearoyl-CoA 9-desaturase 1 protects human mesenchymal stromal cells against palmitic acid-
induced lipotoxicity and inammation. Front. Endocrinol. 10, 726 (2019).
30. Yang, C., Lim, W., Bazer, F. W. & Song, G. Down-regulation of stearoyl-CoA desaturase-1 increases susceptibility to palmitic-acid-
induced lipotoxicity in human trophoblast cells. J. Nutr. Biochem. 54, 35–47 (2018).
31. Minville-Walz, M. et al. Distinct regulation of stearoyl-CoA desaturase 1 gene expression by cis and trans C18:1 fatty acids in
human aortic smooth muscle cells. Genes Nutr. 7, 209–216 (2012).
32. ALJohani, A. M., Syed, D. N. & Ntambi, J. M. Insights into Stearoyl-CoA Desaturase-1 Regulation of Systemic Metabolism. Trends
Endocrinol. Metabol 28, 831–842 (2017).
33. Liu, Y., Li, J. & Liu, Y. Eects of epoxy stearic acid on lipid metabolism in HepG2 cells. J. Food Sci. 85, 3644–3652 (2020).
34. Sarnyai, F. et al. Dierent metabolism and toxicity of TRANS fatty acids, elaidate and vaccenate compared to cis-oleate in HepG2
cells. Int. J. Mole. Sci. 23, 7298 (2022).
35. Hirata, Y. et al. trans-Fatty acids promote proinammatory signaling and cell death by stimulating the apoptosis signal-regulating
kinase 1 (ASK1)-p38 pathway. J. Biol. Chem. 292, 8174–8185 (2017).
36. Oteng, A. B. et al. Feeding Angptl4(-/-) mice trans fat promotes foam cell formation in mesenteric lymph nodes without leading
to ascites. J. Lipid Res. 58, 1100–1113 (2017).
37. Monguchi, T. et al. Excessive intake of trans fatty acid accelerates atherosclerosis through promoting inammation and oxidative
stress in a mouse model of hyperlipidemia. J. Cardiol. 70, 121–127 (2017).
38. Oteng, A. B., Loregger, A., van Weeghel, M., Zelcer, N. & Kersten, S. Industrial trans fatty acids stimulate SREBP2-mediated
cholesterogenesis and promote non-alcoholic fatty liver disease. Mole. Nutr. Food Res. 63, e1900385 (2019).
39. Jeyapal, S. et al. Chronic consumption of fructose in combination with trans fatty acids but not with saturated fatty acids induces
nonalcoholic steatohepatitis with brosis in rats. Eur. J. Nut r. 57, 2171–2187 (2018).
40. Stender, S., Astrup, A. & Dyerberg, J. Ruminant and industrially produced trans fatty acids: health aspects. Food Nutr. Res. 52,
1651 (2008).
41. Estadella, D. et al. Lipotoxicity: eects of dietary saturated and transfatty acids. Media. Inamm. 2013, 137579 (2013).
42. Jakobsen, M. U., Over vad, K., Dyerberg, J. & Heitmann, B. L. Intake of ruminant trans fatty acids and risk of coronary heart disease.
Int. J. Epidemiol. 37, 173–182 (2008).
43. Gebauer, S. K. et al. Eects of ruminant trans fatty acids on cardiovascular disease and cancer: a comprehensive review of epide-
miological, clinical, and mechanistic studies. Adv. Nutrition 2, 332–354 (2011).
44. Brouwer, I. A., Wanders, A. J. & Katan, M. B. Eect of animal and industrial trans fatty acids on HDL and LDL cholesterol levels
in humans–a quantitative review. PloS One 5, e9434 (2010).
45. Gebauer, S. K., Destaillats, F., Dionisi, F., Krauss, R. M. & Baer, D. J. Vaccenic acid and trans fatty acid isomers from partially hydro-
genated oil both adversely aect LDL cholesterol: A double-blind, randomized controlled trial. Am. J. Clin. Nutr. 102, 1339–1346
(2015).
46. Da Silva, M. S., Julien, P., Bilodeau, J. F., Barbier, O. & Rudkowska, I. Trans fatty acids suppress TNF-α-induced inammatory gene
expression in endothelial (HUVEC) and hepatocellular carcinoma (HepG2) Cells. Lipids 52, 315–325 (2017).
47. Yang, C., Lim, W., Bazer, F. W. & Song, G. Oleic acid stimulation of motility of human extravillous trophoblast cells is mediated
by stearoyl-CoA desaturase-1 activity. Mole. Human Reprod. 23, 755–770 (2017).
48. Jaudszus, A. et al. Vaccenic acid-mediated reduction in cytokine production is independent of c9, t11-CLA in human peripheral
blood mononuclear cells. Biochimica et Biophysica Acta 1821, 1316–1322 (2012).
49. Min, J. Y. & Kim, D. H. Stearoyl-CoA desaturase 1 as a therapeutic biomarker: Focusing on cancer stem cells. Int. J. Mole. Sci. 24,
8951 (2023).
50. Sen, U., Coleman, C. & Sen, T. Stearoyl coenzyme A desaturase-1: multitasker in cancer, metabolism, and ferroptosis. Tren ds
Cancer 9, 480–489 (2023).
51. Stryjecki, C. & Mutch, D. M. Fatty acid-gene interactions, adipokines and obesity. Eur. J. Clin. Nutr. 65, 285–297 (2011).
52. Arregui, M. et al. Heterogeneity of the Stearoyl-CoA desaturase-1 (SCD1) gene and metabolic risk factors in the EPIC-Potsdam
study. PloS One 7, e48338 (2012).
53. Merino, D. M., Ma, D. W. & Mutch, D. M. Genetic variation in lipid desaturases and its impact on the development of human
disease. Lipids Health Disease 9, 63 (2010).
54. Verschoor, M. L., Verschoor, C. P. & Singh, G. Ets-1 global gene expression prole reveals associations with metabolism and oxida-
tive stress in ovarian and breast cancers. Cancer Metabol. 1, 17 (2013).
55. Verschoor, M. L., Wilson, L. A., Verschoor, C. P. & Singh, G. Ets-1 regulates energy metabolism in cancer cells. PloS One 5, e13565
(2010).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
14
Vol:.(1234567890)
 | (2024) 14:177 | https://doi.org/10.1038/s41598-023-50700-5
www.nature.com/scientificreports/
56. Oballa, R. M. et al. Development of a liver-targeted stearoyl-CoA desaturase (SCD) inhibitor (MK-8245) to establish a therapeutic
window for the treatment of diabetes and dyslipidemia. J. Med. Chem. 54, 5082–5096 (2011).
57. Elk, N. & Iwuchukwu, O. F. Using personalized medicine in the management of diabetes mellitus. Pharmacotherapy 37, 1131–1149
(2017).
58. Masulli, M. et al. e Pro12Ala polymorphism of PPARγ2 modulates beta cell function and failure to oral glucose-lowering drugs
in patients with type 2 diabetes. Diabetes Metabol. Res. Rev. 37, e3392 (2021).
59. Castro-Mondragon, J. A. et al. JASPAR 2022: the 9th release of the open-access database of transcription factor binding proles.
Nucl. Acids Res. 50, D165-d173 (2022).
60. McLaren, W. et al. e ensembl variant eect predictor. Genome Biol. 17, 122 (2016).

We thank Ms. Valéria Mile and Ms. Viktória Molnár for their skillful technical assistance.

K.T.: methodology, investigation, visualization, writingoriginal dra preparation; V.Z.: methodology, investi-
gation, writingoriginal dra preparation, funding acquisition; G.O.: validation; P.S.: visualization; F.S.: valida-
tion; V.T.: writingreview and editing; Z.R.: soware, writingreview and editing, funding acquisition; M.C.:
writingreview and editing, funding acquisition; É.K.: conceptualization, writingoriginal dra preparation,
writingreview and editing, visualization, funding acquisition.

is work was supported by the Hungarian National Research, Development and Innovation Oce (NKFIH
grant numbers: FK138115, K131680 and PD142709). Project no. TKP2021-EGA-24 was implemented with
the support provided by the Ministry of Innovation and Technology of Hungary from the National Research,
Development and Innovation Fund, and nanced under the TKP2021-EGA funding scheme.

e authors declare no competing interests.

Supplementary Information e online version contains supplementary material available at https:// doi. org/
10. 1038/ s41598- 023- 50700-5.
Correspondence and requests for materials should be addressed to V.Z.orÉ.K.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.
© e Author(s) 2024
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Four SNPs (rs1054411, rs670213, rs2275657, and rs2275656) have been identified in the 5′ regulatory region of SCD1 and demonstrated not to affect promoter activity per se. Moreover, it has been evidenced that rs1054411 enhances transcription in a FA-dependent manner, while the same polymorphism has been shown to reduce gene expression through ETS1 TF-binding [24]. In contrast, several genetic variations in the 3′UTR have been associated with specific conditions affecting lipid metabolism. ...
... Four SNPs (rs1054411, rs670213, rs2275657, and rs2275656) have been identified in the 5 ′ regulatory region of SCD1 and demonstrated not to affect promoter activity per se. Moreover, it has been evidenced that rs1054411 enhances transcription in a FA-dependent manner, while the same polymorphism has been shown to reduce gene expression through ETS1 TF-binding [24]. In contrast, several genetic variations in the 3 ′ UTR have been associated with specific conditions affecting lipid metabolism. ...
... Nevertheless, it is important to note that, in addition to in silico analyses and in vitro or in vivo effect studies, the impact of environmental factors fine-tuned by genetic mutations can only be fully understood when they are considered in combination. The role of gene-environment interactions is particularly pronounced in lipid metabolism-related conditions [8,[57][58][59], as evidenced by the influence of natural genetic variants on SCD1 promoter activity [24] or protein stability [23], among other factors, in a FA-dependent manner. ...
Article
Full-text available
Background: A considerable proportion of the symptoms associated with excessive dietary intake can be attributed to systemic imbalances in lipid metabolism. The prominent toxicity of saturated fatty acids has been repeatedly demonstrated and sheds light on the protective role of stearoyl-CoA desaturase-1 (SCD1), the key enzyme for fatty acid desaturation. SCD1 protein expression is regulated at the levels of transcription, translation, and degradation. However, the modulating effect of the variability of the human genome must also be taken into account. Therefore, we aimed to ascertain whether natural missense or frameshift mutations in SCD1 (p.H125P, p.M224L, p.A333T, p.R253AfsTer7) could influence the expression, degradation, or function of the enzyme. Methods: In silico and in vitro experiments were conducted to comprehensively evaluate the consequences associated with each genetic variation, with the objective of using the results to propose a risk or severity ranking of SCD1 variants. Results: As anticipated, the p.R253AfsTer7 variant was identified as the most deleterious in structural, functional, and quantitative terms. The p.H125P variant also reduced the desaturation capacity of the enzyme in accordance with the predicted structural alterations and augmented degradation resulting from folding complications. This was aggravated by increased mRNA instability and accompanied by mild endoplasmic reticulum stress induction. The p.A333T protein exhibited an intermediate phenotype, whereas p.M224L showed no deleterious effects and even increased the amount of SCD1. Conclusions: In conclusion, the large-scale identification of genetic variations needs to be supplemented with comprehensive functional characterization of these variations to facilitate adequate personalized prevention and treatment of lipid metabolism-related conditions.
... Stearyl CoA desaturase-1 (SCD1) is an essential enzyme that regulates the transformation of saturated fatty acids (SFA) into monounsaturated fatty acids (MUFA), and plays a crucial role in controlling fatty acid metabolism and the balance between SFA and MUFA (Balatskyi & Dobrzyn, 2023). Activated SCD1 promotes MUFA biosynthesis, fat accumulation, and the development of T2DM (Igal & Sinner, 2021;Tibori et al., 2022Tibori et al., , 2024. Recently, multiple studies have demonstrated that reducing the expression levels of sbp-1, fat-5, fat-6, and fat-7 significantly decreases intestinal fat accumulation in C. elegans (Bai et al., 2021;Nomura et al., 2010). ...
Article
Full-text available
Sweet pepper, a globally commercialized horticultural crop, has been demonstrated to impede fat accumulation, but its mechanism remains incompletely understood. This study was designed to explore the potential mechanism of sweet pepper in reducing fat accumulation in Caenorhabditis elegans through RNA‐seq and metabolome analysis. A total of 22 metabolites were identified from sweet pepper by UHPLC‐ESI‐TOF‐MS analysis. In vivo, sweet pepper significantly inhibited α‐glucosidase activity and reduced the levels of glucose, triglycerides (TG), total cholesterol (TC), and the area stained with oil red O. Additionally, it increased body length and the number of head swings in C. elegans compared to the control group. The KEGG enrichment analysis revealed significant enrichment of the biosynthesis of unsaturated fatty acids signaling pathway among the differentially expressed genes and metabolites. Furthermore, the mRNA levels of sterol regulatory element‐binding proteins (SREBPs) ortholog SBP‐1, as well as the stearyl CoA desaturase‐1 (SCD1), including fat‐5, fat‐6, and fat‐7, were significantly decreased after treatment with sweet pepper. Collectively, sweet pepper effectively reduces fat accumulation, which is probably related to downregulating the SREBP‐SCD axis, offering new insights for future functional food development.
Article
Scope The study aims to analyze transcriptomic profiles in adipose tissues postconsumption of elaidic acid (EA; trans ‐18:1 n ‐9) and trans ‐palmitoleic acid (TPA; trans ‐16:1 n ‐7), elucidating their different effects on inflammation and glucose metabolism. Methods and results Twenty C57BL/6 mice are divided into four groups. Each group receives one of the following formulations in drinking water: lecithin nanovesicles, nanovesicles containing either lecithin with EA or TPA (86:14 w/w), or water (control) for 28 days with a regular fat diet (18% calories from fat). Total RNA is extracted, and paired‐end sequencing is performed. TPA intake alters the expression of 351 genes compared to EA intake, including 11 downregulated and 340 upregulated genes (fold change [FC] >1.5, p < 0.05). TPA compares to EA upregulated: Slc5a8, Lcn2, Csf3, Scube1, Mapk13, Bdkrb2, Ctla2a, Slc2a1, Oas3, Cx3cl1, Oas2, Nlrp6, Pycard, Cyba, Ddr1, and Prkab1 and downregulated Fas gene. These genes are related to the NOD‐like receptor, lipid and atherosclerosis, IL‐17 signaling, TNF, nonalcoholic fatty liver disease, cytokine–cytokine receptor interaction, adipocytokine, glucagon, insulin resistance, and inflammatory mediator regulation of TRP channels signaling. Conclusion TPA intake has a distinct impact on the regulation of inflammation and diabetes‐related pathways in adipose tissue compared to EA.
Article
Full-text available
The dysregulation of lipid metabolism and alterations in the ratio of monounsaturated fatty acids (MUFAs) to saturated fatty acids (SFAs) have been implicated in cancer progression and stemness. Stearoyl-CoA desaturase 1 (SCD1), an enzyme involved in lipid desaturation, is crucial in regulating this ratio and has been identified as an important regulator of cancer cell survival and progression. SCD1 converts SFAs into MUFAs and is important for maintaining membrane fluidity, cellular signaling, and gene expression. Many malignancies, including cancer stem cells, have been reported to exhibit high expression of SCD1. Therefore, targeting SCD1 may provide a novel therapeutic strategy for cancer treatment. In addition, the involvement of SCD1 in cancer stem cells has been observed in various types of cancer. Some natural products have the potential to inhibit SCD1 expression/activity, thereby suppressing cancer cell survival and self-renewal activity.
Article
Full-text available
Cancer progression is a highly balanced process and is maintained by a sequence of finely tuned metabolic pathways. Stearoyl coenzyme A desaturase-1 (SCD1), the fatty enzyme that converts saturated fatty acids into monounsaturated fatty acids, is a critical modulator of the fatty acid metabolic pathway. SCD1 expression is associated with poor prognosis in several cancer types. SCD1 triggers an iron-dependent cell death called ferroptosis and elevated levels of SCD1 protect cancer cells against ferroptosis. Pharmacological inhibition of SCD1 as monotherapy and in combination with chemotherapeutic agents shows promising antitumor potential in preclinical models. In this review, we summarize the role of SCD in cancer cell progression, survival, and ferroptosis and discuss potential strategies to exploit SCD1 inhibition in future clinical trials.
Article
Full-text available
The combined prevalence of type 1 (T1DM) and type 2 (T2DM) diabetes mellitus is 10.5% worldwide and this is constantly increasing. The pathophysiology of the diseases include disturbances of the lipid metabolism, in which acyl-CoA desaturases play a central role as they synthesize unsaturated fatty acids, thereby providing protection against lipotoxicity. The stearoyl-CoA desaturase-5 (SCD5) isoform has received little scientific attention. We aimed to investigate the SCD5 promoter and its polymorphisms in vitro, in silico and in a case-control study. The SCD5 promoter region was determined by a luciferase reporter system in HepG2, HEK293T and SK-N-FI cells and it was proved to be cell type-specific, but it was insensitive to different fatty acids. The effect of the SCD5 promoter polymorphisms rs6841081 and rs3811792 was tested in the transfected cells. The T allele of rs3811792 single nucleotide polymorphism (SNP) significantly reduced the activity of the SCD5 promoter in vitro and modified several transcription factor binding sites in silico. A statistically significant association of rs3811792 SNP with T1DM and T2DM was also found, thus supporting the medical relevance of this variation and the complexity of the molecular mechanisms in the development of metabolic disorders. In conclusion, the minor allele of rs3811792 polymorphism might contribute to the development of diabetes by influencing the SCD5 promoter activity.
Article
Full-text available
Trans fatty acids (TFAs) are not synthesized in the human body but are generally ingested in substantial amounts. The widespread view that TFAs, particularly those of industrial origin, are unhealthy and contribute to obesity, cardiovascular diseases and diabetes is based mostly on in vivo studies, and the underlying molecular mechanisms remain to be elucidated. Here, we used a hepatoma model of palmitate-induced lipotoxicity to compare the metabolism and effects of the representative industrial and ruminant TFAs, elaidate and vaccenate, respectively, with those of cis-oleate. Cellular FAs, triacylglycerols, diacylglycerols and ceramides were quantitated using chromatography, markers of stress and apoptosis were assessed at mRNA and protein levels, ultrastructural changes were examined by electron microscopy and viability was evaluated by MTT assay. While TFAs were just slightly more damaging than oleate when applied alone, they were remarkably less protective against palmitate toxicity in cotreatments. These differences correlated with their diverse incorporation into the accumulating diacylglycerols and ceramides. Our results provide in vitro evidence for the unfavorable metabolic features and potent stress-inducing character of TFAs in comparison with oleate. These findings strengthen the reasoning against dietary trans fat intake, and they can also help us better understand the molecular mechanisms of lipotoxicity.
Article
Full-text available
Disturbances in lipid metabolism related to excessive food intake and sedentary lifestyle are among major risk of various metabolic disorders. Stearoyl-CoA desaturase-1 (SCD1) has an essential role in these diseases, as it catalyzes the synthesis of unsaturated fatty acids, both supplying for fat storage and contributing to cellular defense against saturated fatty acid toxicity. Recent studies show that increased activity or over-expression of SCD1 is one of the contributing factors for type 2 diabetes mellitus (T2DM). We aimed to investigate the impact of the common missense rs2234970 (M224L) polymorphism on SCD1 function in transfected cells. We found a higher expression of the minor Leu224 variant, which can be attributed to a combination of mRNA and protein stabilization. The latter was further enhanced by various fatty acids. The increased level of Leu224 variant resulted in an elevated unsaturated: saturated fatty acid ratio, due to higher oleate and palmitoleate contents. Accumulation of Leu224 variant was found in a T2DM patient group, however, the difference was statistically not significant. In conclusion, the minor variant of rs2234970 polymorphism might contribute to the development of obesity-related metabolic disorders, including T2DM, through an increased intracellular level of SCD1.
Article
Full-text available
JASPAR (http://jaspar.genereg.net/) is an open-access database containing manually curated, non-redundant transcription factor (TF) binding profiles for TFs across six taxonomic groups. In this 9th release, we expanded the CORE collection with 341 new profiles (148 for plants, 101 for vertebrates, 85 for urochordates, and 7 for insects), which corresponds to a 19% expansion over the previous release. We added 298 new profiles to the Unvalidated collection when no orthogonal evidence was found in the literature. All the profiles were clustered to provide familial binding profiles for each taxonomic group. Moreover, we revised the structural classification of DNA binding domains to consider plant-specific TFs. This release introduces word clouds to represent the scientific knowledge associated with each TF. We updated the genome tracks of TFBSs predicted with JASPAR profiles in eight organisms; the human and mouse TFBS predictions can be visualized as native tracks in the UCSC Genome Browser. Finally, we provide a new tool to perform JASPAR TFBS enrichment analysis in user-provided genomic regions. All the data is accessible through the JASPAR website, its associated RESTful API, the R/Bioconductor data package, and a new Python package, pyJASPAR, that facilitates serverless access to the data.
Article
Full-text available
Background and Aims Many nutritional and epidemiological studies have shown that high consumption of trans fatty acids can cause several adverse effects on human health, including cardiovascular disease, diabetes, and cancer. In the present study, we investigated the effect of trans fatty acids on innate immunity in the gut by observing mice fed with a diet high in trans fatty acids, which have been reported to cause dysbiosis. Methods We used C57BL6/J mice and fed them with normal diet (ND) or high-fat, high-sucrose diet (HFHSD) or high-trans fatty acid, high-sucrose diet (HTHSD) for 12 weeks. 16S rRNA gene sequencing was performed on the mice stool samples, in addition to flow cytometry, real-time PCR, and lipidomics analysis of the mice serum and liver samples. RAW264.7 cells were used for the in vitro studies. Results Mice fed with HTHSD displayed significantly higher blood glucose levels and advanced fatty liver and intestinal inflammation, as compared to mice fed with HFHSD. Furthermore, compared to mice fed with HFHSD, mice fed with HTHSD displayed a significant elevation in the expression of CD36 in the small intestine, along with a reduction in the expression of IL-22. Furthermore, there was a significant increase in the populations of ILC1s and T-bet-positive ILC3s in the lamina propria in mice fed with HTHSD. Finally, the relative abundance of the family Desulfovibrionaceae, which belongs to the phylum Proteobacteria, was significantly higher in mice fed with HFHSD or HTHSD, than in mice fed with ND; between the HFHSD and HTHSD groups, the abundance was slightly higher in the HTHSD group. Conclusions This study revealed that compared to saturated fatty acid intake, trans fatty acid intake significantly exacerbated metabolic diseases such as diabetes and fatty liver.
Article
Full-text available
Aims To systematically evaluate the evidence regarding the effects of foods on LDL cholesterol levels and to compare the findings with current guidelines. Data Synthesis From inception through June 2019, we searched PubMed, Cochrane Database of Systematic Reviews, and Cochrane Central Register of Controlled Trials for guidelines, systematic reviews, and RCTs (for coffee intake only) of at least 13 days duration. Additionally, we searched Trip database for guidelines from 2009 through Oct 2019. Language was restricted to English. The strength of evidence was evaluated using The Grading of Recommendations Assessment, Development, and Evaluation (GRADE). A total of 37 guidelines, 108 systematic reviews, and 20 RCTs were included. With high evidence, foods high in unsaturated and low in saturated and trans fatty acids (e.g. rapeseed/canola oil), with added plant sterols/stanols, and high in soluble fibre (e.g. oats, barley, and psyllium) caused at least moderate (i.e. 0.20–0.40 mmol/L) reductions in LDL cholesterol. Unfiltered coffee caused a moderate to large increase. Soy protein, tomatoes, flaxseeds, and almonds caused small reductions. With moderate evidence, avocados and turmeric caused moderate to large reductions. Pulses, hazelnuts, walnuts, high-fibre/wholegrain foods, and green tea caused small to moderate reductions, whereas sugar caused a small increase. Other identified foods were either neutral or had low or very low evidence regarding their effects. Conclusions Several foods distinctly modify LDL cholesterol levels. The results may aid future guidelines and dietary advice for hypercholesterolemia.
Article
Full-text available
Context Apart from ruminant fat, trans-fatty acids are produced during the partial hydrogenation of vegetable oils, (eg, in the production of ultraprocessed foods). Harmful cardiovascular effects of trans-fatty acids are already proven, but the link with cancer risk has not yet been summarized. Objective A systematic review (following PRISMA guidelines) – including observational studies on the association of trans-fatty acid intake with any cancer risk – was conducted, with no limitations on population types. Data Sources The electronic databases PubMed and Embase were searched to identify relevant studies. Data Extraction This systematic review included 46 articles. Quality was assessed via the Newcastle-Ottawa scale. Meta-analyses were conducted if at least 4 articles exploring the same transfat-cancer pairings were found. Data analysis Nineteen cancer types have been researched in cohort and case-control studies on trans-fatty acids, with breast cancer (n = 17), prostate cancer (n = 11), and colorectal cancer (n = 9) as the most researched. The meta-analyses on total trans-fat showed a significant positive association for prostate cancer (odds ratio [OR] 1.49; 95%CI, 1.13–1.95) and colorectal cancer (OR 1.26; 95%CI, 1.08–1.46) but not for breast cancer (OR 1.12; 95%CI, 0.99–1.26), ovarian cancer (OR 1.10; 95%CI, 0.94–1.28), or non-Hodgkin lymphoma (OR 1.32; 95%CI, 0.99–1.76). Results were dependent on the fatty acid subtype, with even cancer-protective associations for some partially hydrogenated vegetable oils. Enhancing moderators in the positive transfat-cancer relation were gender (direction was cancer-site specific), European ancestry, menopause, older age, and overweight. Conclusion Despite heterogeneity, higher risk of prostate and colorectal cancer by high consumption of trans-fatty acids was found. Future studies need methodological improvements (eg, using long-term follow-up cancer data and intake biomarkers). Owing to the lack of studies testing trans-fatty acid subtypes in standardized ways, it is not clear which subtypes (eg, ruminant sources) are more carcinogenic. Systematic Review Registration PROSPERO registration no. CRD42018105899
Article
The stearoyl-CoA desaturase 1 (SCD1) gene at 10q24.31 encodes the rate limiting enzyme SCD1 that catalyzes the biosynthesis of monounsaturated fatty acids (MUFAs) from saturated fatty acids (SFAs). Dysregulated SCD1 activity has been observed in many human diseases including non-alcoholic fatty liver disease (NAFLD), obesity, hypertension, hyperlipidemia, metabolic syndrome and several types of cancer. HNF4A is a central regulator of glucose and lipid metabolism and previous studies suggested that it is deeply involved in regulating the SCD1 activity in the liver. However, the underlying mechanisms on whether and how SCD1 is regulated by HNF4A have not been explored in detail. In this study, we found that HNF4A regulates SCD1 expression by directly binding to the key regulatory regions in the SCD1 locus. Knocking down of HNF4A significantly downregulated the expression of SCD1. Variants rs55710213 and rs56334587 in intron 5 of SCD1 directly reside in a canonical HNF4A binding site. The GG haplotype of rs55710213 and rs56334587 is associated with decreased SCD1 activity by disrupting the binding of HNF4A, which further decreased the enhancer activity and SCD1 expression. In conclusion, our study demonstrated that SCD1 is directly regulated by HNF4A, which may be helpful in the understanding of the altered metabolic pathways in many diseases associated with dysregulated SCD1 or HNF4A or both.