You are on page 1of 10

www.nature.

com/scientificreports

Correction: Retraction

OPEN Impact of different promoters,

LE
promoter mutation, and an
enhancer on recombinant protein

C
Received: 22 March 2017
Accepted: 17 August 2017 expression in CHO cells

TI
Published online: 05 September 2017
Wen Wang1,2, Yan-long Jia2, Yi-chun Li1, Chang-qin Jing3, Xiao Guo2, Xue-fang Shang1,
Chun-peng Zhao1 & Tian-yun Wang1

R
In the present study, six commonly used promoters, including cytomegalovirus major immediate-
early (CMV), the CMV enhancer fused to the chicken beta-actin promoter (CAG), human elongation
factor-1α (HEF-1α), mouse cytomegalovirus (mouse CMV), Chinese hamster elongation factor-1α
A
(CHEF-1α), and phosphoglycerate kinase (PGK), a CMV promoter mutant and a CAG enhancer, were
evaluated to determine their effects on transgene expression and stability in transfected CHO cells.
The promoters and enhancer were cloned or synthesized, and mutation at C-404 in the CMV promoter
D
was generated; then all elements were transfected into CHO cells. Stably transfected CHO cells were
identified via screening under the selection pressure of G418. Flow cytometry, qPCR, and qRT-PCR were
used to explore eGFP expression levels, gene copy number, and mRNA expression levels, respectively.
TE

Furthermore, the erythropoietin (EPO) gene was used to test the selected strong promoter. Of the six
promoters, the CHEF-1α promoter yielded the highest transgene expression levels, whereas the CMV
promoter maintained transgene expression more stably during long-term culture of cells. We conclude
that CHEF-1α promoter conferred higher level of EPO expression in CHO cells, but the CMV promoter
with its high levels of stability performs best in this vector system.
C

Currently, recombinant proteins are used in the treatment of different diseases1–3, and these therapeutic proteins
A

are produced in a large number of cell types4. Chinese hamster ovary (CHO) cells are the preferred producers
because of their capacity to perform proper protein folding, assembly, and post-translational modifications, sim-
ilar to human cells5, 6. However, problems of instability and recombinant protein expression silencing need to
R

be resolved urgently4–7 because they limit the application of recombinant proteins8, 9. To resolve these problems,
many elements have been investigated, including promoters, matrix attachment regions (MARs), introns, and
other cis-acting elements10–13.
A promoter is the region upstream of a specific gene that initiates its transcription. Promoters show different
ET

activities because of their composition or other cis-acting elements in the expression vector 14. In addition, the
promoter also shows cell compatibility in the episomal vector driven by human cytomegalovirus major (CMV)
immediate early gene promoter4. Mouse CMV promoter is suggested to be stronger than human CMV pro-
moter15, 16. The CMV promoter is a commonly used promoter for the production of high level recombinant pro-
tein in mammalian cells17. However, the expression level of the transgene driven by CMV promoter decreases
R

with extended culture times because of transcriptional silencing, which is associated with DNA methylation18, 19.
To achieve stable and increased expression of the transgene, other strong promoters of mammalian origin, com-
bined with cis-acting elements and synthetic promoters, have been investigated. Human eukaryotic translation
elongation factor 1 alpha (HEF-1α, gene symbol EEF1A1) promoter is constitutively active in a broad range of
cell types and often active in cells in which viral promoters fail to express downstream genes and in cells in which
the viral promoters are gradually silenced20, 21. The CHEF1-α promoter is more active in CHO cells compared

1
Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, 453003, Henan,
China. 2Pharmacy Collage, Xinxiang Medical University, Xinxiang, 453003, Henan, China. 3School of Life Sciences
and Technology, Xinxiang Medical University, Xinxiang, 453003, Henan, China. Wen Wang and Yan-long Jia
contributed equally to this work. Correspondence and requests for materials should be addressed to T.-y.W. (email:
wtianyuncn@126.com)

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 1


www.nature.com/scientificreports/

LE
C
TI
R
A
Figure 1.  Effect of different promoter on transfection efficiency and transient transgene expression. The pIRES-
mediated vectors containing CMV, CAG, CHEF-1α, CMV mutant, HEF1-α, mouse CMV, CAG and PGK were
transfected into CHO cells, and CHO cells were cultured in absence of G418 selection pressure for 48 h. (A) The
D
eGFP of cells fluorescence profile was observed under fluorescence microscope; (B) The transfection efficiency
were obtained using eGFP antibody analysis. (C) eGFP proteins transient expression levels. CMV promoter
was regarded as 100, the MFI of other promoter were calculated. CMV, Cytomegalovirus major immediate-
TE

early; CAG, the CMV enhancer fused to the chicken beta-actin promoter; CHEF-1α, Chinese hamster
elongation factor-1α; mouse CMV, mouse cytomegalovirus; HEF-1α, human elongation factor-1α; PGK,
phosphoglycerate kinase; and CMV protein mutant, CAG enhancer. EPO, erythropoietin; SpA, simian virus 40
early polyadenylation signal; eGFP, enhanced green fluorescence protein.
C

to CMV and SV40 promoters alone20, 22. Simian virus 40 (SV40) is also a strong promoter for the production
of therapeutic proteins in mammalian cells, providing a lower expression yield, but higher stability than the
A

EF1a and CMV promoters14. Mouse phosphoglycerate kinase 1 (PGK) promoter was also used to drive transgene
expression in different cell lines23, 24. Besides natural promoters, hybrid promoters and synthetic promoters were
also developed to drive higher and long-term expression of a transgene24, 25. CAG promoter is a hybrid construct
R

consisting of the cytomegalovirus (CMV) enhancer fused to the chicken beta-actin promoter, which is also used
to drive transgene expression in different cell lines26, 27. Brown et al. designed 140 synthetic promoters to spe-
cifically regulate the expression of recombinant genes in CHO cells, which offer precise control of recombinant
ET

transcriptional activity in CHO cells spanning over two orders of magnitude12. Schlabach et al. took a synthetic
biology approach for the generation and screening of transcription factor binding sites for activity in human cells,
and yielded compound enhancers that were capable of a twofold greater enhancement activity than the CMV
enhancer, with higher levels of activity than the original synthetic enhancer across multiple cell lines28.
Although previous reports attempted to identify strong promoters for transgene expression, none of the ideal
R

promoters can significantly increase and maintain stable transgene expression. In this study, we report a system-
atic comparison of six commonly used promoters (CMV, mouse CMV, CHEF-1α, PGK, CAG, and HEF-1α), a
CMV mutant, and a CAG enhancer in transfected CHO cell system. Our findings will benefit those choosing pro-
moters during vector design to generate transfected CHO cell lines with both high expression level and long-term
expression stability.

Results
Transfection efficiency and transient transgene expression.  Transient gene expression is especially
useful in early research when many potential therapeutic candidates are needed for evaluation or when a mole-
cule is needed at short notice29, 30. At 48 h post-transfection, the fluorescence intensity of CHO cells transfected
with different types of promoter was observed under a fluorescence microscope (Fig. 1A). CAG, HEF-1α, mouse
CMV, and CHEF-1α showed enhanced transgene expression when compared with CMV. The transfection effi-
ciency of CAG, HEF-1α, mouse CMV, and CHEF-1α was better than that of the CMV mutant and the CAG

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 2


www.nature.com/scientificreports/

LE
C
TI
R
A
D
Figure 2.  Cells were collected at 10 generation post-transfection and the eGFP MFI was measured by flow
cytometry. (A) The stably transfected cells were screened in medium containing G418 (800 μg/mL). The eGFP
TE

MFI of stably transfected cell lines containing different promoters were detected. Black bar represent the results
from after 10 generations analyzed by flow cytometry. (B) Fold statistical analysis results of expression level, and
the eGFP MFI was normalized to CMV promoter. Three stably transfected pools were generated for each vector.
Cells were collected and measured for the eGFP MFI with the FACS Calibur (*P < 0.05, **P < 0.01).
C

enhancer. We also observed that the mouse CMV presented a significantly higher transient efficiency (about
93.5%) than the control vector (about 57.6%), followed by CHEF1-α (90.7%), CAG (78.8%), and CAG enhancer
A

(74.3%) (Fig. 1B). DNA length influences the transfection efficiency, but the PGK promoter was the sixth shortest
in length, shorter than CAG, HEF1-α, CHEF-1α, the CMV mutant, and CMV, and showed the lowest activity.
This demonstrated that the length of the promoter is not a major factor affecting transfection efficiency.
R

Compared with the CMV promoter, the enhancement was the highest for CHEF1-α, which improved trans-
genic eGFP expression by 2.9-fold, followed by HEF1-α (2.4-fold). However, mouse CMV and CAG resulted in a
slight increase in transient transgene expression (Fig. 1C).
ET

Recombinant protein expression of stably transfected cells.  Transient expression cannot fully
reveal the function of different promoters, and it is important that vectors can be stably expressed in cells. When
stably transfected cell lines were screened out, the eGFP protein levels (MFI) were measured by using flow cytom-
etry (Fig. 2A). The cells transfected with CHEF-1α promoter-containing vectors exhibited the highest expression
R

levels, followed by those containing HEF-1α, CMV mutant, CMV, mouse CMV, CAG, CAG enhancer, and PGK.
When the eGFP expression level under the CMV promoter was considered as 100, the expression levels under the
CHEF-1α, HEF-1α, CMV mutant, mouse CMV, CAG, CAG enhancer, and PGK promoters were 218.13, 184.21,
164.33, 49.12, 47.95, 26.31, and 8.77, respectively (Fig. 2B). Therefore, expression under the HEF-1α, CHEF-1α,
CMV mutant, and CMV promoters was higher than that under the mouse CMV, CAG, CAG enhancer, and PGK
promoter. The highest activity was exhibited by the CHEF-1α promoter, with an MFI 2.18-fold that of the CMV
promoter, 24.86-fold that of the PGK promoter, and 8.29-fold that of the CAG enhancer.

Recombinant mRNA expression.  The mRNA expression level is closely related to that of recombinant
protein31. We measured the mRNA expression levels in cells transfected with CMV, CAG, HEF-1α, and CHEF-1α
promoters by qRT-PCR using eGFP as the target gene and GAPDH as the internal control. We found differences
in recombinant mRNA expression levels among the cells transfected with CMV, CAG, HEF-1α, and CHEF-1α
promoters; the highest level was found in CHEF-1α promoter-containing cells, followed by those containing the

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 3


www.nature.com/scientificreports/

LE
C
TI
R
Figure 3.  Recombinant expression at mRNA level in cells transfected with CMV, CAG, HEF-1α, and CHEF-1α
promoters at 10 generations post-transfection. (A) Target gene (eGFP) and internal reference gene (GAPDH)
A
were measured by qRT-PCR. (B) The mRNA expression levels of cells were calculated using percentage of
eGFP/GAPDH qRT-PCR values. qRT-PCR results were obtained three independent measurements.
D
CMV, HEF-1α, and CAG promoters (Fig. 3). The mRNA expression level was consistent with the protein expres-
sion level, but the increasing fold was not directly proportional, suggesting that a promoter can increase transgene
TE

expression not only at the mRNA level, but also by affecting post-transcriptional regulation.
Analysis of the stability of recombinant protein expression. After un-transfected cells were killed, the stably
transfected cells were screened out and divided into two groups: cultured in medium with G418 (500 μg/mL) to
maintain the selection pressure, or without G418. MFI was detected to evaluate the intensity of the expressed
eGFP until generation 60 post-transfection (Fig. 4A,B), and the relative retention of cells transfected with CMV,
CMV mutant, CAG, HEF-1α, CAG enhancer, mouse CMV, PGK, and CHEF-1α vectors was calculated (Fig. 4C).
C

Cells transfected with the vector containing the CMV promoter showed the most stable expression levels, the rel-
ative retention was 74.53% (with G418) and 70.59% (without G418) of the original expression level at 60 passages
post-transfection, followed by CHEF1-α, 43.49% (with G418) and 40.97% (without G418), mouse CMV, 63.75%
A

(with G418) and 59.78% (without G418), and CAG enhancer 56.58% (with G418) and 54.02% (without G418).
Cells containing the CMV mutant, CHEF-1α, HEF-1α, and PGK promoters were unstable, with only 12.31%
(with G418) and 11.37% (without G418), 43.49% (with G418) and 40.97% (without G418), 40.83% (with G418)
R

and 38.10% (without G418) retention, and 32.85% (without G418), 23.84% (with G418), respectively. The trans-
gene expression levels also differed and decreased gradually in stably transfected CHO cells.
ET

Gene copy number analysis.  To further study the mechanism of the promoters, 30 single cell clones trans-
fected with different vectors, HEF-1α, CHEF-1α, CMV, and CAG, were selected and the gene copy number was
further analyzed by using qPCR. Some differences in the number of plasmids per genome were detected among
the cells, indicating that the ratios of transgene copy number per genome were disparate for all plasmids in this
experiment. The copy numbers are presented in Fig. 5A. The relative mean gene copy numbers were 0.73 ± 0.09
(CAG), 6.89 ± 1.43 (CHEF-1α), 10.98 ± 0.94 (HEF-1α), 1 ± 0.17 (CMV) (Fig. 5B). This finding indicated that
R

transgene expression level was not related with gene copy number, suggesting that the promoter enhancing activ-
ity did not involve an increase in gene copy numbers.

EPO expression levels.  To evaluate the effect of promoter on the secreted protein expression, we selected a
therapeutic protein, erythropoietin (EPO), and analyzed its expression in CHO cells under serum-free medium
culture conditions. We transfected the EPO-expressing vectors driven by the CHEF1-α or CMV promoter into
CHO cells and analyzed the expression levels of EPO by using ELISA and Western blot. The mean EPO expres-
sion level of cells transfected with the CHEF1-α driven vector yielded 49.07 ± 2.4 mg/L (Fig. 6A), and those trans-
fected with the CMV driven vector yielded 23.56 ± 0.9 mg/L (Fig. 6B). The results showed that EPO expression
driven by the CHEF1-α promoter was about 2.08-fold higher than that driven by the CMV promoter. The highest
production of EPO in the single cell clone driven by CHEF1-α and CMV was 75.3 mg/L, 54.9 mg/L, respectively.

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 4


www.nature.com/scientificreports/

Figure 4.  The stability of eGFP expression in transfected CHO cells grown in the presence G418 selection
pressure or absence of G418. (A) The stably transfected cells were passaged until 60 generation in the presence
of G418 selection pressure (B) or in the absence of G418 selection pressure. The intensity of eGFP of cells were

LE
detected by flow cytometry at the passage 0, 10, 20, 30,40, 50, 60, respectively. The experiments were performed
in triplicates. (C) The eGFP expression retention was calculated as the ratio of the MFI at the 60 generations
stability testing to the MFI at the transient expression MFI testing. Retention of eGFP expression levels in cells
transfected with CMV, CAG, CHEF-1α, CMV mutant, HEF1-α, mouse CMV, and CAG enhancer promoter-
containing vectors (n = 3).

C
TI
R
A
D
Figure 5.  Gene copies per genome as determined by qPCR analysis. The gene copy number of CHO cells that
stably transfected with CMV, CAG, CHEF-1α, HEF1-α element-containing vector was detected using qPCR at
TE

10 generations post-transfection. (A) Thirty single stably clones was picked out and the gene copy number were
analyzed by qPCR to detected the relationship between gene copy number. (B) The cells transfected with CMV
promoter-containing vector was considered as 1.0. The relative copy number of the CAG, CHEF-1α, HEF1-α
gene was calculated (*P < 0.05).
C

In addition, Western blot results were further demonstrated that EPO expression driven by the CHEF1-α pro-
moter was higher than that of CMV promoter (Fig. 6C).
A

Discussion
An ideal vector should ensure persistent transgene expression without epigenetic effects32. The landscape of epi-
R

genetic silencing is complex, and many factors could influence transgene silencing33, 34. The promoter is a major
element in the expression cassette of gene therapy vectors, and optimal promoter selection can increase target
specificity and gene expression14. Several reports investigated the effect of promoters on transgene expression35, 36.
ET

At present, CMV is the most commonly used promoter for the production of recombinant protein 37. However,
the CMV promoter cannot maintain production stability over time and it has many potential methylation sites;
mutations and methylation will lead to lower productivity of recombinant proteins 38. Nonetheless, we demon-
strated that the CMV promoter is more stable than the mouse CMV, CHEF-1α, PGK, CAG, HEF-1α, and CMV
mutant promoters, and the CAG enhancer. Previous studies revealed that EF-1α can enhance transgene expres-
R

sion in a lentiviral vector-mediated system and produce high-level, stable transgene expression 39. We also found
that CHEF-1α can significantly improve exogenous gene expression, but with reduced stability in CHO cells. One
report demonstrated that the CAG promoter could enhance exogenous gene expression when it was cloned into
the pCAGGS-GFP vector and transfected into Balb/c mice40. However, we found that the CAG promoter cannot
enhance transgene expression in long-term culture. This phenomenon may result from methylation of the CAG
promoter41. Mammalian DNA is frequently methylated at cytosine bases that are part of CpG dinucleotides42.
In this study, the CMV mutant was unable to improve exogenous gene expression when the cytosine at position
404 was point-mutated to guanine; hence, we speculate that C-404-G point mutations may inhibit transgene
expression.
In the present study, the relative activities of six commonly used promoters (CMV, CAG, HEF-1α, mouse
CMV, CHEF-1α, and PGK), the CAG enhancer, and a CMV mutant were compared to explore the effects on
transgene expression in CHO cells. During the long-term passage of cells, non-viral-mediated vectors that con-
tain large DNA fragments can cause deficient transgene expression. Among the six promoters, CMV mutant, and

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 5


www.nature.com/scientificreports/

Figure 6.  Analysis of EPO protein. The vectors containing EPO were transfected into CHO cells, and the stably

LE
transfected cells were screened under G418 selective pressure and thirty single cell clones were picked out.
Cells supernatant were collected for analysis by ELISA to determine volumetric EPO production (mg/L). (A)
CHEF-1α-EPO-containinng vector; (B) CMV-EPO-containing vector; (C) Western blot analysis. Lane 1, Un-
transfected cells; Lane 2, CMV promoter; 3, CHEF1-α promoter, 4, Positive control.

C
CAG enhancer tested here, the CAG enhancer was the shortest in length, followed by the mouse CMV and PGK
promoters. The highest positive eGFP gene expression rate was achieved with the HEF1-α promoter-containing

TI
vector, indicating that transgene expression is not only affected by the length of the fragment, but also by multiple
factors, including cell growth conditions, cell type, and cell state. Our results show that, among the six promoters,
CAG enhancer and CMV mutant, CMV and CHEF-1α, are better promoters than the CMV mutant, CAG, CAG
enhancer, mouse CMV, HEF-1α, and PGK in mammalian cells for long-term cell culture. Results obtained by
flow cytometry and qPCR experiments revealed that the vector containing HEF-1α was integrated into the host

R
chromosome with the highest copy number, followed by CHEF-1α, CMV, and CAG. These results demonstrated
that there was no direct relationship between exogenous gene expression levels and gene copy number. qRT-PCR
was used to analyze the expression levels of mRNA and showed that the cells transfected with CHEF-1α had the

eGFP expression. A
highest expression levels, followed by HEF-1α, CMV, and CAG. These results are consistent with the results of

To further investigate the effect of strong promoter on the expression of a gene of interest, we evaluated the
expression of a therapeutic secreted protein, erythropoietin (EPO), driven by the CHEF-1α and CMV promot-
D
ers under serum-free medium culture conditions. The results showed that EPO expression level in CHO cells
transfected with the vector containing CHEF-1α was significantly higher than of that of cells transfected with the
vector containing the CMV promoter, which was consistent with eGFP gene expression.
TE

Many studies investigated synthetic promoters43. Human CMV and HEF-1α constructs increased transgene
expression by up to ten-fold44. However, human CMV usually peaks 1–2 days after transfection and the activity
is rapidly lost43. Considering the effects of CMV on the relative maintenance of transgenes and the activity of
CHEF-1α on transgene expression, we can use a CMV core and CHEF-1α to construct a synthetic promoter. The
synthetic promoter would shorten the vector length and potentially contribute to the improvement of transfec-
tion efficiency.
C

In conclusion, we found that CHEF-1α showed high transgene expression activity and CMV derived from
an IRES-mediated vector presented the highest retention rate. However, this experiment was performed only in
CHO cells and the results cannot be extrapolated to other cell lines. The development of such expression systems
A

is a major strategic task continually required for the expression of target proteins for research and is necessary
for any future long-term clinical application. Our study makes a significant contribution to such research and
applications.
R

Materials and Methods


Vector construction.  The vector containing the CMV promoter was obtained by cloning the enhanced
ET

green fluorescent protein (eGFP) from pEGFP-C1 (Clontech, Mountain View, USA) into the pIRES-neo vec-
tor (Clontech, Mountain View, USA). Vectors containing the CMV mutant, CAG enhancer (GenBank no:
AJ575208.1, position 100-386), CAG (GenBank no: GU299216.1, position 3-1664), HEF-1α (GenBank no:
AY188393.1, position 10964-12623), mouse CMV (GenBank no: KT343252.1, position 1103-1625), CHEF-1α
(GenBank no: KY447299.1, position 12-1346), and PGK (GenBank no: KJ175229.1, position 641-1195) were
constructed by using the vector containing the CMV promoter, which acted as the original vector based on pre-
R

viously described IRES-mediated vectors45 (Fig. 7A, supplement 1). The CMV mutant, CAG enhancer, and CAG,
CHEF-1α, mouse CMV, HEF1-α, PGK, and CMV promoters were 589 bp, 287 bp, 1662 bp, 1335 bp, 523 bp, 1659
bp, 555 bp, and 589 bp in length, respectively (Fig. 7B); eGFP was inserted into the vectors as a reporter gene. The
sequences of elements were synthesized by General Biosystems (Chuzhou, China). Meanwhile, to detect the effect
of promoter on secreted protein, we selected two promoters, CMV and CHEF1-α, to construct vectors contain-
ing the EPO cDNA (Fig. 7A). The EPO gene was synthesized according to the codon optimization sequence and
further cloned into the vector using standard methods46.

Cell culture and transfection.  CHO-S cells (Life Technologies # A11557-01) were maintained in
Dulbecco’s modified Eagle’s medium + F12 (Gibco, Carlsbad, CA, USA) supplemented with 10% fetal bovine
serum (Gibco, arlsbad, CA, USA), 1% penicillin and streptomycin (Beyotime, Shanghai, China). The cells were
cultured in a humidified atmosphere at 37 °C and 5% CO2. The cells were plated at a concentration of 2 × 105 cells/

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 6


www.nature.com/scientificreports/

LE
C
TI
R
A
Figure 7.  Schematic representation of vectors containing different promters and the length of different
elements. (A) The vectors construction that containing different element. (B) The lengths of the six different
D
promoters, and CMV mutant, CAG enhancer.
TE

well in 24-well plates and allowed to attach overnight. On the second day, after reaching 80% confluence, the cells
in each well were transfected with the vectors containing CMV, CMV mutant, CAG enhancer, CAG, CHEF-1α,
mouse CMV, HEF-1α, or PGK using 1 μL Lipofectamine 3000 Transfection Reagent (Invitrogen, Carlsbad, CA,
USA) per 1 μg vector according to the manufacturer’s instructions. At 48 h post-transfection, G418 (800 μg/mL)
was used to kill the un-transfected cell lines. Meanwhile, cells transfected with the different vectors were digested
C

and reseeded at a concentration of 5 × 104 cells/well in 12-well plates. Each cell type was plated into three wells.

Transient expression.  At 48 h post-transfection, transfection efficiency and transient eGFP expression
A

levels were analyzed by assessing the fluorescence intensity of transfected cells using fluorescence microscopy
(Nikon ECLIPSE Ti, Nikon, Japan). In order to more accurately analyze the results, the cells were assessed at
200 × magnification with an emission wavelength of 530 nm using a 530/15 bandpass filter for green fluorescence.
R

Briefly, cells were harvested after digestion with trypsin at 48 h post transfection. The MFI and transfection effi-
ciency were detected. Cells were resuspended in 100 µL of mouse anti-GFP antibody (ZSGB-Bio, Beijing, China)
and analyzed by using a flow cytometer. The numbers of eGFP-positive and eGFP-negative cells were calculated
ET

according to the flow cytometry results, and the transfection efficiency was represented by the ratio of the number
of eGFP-positive cells to the total cell number.

Stable expression of transfected cells and flow cytometry analysis.  Long-term stable expression
of the target gene is required for industrial production of recombinant proteins. Thus, cells transfected with plas-
R

mid vectors were passaged in pools and further cultured under G418 (800 μg/mL) selection for 16 days. When
the non-transfected cells were dead, the stably transfected cell colonies were screened out and the concentration
of G418 was reduced to 500 μg/mL to maintain the selection pressure. When cell colonies reached 90% conflu-
ence, the cells were harvested by using 0.25% trypsin/EDTA. Analyses were performed with a Guava EasyCyte
8HT flow cytometer (MilliporeSigma, Darmstadt, Germany) using FlowJo software 7.6 (Tree Star, Ashland, OR,

USA). This software provides values for the media fluorescence intensity (MFI) based on the fluorochromes in
each cell captured by flow cytometry. Therefore, the eGFP MFI of each sample can be used as a reporter of trans-
gene expression level. For EPO gene expression, the cells transfected with EPO gene were screened under G418
(800 μg/mL) selection for about 15 days and the stably transfected cells appeared. When cell colonies reached
90% confluence, the cells were harvested and then were seeded into 96-well plate with a limited dilution method
to produce the one cell/well. The cell colonies took up about 15% of the surface of the well when cells grew about
one week, and further transferred into 24- well, 6-well, gradualy. When the total cell number achieved 1 × 107,
the cells were cultured in protein-free, serum-free, chemically-defined CD CHO medium (Life Technologies #

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 7


www.nature.com/scientificreports/

Primer designation Type of analysis Primer sequence (5′-3′)


EGFP-Fwd qRT-PCR/qPCR 5′-CTACGTCCAGGAGCGCACCATCT-3′
EGFP-Rev qRT-PCR/qPCR 5′-GTTCTTCTGCTTGTCGGCCATGATAT-3′
GAPDH-Fwd qRT-PCR/qPCR 5′-CGACCCCTTCATTGACCTC-3′
GAPDH-Rev qRT-PCR/qPCR 5′-CTCCACGACATACTCAGCACC-3′

Table 1.  Primer used in the analysis of gene copy number and mRNA expression levels.

10743-029) supplemented with 8 mM L-glutamine (Life Technologies # 25030-024) in 125 mL Corning shake
flasks (Sigma # 431255) with 20 mL medium for about 5 days and the supernatant were collected for EPO expres-
sion level analysis.

LE
Analysis of long-term transgene expression stability.  CHO cells stably transfected with the vectors
containing human CMV, CMV mutant, CAG, CHEF-1α, CAG enhancer, mouse CMV, and HEF-1α were pas-
saged in pools and further cultured. The MFI for each vector type was measured by using the Guava EasyCyte
8HT flow cytometer, and the relative retention of eGFP expression for each vector was calculated as the ratio of

the MFI after generation 60 to that at the start of stability testing. Each ten passages, the expression level of eGFP

C
was determined by fluorescence intensity.

qPCR analysis of gene copy number.  Genomic DNA was isolated from 4 × 106 transfected CHO-S

TI
cells using a genomic DNA Mini Preparation kit with spin column (Beyotime). The gene copy numbers were
determined through qPCR technique using the eGFP and the GAPDH reference gene. Oligonucleotide primer
sequences are provided in Table 1. Copy numbers were determined in triplicate and are presented as ratios of
individual copy numbers relative to the control. Double-stranded DNA fragments were prepared from the oli-

R
gonucleotides by polymerization: a 10 μL reaction mix consisting of 4 μL template DNA (0.05 μg/μL), 5 μL SYBR
Green (TAKARA, Dalian, China), 0.2 μL of each forward and reverse oligonucleotide (10 μM each), and 0.6 μL
deionized water was subjected to heating at 95 °C for 3 min, 30 cycles of 94 °C for 30 s, 50 °C for 30 s, and elonga-

A
tion for 30 s at 72 °C, then 60 °C for 30 min for data acquisition.

Recombinant mRNA expression analysis.  After approximately 10 passages, the expression levels of
recombinant mRNA were analyzed. We isolated total RNA from cells transfected with each plasmid, and reverse
D
transcription reaction was performed with a HiFiScript first strand cDNA synthesis kit (CWBIO, Beijing, China)
according to the manufacturer’s protocol. qRT-PCR was undertaken using a PikoReal Software 2.2 Real-Time
PCR System (Thermo Scientific, Waltham, MA, USA). cDNA template (40 ng/reaction) was quantified by using

a Multiscan Spectrum spectrophotometer (SpectraMax i3x, Silicon Valley, CA, USA). PCR reactions were per-
TE

formed according to standard procedures using primer sets designed for the eGFP and GAPDH sequences
(Table 1). The mRNA expression level of the target gene was calculated by comparison with that of the internal
reference gene.

ELISA analysis
C

Cells transfected vectors were screened using G418 (800 μg/mL) after 48 h post-tranfection. Colonies arose after
10–14 days. Briefly, like the method demonstrated in stable expression of transfected cells and flow cytome-
try analysis. 30 stable clones were picked out and transferred to 24-well plates. Volumetric EPO production
A

(mg/L) for initial CHO-EPO cell lines cloned were scaled up to 125 mL Corning shake flasks. Cells were grown in
protein-free, serum-free, chemically-defined CD CHO medium supplemented with 8 mM L-glutamine in 125 mL
Corning shake flasks with 30 mL medium; at 60% density compared with 1.5 × 107. Cells supernatant were col-
R

lected for analysis by ELISA to determine volumetric EPO production as previously described47.

Western blot analysis.  The cells transfected with EPO-containing vector were suspended. When the
cell number reached 8 × 106/mL, the supernatant was collected and EPO was detected by immunoblotting.
ET

The supernatant containing EPO mixed with 5× SDS sample buffer was boiled. Ten microliters of sample were
subjected to electrophoresis on a 15% SDS–polyacrylamide gel and transferred to a nitrocellulose membrane
by electro-blotting. A 1:1500 dilution of an anti-EPO (EPO resistance protein) rabbit antiserum (Baoankang
Biotechnology Co., Ltd., Shenzhen, China) was incubated with the membrane followed by a secondary incuba-
tion with a 1:2000 dilution of goat anti-rabbit antibody conjugated to alkaline phosphatase (Jackson Immuno
R

Research Lab, West Grove, PA, USA). Densitometric analysis was performed by using ImageJ v2.1.4.7 software
(National Institutes of Health, Bethesda, MD, USA).

Statistical analysis.  All experimental data were analyzed by using SPSS 18.0 software (SPSS Inc., Chicago,
IL, USA). Data are reported as mean ± standard deviation. All experiments were performed three times and
t-tests were used for comparisons. Differences with P values <0.05 were considered statistically significant.

References
1. Hoban, M. D., Orkin, S. H. & Bauer, D. E. Genetic treatment of a molecular disorder: gene therapy approaches to sickle cell disease.
Blood. 18, 839–848 (2016).
2. Garcia-Gomez, M. et al. Safe and Efficient Gene Therapy for Pyruvate Kinase Deficiency. Mol. Ther. 24, 1187–1198 (2016).
3. Flynn, R. et al. CRISPR-mediated genotypic and phenotypic correction of a chronic granulomatous disease mutation in human iPS
cells. Exp. Hematol. 43, 838–848 (2015).

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 8


www.nature.com/scientificreports/

4. Wang, T. Y. et al. Cell compatibility of an eposimal vector mediated by the characteristic motifs of matrix attachment regions. Curr.
Gene Ther. (2016).
5. Hansen, H. G., Pristovšek, N., Kildegaard, H. F. & Lee, G. M. Improving the secretory capacity of Chinese hamster ovary cells by
ectopic expression of effector genes: lessons learned and future directions. Biotechnol. Adv. 35, 64–76 (2017).
6. Walsh, G. Biopharmaceutical benchmarks. Nat. Biotechnol. 32, 992–1000 (2014).
7. Wang, X. Y. et al. Impact of Different Promoters on Episomal Vectors Harbouring Characteristic Motifs of Matrix Attachment
Regions. Sci. Rep. 6, 26446 (2016).
8. Santilli, G. et al. Biochemical correction of X-CGD by a novel chimeric promoter regulating high levels of transgene expression in
myeloid cells. Mol. Ther. 19, 122–132 (2011).
9. Stein, S. et al. Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for
chronic granulomatous disease. Nat. Med. 16, 198–204 (2010).
10. Girod, P. A. et al. Genome-wide prediction of matrix attachment regions that increase gene expression in mammalian cells. Nat.
Methods. 4, 747–753 (2007).
11. Allen, M. L. & Antoniou, M. Correlation of DNA methylation with histone modifications across the HNRPA2B1-CBX3
Ubiquitously-Acting Chromatin Open Element (UCOE). Epigenetics. 2, 227–236 (2007).

LE
12. Brown, A. J., Sweeney, B., Mainwaring, D. O. & James, D. C. Synthetic promoters for CHO cell engineering. Biotechnol. Bioeng. 111,
1638–1647 (2014).
13. Kim, S. Y., Lee, J. H., Shin, H. S., Kang, H. J. & Kim, Y. S. The human elongation factor 1 alpha (EF-1 alpha) first intron highly
enhances expression of foreign genes from the murine cytomegalovirus promoter. J. Biotechnol. 93, 183–187 (2002).
14. Ho, S. C., Mariati, Yeo, J. H., Fang, S. G. & Yang, Y. Impact of using different promoters and matrix attachment regions on
recombinant protein expression level and stability in stably transfected CHO cells. Mol. Biotechnol. 57, 138–144 (2015).

C
15. Rotondaro, L., Mele, A. & Rovera, G. Efficiency of different viral promoters in directing gene expression in mammalian cells: effect
of 3′-untranslated sequences. Gene 168, 195–198 (1996).
16. Addison, C. L., Hitt, M., Kunsken, D. & Graham, F. L. Comparison of the human versus murine cytomegalovirus immediate early
gene promoters for transgene expression by adenoviral vectors. J. Gen. Virol. 78, 1653–1661 (1997).

TI
17. Rita Costa, A., Elisa Rodrigues, M., Henriques, M., Azeredo, J. & Oliveira, R. Guidelines to cell engineering for monoclonal antibody
production. Eur. J. Pharm. Biopharm. 74, 127–138 (2010).
18. Bailey, L. A., Hatton, D., Field, R. & Dickson, A. J. Determination of Chinese hamster ovary cell line stability and recombinant
antibody expression during long-term culture. Biotechnol. Bioeng. 109, 2093–2103 (2012).
19. Hsu, C. C. et al. Targeted methylation of CMV and E1A viral promoters. Biochem. Biophys. Res. Comm. 402, 228–234 (2010).

R
20. Deer, J. R. & Allison, D. S. High-level expression of proteins in mammalian cells using transcription regulatory sequences from the
Chinese hamster EF-1 alpha gene. Biotechnol. Progr. 20, 880–889 (2004).
21. Byun, H. M. et al. Plasmid vectors harboring cellular promoters can induce prolonged gene expression in hematopoietic and
mesenchymal progenitor cells. Biochem. Biophys. Res. Comm. 332, 518–523 (2005).

A
22. Chan, K. K., Wu, S. M., Nissom, P. M., Oh, S. K. & Choo, A. B. Generation of high-level stable transgene expressing human
embryonic stem cell lines using Chinese hamster elongation factor-1 alpha promoter system. Stem cells Dev. 17, 825–836 (2008).
23. Dighe, N. et al. Long-term reproducible expression in human fetal liver hematopoietic stem cells with a UCOE-based lentiviral
vector. PLoS One 9, e104805 (2014).
24. Farokhimanesh, S., Rahbarizadeh, F., Rasaee, M. J., Kamali, A. & Mashkani, B. Hybrid promoters directed tBid gene expression to
D
breast cancer cells by transcriptional targeting. Biotechnol. Prog. 26, 505–511 (2010).
25. Yew, N. S., Przybylska, M., Ziegler, R. J., Liu, D. & Cheng, S. H. High and sustained transgene expression in vivo from plasmid vectors
containing a hybrid ubiquitin promoter. Mol. Ther. 4, 75–82 (2001).
TE

26. Sladitschek, H. L. & Neveu, P. A. Bidirectional Promoter Engineering for Single Cell MicroRNA Sensors in Embryonic Stem Cells.
PLoS One 11, e0155177 (2016).
27. Seo, H. W. et al. Evaluation of combinatorial cis-regulatory elements for stable gene expression in chicken cells. BMC. Biotechnol.
19(10), 69 (2010).
28. Schlabach, M. R., Hu, J. K., Li, M. & Elledge, S. J. Synthetic design of strong promoters. Proc. Natl. Acad. Sci. USA 107, 2538–2543
(2010).
29. Estes, B. et al. Uncovering Methods for the Prevention of Protein Aggregation and Improvement of Product Quality in a Transient
C

Expression System. Biotechnol. Prog. 31, 258–267 (2015).


30. Pham, P. L., Kamen, A. & Durocher, Y. Large-scale transfection of mammalian cells for the fast production of recombinant protein.
Mol. Biotechnol. 34, 225–237 (2006).
31. Veith, N., Ziehr, H., MacLeod, R. A. & Reamon-Buettner, S. M. Mechanisms underlying epigenetic and transcriptional heterogeneity
A

in Chinese hamster ovary (CHO) cell lines. BMC. Biotechnol. 16, 6 (2016).
32. Lin, Y. et al. MAR characteristic motifs mediate episomal vector in CHO cells. Gene 559, 137–143 (2015).
33. Kelly, L. E., Martinez-De Luna, R. I. & El-Hodiri, H. M. Autoregulation of retinal homeobox (rax) gene promoter activity through a
R

highly conserved genomic element. Genesis. 54, 562–567 (2016).


34. Ford, K. L., Baumgartner, K., Henricot, B., Bailey, A. M. & Foster, G. D. A native promoter and inclusion of an intron is necessary for
efficient expression of GFP or mRFP in Armillaria mellea. Sci. Rep. 6, 29226 (2016).
35. Brown, A. J., Sweeney, B., Mainwaring, D. O. & James, D. C. NF-κB, CRE and YY1 elements are key functional regulators of CMV
ET

promoter-driven transient gene expression in CHO cells. Biotechnol. J. 10, 1019–1028 (2015).
36. Román, R. et al. Enhancing heterologous protein expression and secretion in HEK293 cells by means of combination of CMV
promoter and IFN2 signal peptide. Biotechnol. J. 10, 1019–1028 (2015).
37. Humby, M. S. & O’Connor, C. M. Human Cytomegalovirus US28 Is Important for Latent Infection of Hematopoietic Progenitor
Cells. J. Virol. 90, 2959–2970 (2015).
38 Moritz, B., Becker, P. B. & Göpfert, U. CMV promoter mutants with a reduced propensity to productivity loss in CHO cells. Sci. Rep.
R

5, 16952 (2015).
39. Montiel-Equihua, C. A. et al. The β-globin locus control region in combination with the EF1α short promoter allows enhanced
lentiviral vector-mediated erythroid gene expression with conserved multilineage activity. Mol. Ther. 20, 1400–1409 (2012).
40. Roodbari, F. et al. Immune responses against a new HIV-1p24-gp41/pCAGGS-IL-12 DNA vaccine in Balb/c mice. Iran. J. Immunol.
9, 86–97 (2012).
41. Yang, C. et al. DNMT 1 maintains hypermethylation of CAG promoter specific region and prevents expression of exogenous gene
in fat-1 transgenic sheep. PLoS One 12, e0171442 (2017).
42. Wippermann, A., Rupp, O., Brinkrolf, K., Hoffrogge, R. & Noll, T. The DNA methylation landscape of Chinese hamster ovary
(CHO) DP-12 cells. J. Biotechnol. 199, 38–46 (2015).
43. Tornøe, J., Kusk, P., Johansen, T. E. & Jensen, P. R. Generation of a synthetic mammalian promoter library by modification of
sequences spacing transcription factor binding sites. Gene 297, 21–32 (2002).
44. Magnusson, T., Haase, R., Schleef, M., Wagner, E. & Ogris, M. Sustained, high transgene expression in liver with plasmid vectors
using optimized promoter-enhancer combinations. J. Gene Med. 13, 382–391 (2011).

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 9


www.nature.com/scientificreports/

45. Ho, S. C. et al. IRES-mediated Tricistronic vectors for enhancing generation of high monoclonal antibody expressing CHO cell lines.
J.Biotechnol. 157, 130–139 (2012).
46. Kim, C. H., Oh, Y. & Lee, T. H. Codon optimization for high-level expression of human erythropoietin (EPO) in mammalian cells.
Gene. 199, 293–301 (1997).
47. Yang, M. & Butler, M. Effect of ammonia on the glycosylation of human recombinant erythropoietin in culture. Biotechnol. Prog. 16,
751–759 (2000).

Acknowledgements
This work was supported by the Grants from the National Natural Science Foundation of China (No. 81673337),
Plan for Scientific Innovation Talent of Henan Province, China (No. 164200510003)and the Key Scientific
Research Projects in Universities of Henan Province (No. 18A350008).

Author Contributions
Tian-Yun Wang designed, analyzed the experiments and revised the manuscript. Wen Wang and Yan-long Jia

LE
performed the experiments and wrote the manuscript. Yi-chun Li and Chang-qin Jing performed experiments for
stable expression of transfected cells. Xue-fang Shang performed experiments for Figure 5, interpreted these data
and edited the manuscript. Xiao Guo and Chun-peng Zhao performed the experiment for vector construction
and cultured cells.

Additional Information

C
Supplementary information accompanies this paper at doi:10.1038/s41598-017-10966-y
Competing Interests: The authors declare that they have no competing interests.

TI
Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional affiliations.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or

R
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Cre-
ative Commons license, and indicate if changes were made. The images or other third party material in this
article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the
A
material. If material is not included in the article’s Creative Commons license and your intended use is not per-
mitted 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 license, visit http://creativecommons.org/licenses/by/4.0/.
D
© The Author(s) 2017
TE
C
A
R
ET
R

Scientific Reports | 7:10416 | DOI:10.1038/s41598-017-10966-y 10

You might also like