ArticlePDF Available

Activation of stress-activated MAP protein kinases up-regulates expression of transgenes driven by the cytomegalovirus immediate/early promoter

Authors:

Abstract and Figures

The immediate/early promoter/enhancer of cytomegalovirus (CMV promoter) is one of the most commonly used promoters for expression of transgenes in eukaryotic cells. In practice, the CMV promoter is often thought of as a constitutively active unregulated promoter. However, we have observed that transcription from the CMV promoter can be up-regulated by a variety of environmental stresses. Many forms of cellular stress stimulate MAP kinase signalling pathways, resulting in activation of stress-activated protein kinases [SAPKs, also called Jun N-terminal kinases (JNKs)] and p38 kinases. We have found that the same conditions that lead to activation of SAPK/JNKs and p38 kinases can also dramatically increase expression from the CMV promoter. Inhibitors of p38 kinases abolished basal transcription from the CMV promoter and completely blocked stress-induced up-regulation of the CMV promoter. Overexpression of a dominant negative JNK kinase had no effect on basal transcription, but significantly reduced up-regulation caused by stress. These results have grave implications for use of the CMV promoter. If the CMV promoter can be upregulated by cellular stresses, inadvertent activation of the stress kinase pathways may complicate, if not invalidate, the interpretation of a wide range of experiments.
Content may be subject to copyright.
1998 Oxford University Press
486–489 Nucleic Acids Research, 1998, Vol. 26, No. 2
Activation of stress-activated MAP protein kinases
up-regulates expression of transgenes driven by the
cytomegalovirus immediate/early promoter
W. Bruening+, B. Giasson1, W. Mushynski1 and H. D. Durham*
Department of Neurotoxicology, Montreal Neurological Institute and Department of Neurology/Neurosurgery,
3801 University Street, Montreal, Quebec H3A 2B4, Canada and 1Department of Biochemistry, McGill University,
Montreal, Quebec, Canada
Received September 25, 1997; Revised and Accepted November 20, 1997
ABSTRACT
The immediate/early promoter/enhancer of cytomega-
lovirus (CMV promoter) is one of the most commonly
used promoters for expression of transgenes in
eukaryotic cells. In practice, the CMV promoter is often
thought of as a constitutively active unregulated
promoter. However, we have observed that transcription
from the CMV promoter can be up-regulated by a variety
of environmental stresses. Many forms of cellular
stress stimulate MAP kinase signalling pathways,
resulting in activation of stress-activated protein
kinases [SAPKs, also called Jun N-terminal kinases
(JNKs)] and p38 kinases. We have found that the same
conditions that lead to activation of SAPK/JNKs and
p38 kinases can also dramatically increase expression
from the CMV promoter. Inhibitors of p38 kinases
abolished basal transcription from the CMV promoter
and completely blocked stress-induced up-regulation of
the CMV promoter. Overexpression of a dominant
negative JNK kinase had no effect on basal transcription,
but significantly reduced up-regulation caused by
stress. These results have grave implications for use of
the CMV promoter. If the CMV promoter can be up-
regulated by cellular stresses, inadvertent activation of
the stress kinase pathways may complicate, if not
invalidate, the interpretation of a wide range of
experiments.
INTRODUCTION
It has recently been discovered that a variety of environmental
insults, such as UV irradation, osmotic shock and oxidative stress,
mediate some of their effects on the cell through activation of
MAP kinase signalling pathways (1,2). These kinase cascades
ultimately activate the c-Jun N-terminal kinases (JNKs), also
called stress-activated protein kinases (SAPKs, here referred to as
SAPK/JNKs), a family of kinases which increase the activity of
transcription factors such as c-Jun and ATF-2 (3,4). A parallel
pathway which responds to many of the same stimuli results in
activation of the p38 MAP kinases (5), a family of kinases that
increase the activity of MAPKAP kinase 2 (6), ATF-2 (7) and
Max (8).
During the course of our work we observed that many of the
same stresses that activate the SAPK/JNK pathway could also
dramatically increase expression of transgenes regulated by the
immediate/early cytomegalovirus promoter/enhancer complex
(CMV promoter). This is one of the most commonly used
promoters in eukaryotic expression vectors. Its popularity derives
from its ability to drive high levels of expression in nearly all
mammalian cells. If the CMV promoter can be up-regulated by
stress, its use may complicate a wide range of experiments,
particularly those involving components of the stress-activated
kinase pathways.
MATERIALS AND METHODS
Cell culture
All cell lines were originally from ATCC. Cell lines were maintained
in DMEM supplemented with 10% fetal calf serum. Cells were
transfected with Lipofectamine (Gibco-BRL, Burlington, Ontario)
following the manufacturer’s instructions. Equal amounts of
expression vector were added in each transfection; when necessary
an empty CMV expression vector (pcDNA-3; Invitrogen,
San Diego, CA) was added to equalize the total amount of plasmid
DNA. Under these conditions 80% of NIH 3T3 cells are
transfected.
β-Galactosidase activity assays
Forty-eight hours after transfection cells were lysed in PBS, 0.5%
Nonidet P-40. Insoluble debris was pelleted and the supernatant
diluted 10-fold with Z buffer [140 mM sodium phosphate, pH 7.4,
1 mM magnesium sulfate, 140 mM potassium chloride, 0.27%
β-mercaptoethanol, 1 mg/ml o-nitrophenyl-β-D-galactopyranoside
(ONPG)]. After incubation at 37C for 30 min, the absorbance at
420 nm was measured with an LKB Ultrospec 4050
*To whom correspondence should be addressed. Tel: +1 514 398 8509; Fax: +1 514 398 1509; Email: mddm@musica.mcgill.ca
+Present address: FoxChase Cancer Center, Philadelphia, PA 19111, USA
487
Nucleic Acids Research, 1994, Vol. 22, No. 1
Nucleic Acids Research, 1998, Vol. 26, No. 2 487
Figure 1. NIH 3T3 cells exposed to arsenite have elevated levels of heat
shock/stress proteins. Western blots were probed with an antibody that
recognizes only the inducible form of HSP70 (K20; Santa Cruz Biotechnology)
and an antibody against HSP27 (M20; Santa Cruz Biotechnology). Lane 1,
untreated control; lane 2, 1 µM arsenite; lane 3, 10 µM arsenite; lane 4, 50 µM
arsenite. Cells were treated with sodium arsenite for 6 h before harvesting and
blotting as described in Materials and Methods.
spectrophotometer. Protein concentration of the extracts was
measured with a bicichoninic acid–copper assay (Sigma, St Louis,
MO). β-Galactosidase activity is defined as (OD420 nm × 1000)
(time of incubation)(mg extract). All assays were done in duplicate.
Immunoblots
Cells were lysed in RIPA buffer (50 mM Tris, pH 8.0, 150 mM
NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS).
Insoluble debris was pelleted and the protein concentration of the
supernatant determined using a bichichoninic acid–copper assay
(BioRad, Mississauga, Ontario). Samples of 20 µg were
electrophoresed by standard SDS–PAGE and transferred to
Immobilon-P membranes (Millipore). Blots were probed with the
indicated antibodies using standard techniques and developed with
ECL chemiluminescence reagents (Amersham).
Northern blots
Forty-eight hours after transfection, RNA was extracted from the
cells with Trizol (Gibco-BRL), following the manufacturer’s
instructions. The RNA was quantified by measuring optical density
at 260 nm and the values were confirmed by ethidium staining after
gel electrophoresis. Five micrograms of RNA were separated on a
MOPS/formaldehyde–agarose gel and transferred to Zetabind
membrane (BioRad) by capillary blotting. The blot was probed with
full-length lacZ cDNA using standard techniques.
Immunoprecipitation kinase assay of JNK1
Activation of JNK1 was assayed as described previously (9).
Briefly, following cell lysis and centrifugation to remove cell
debris the enzyme was immunoprecipitated using anti-JNK1
polyclonal antibody (C-17; Santa Cruz Biotechnology, Santa
Cruz, CA) and GST–cJun (Santa Cruz Biotechnology) was used
as substrate to determine the activity.
Plasmids and reagents
Unless otherwise stated, chemicals were purchased from Sigma
and cell culture reagents were from Gibco-BRL. The p38
inhibitor SB203580 was from Calbiochem. The pCMV-lacZ
plasmid was pUT535 (Cayla, Toulouse, France). Plasmid
pJNKK(–) encodes a JNK kinase (also called SEK1) mutated at
the sites of MEKK1 phosphorylation, such that activated
Figure 2. Activation of stress-activated MAP kinases up-regulates expression
from the CMV promoter. (A) NIH 3T3 cells were transfected with pCMV-lacZ as
described in Materials and Methods. Forty hours after transfection some of the cells
were treated with 50 µM sodium arsenite for 6 h. (B) The indicated cell lines were
transfected with pCMV-lacZ or co-transfected with pCMV-lacZ and pCMV-MEK-
K(Act). Forty-six hours after transfection the cells were harvested and assayed for
β-galactosidase activity as described in Materials and Methods. Each experiment
was done three to five times, in duplicate, and the average was taken.
MEKK1 can bind to the sites but not phosphorylate them, thus
preventing some of the activated MEKK1 from activating
endogenous JNKK. Plasmid pCMV-MEKK(Act) expresses a
constitutively active truncated form of MEKK1 and was a gift
from M.Karin.
RESULTS
Transcription from the CMV promoter can be up-regulated
While performing routine transfection experiments with expression
vectors containing the CMV promoter, we observed that certain
stressors would dramatically increase expression from these
constructs. To study this phenomenon further we employed a
plasmid in which expression of the Escherichia coli gene lacZ,
which encodes β-galactosidase, is regulated by the CMV promoter.
This system allowed us to readily quantify the amount of
CMV-driven expression obtained.
NIH 3T3 cells were transfected with the CMV-lacZ plasmid as
described in Materials and Methods. Forty hours after transfection
the cells were treated with 50 µM sodium arsenite. Six hours were
left between initiation of the treatment and harvesting to allow time
for transcription and translation. The concentration of arsenite used
was chosen to be sublethal but extremely stressful. Under these
conditions of exposure to arsenite induction of a stress response was
also demonstrated by increased levels of two heat shock/stress
proteins, HSP27 and HSP70 (Fig. 1). As shown in Figure 2A,
treatment of transfected NIH 3T3 cells with sodium arsenite, a
reagent which activates SAPK/JNK (10), increased β-galactosidase
activity 3-fold. Other stressors, such as osmotic shock, hydrogen
peroxide and heat shock, were also able to increase β-galactosidase
activity (data not shown).
To confirm that the observed CMV up-regulation was a direct
consequence of stress kinase activation, rather than some
side-effect of drug treatment, we employed a plasmid that
expresses a cDNA encoding an active MEKK1, one of the initial
kinases in the MAP kinase cascade. Low levels of active MEKK1
will preferentially activate SAPK/JNKs, but high levels of
expression, such as are found here, will also activate the p38
kinases (2,11). Co-transfection of NIH 3T3 cells with pCMV-lacZ
Nucleic Acids Research, 1998, Vol. 26, No. 2
488
Figure 3. Stress-induced up-regulation of the CMV promoter occurs at the level
of transcription. NIH 3T3 cells were transfected with pCMV-lacZ or
co-transfected with pCMV-lacZ and pCMV-MEKK(Act). Some cells were
treated with 50 µM sodium arsenite 40 h after transfection for 6 h. The cells
were harvested 46 h after transfection. mRNA was extracted, separated by gel
electrophoresis and blotted as described in Materials and Methods. The relative
amount of β-galactosidase mRNA was measured by phosphorimaging. Lane 1,
mock transfection; lane 2, transfected only with pCMV-lacZ; lane 3, transfected
only with pCMV-lacZ and treated with sodium arsenite; lane 4, transfected with
pCMV-lacZ and pCMV-MEKK(Act). The experiment was repeated several
times and a representative result is shown.
and pCMV-MEKK(Act) resulted in dramatically increased β-ga-
lactosidase activity (Fig. 2A). To see if this effect was cell type
specific a number of different cell lines were tested. Each cell line
was co-transfected with pCMV-lacZ and pCMV-MEKK(Act).
Forty-eight hours later, β-galactosidase levels were measured.
For all cell types tested co-expression of activated MEKK1
caused a significant increase in β-galactosidase levels (Fig. 2B).
The inductive effect was not limited to β-galactosidase, as other
proteins expressed from the CMV promoter were also found to be
up-regulated by stressors (data not shown).
To confirm that up-regulation was at the level of transcription
NIH 3T3 cells were transfected with pCMV-lacZ and treated with
sodium arsenite or co-transfected with pCMV-MEKK(Act).
Levels of β-galactosidase mRNA were examined by Northern
blotting. As can be seen in Figure 3, both sodium arsenite
treatment and co-expression of activated MEKK1 up-regulated
the amount of mRNA driven by the CMV promoter.
Both SAPK/JNKs and p38 kinases are involved in
regulating transcription from the CMV promoter
As mentioned briefly, the stressors that induce CMV up-regulation
are known to activate the SAPK/JNK pathways. To confirm that
SAPK/JNK activation correlates with CMV up-regulation we
assayed SAPK/JNK activity directly. As shown in Figure 4, treating
NIH 3T3 cells with either arsenite or transfection with pCMV-MEK-
K(Act) strongly increases the kinase activity of JNK1. Although
MEKK1 does not activate JNK1 as strongly as does arsenite,
MEKK1 up-regulates expression from the CMV promoter to a
much greater extent than does arsenite (Fig. 1A). However, this
apparent contradiction can easily be explained if we consider that
cells transfected with pCMV-MEKK(Act) are stimulated for 46 h,
allowing a steady accumulation of products expressed from the
CMV promoter, while arsenite treatment only allows 6 h worth
of products to accumulate.
To further investigate the role of SAPK/JNK in CMV
up-regulation we employed a dominant negative JNK kinase. The
Figure 4. JNK1 is activated by the same conditions that up-regulate CMV
expression. NIH 3T3 cells were transfected with pCMV-MEKK(Act) (A, lane 2),
mock transfected (A, lane 1), untreated (B, lane 1) or treated with sodium arsenite
as described in the legend to Figure 1 (B, lane 2). The cells were harvested and
assayed for JNK1 activity as described in Materials and Methods. The relative
JNK1 activity was measured by phosphorimaging and is indicated under each lane.
The experiment was repeated several times and a representative result is shown.
plasmid pJNKK(–) expresses a JNKK (also called SEK1)
mutated such that it partially blocks activation of SAPK/JNKs.
When pCMV-lacZ and pJNKK(–) were co-expressed together in
NIH 3T3 cells basal levels of β-galactosidase were unaltered.
However, when cells co-transfected with pCMV-lacZ and
pJNKK(–) were treated with sodium arsenite the presence of
JNKK(–) reduced up-regulation of β-galactosidase activity by
half (Fig. 5). Since p38 kinases and SAPK/JNKs are both
activated by the same or similar stresses (2), we also investigated
the role of p38 kinases in CMV stress-induced up-regulation. To
study their function we used a specific inhibitor that blocks
activity of the p38 kinases (12). We found that if p38 inhibitor was
added at the time of transfection almost no product was expressed
from the CMV promoter (Fig. 5), indicating that basal levels of
p38 kinase activity are essential for basal CMVpromoter activity.
If, however, p38 inhibitor was added 40 h after transfection, at the
same time sodium arsenite was added, the presence of p38
inhibitor completely blocked stress-induced up-regulation of the
CMV promoter (Fig. 5).
DISCUSSION
Activation of the stress-activated MAP kinase cascades has many
effects on the cell that appear to be mediated primarily through
regulation of transcription. Activated SAPK/JNK is known to
phosphorylate and thereby activate key transcriptional factors such
as c-Jun and ATF-2 (3,4) and the p38 kinases also regulate ATF-2
(7). The immediate/early CMV promoter/enhancer complex
contains AP-1 and ATF binding sites (13). It is, therefore, not
particularly surprising that activity of the CMV promoter should
be altered in a stressed cell. The stress-induced activation of the
CMV promoter that we have observed may simply be a
side-effect caused by a general up-regulation of transcription, but
it is also possible that cytomegalovirus could have evolved to take
advantage of the stress response. Like many other viruses,
cytomegalovirus evades the immune system by remaining
quiescent within the cell until conditions become appropriate for
viral replication (14). Stimulation of cytomegalovirus replication
appears to be primarily mediated by inducing transcription from
the cytomegalovirus immediate/early promoter (15) and many of
the conditions known to stimulate CMV replication, such as
oxidative stress, immunosuppressive drugs and irradiation, are
also conditions that can activate SAPK/JNKs and p38 kinases
(14,13,16). These conditions are also likely to result in
immunosuppression of the host, allowing cytomegalovirus to
replicate only under conditions which it is able to survive. Thus
489
Nucleic Acids Research, 1994, Vol. 22, No. 1
Nucleic Acids Research, 1998, Vol. 26, No. 2 489
Figure 5. MAP kinases regulate expression from the CMV promoter. NIH 3T3
cells were mock transfected (Mock), transfected with pCMV-lacZ alone (βGal),
co-transfected with pCMV-lacZ and pJNKK(–) [βGal + JNKK(–)] or transfected
with pCMV-lacZ and treated with 0.5 µM SB203580 (βGal + p38-I). Forty hours
after transfection some of the cells were treated with arsenite as described in the
legend to Figure 1. Some cells were treated with SB203580 for the entire 46 h
(added at t = 0), while others were only treated with SB203580 40 h after
transfection for 6 h (added at t = 40). All cells were harvested 46 h after transfection
and assayed for β-galactosidase activity as described in Materials and Methods.
Each experiment was done three times in duplicate and the average was taken.
viral promoters may have been selected to be able to maximize
transcription under stressed conditions.
In addition to variation in transcriptional activity of the CMV
promoter according to cell type and developmental age (17), two
previous reports have suggested that the CMV promoter can be
up-regulated under specific conditions. In one the major immediate/
early enhancer of cytomegalovirus was found to be up-regulated by
cAMP in lymphoid cell lines (18). This up-regulation was mediated
through a CRE element contained within the enhancer. In the other
report NF-κB was shown to play a central role in activation of the
CMV promoter by cytomegalovirus gene products (13). Sambucetti
et al. were also able to artificially activate transcription from the
CMV promoter by stimulating the cells with phorbol esters (13).
Our results are the first to show that stress-activated MAP kinases
are involved in regulating transcription from the CMV promoter. We
find that without basal p38 kinase activity the CMV promoter is
transcriptionally silent (Fig. 4). This may be mediated by lower
ATF-2 activity, as p38 kinases have been shown to directly stimulate
ATF-2 activity (7), or it may be mediated indirectly through loss of
NF-κB function. Wesselborg et al. were able to demonstrate that p38
kinases are necessary for NF-κB-dependent gene expression (19).
p38 kinase inhibitors were also able to completely block stress-
induced up-regulation of the CMV promoter (Fig. 4). It is unclear
whether this blockage is simply due to complete shut-off of the
CMV promoter in the absence of p38 kinases or whether p38 kinases
are also essential for stress-induction of the CMV promoter.
It is unlikely that NF-κB activity is involved in stress induction
of the CMV promoter. Although some stresses are able to activate
NF-κB, sodium arsenite does not (19). Thus the observed
up-regulation of the CMV promoter is probably mediated by
increased activity of c-Jun and ATF-2.
The pertinence of the data presented in this paper stems from the
widespread use of the CMV promoter in expression vectors.
Experimental protocols that might activate the stress response could
be complicated to interprete due to up-regulation of the CMV
promoter. For example, the pCMV-MEKK(Act) plasmid used in our
experiments sets up a self-stimulating loop, expressing increasing
amounts of active MEKK1 until the cell dies. Experiments which
use inhibitors of p38 kinases would be devastated by simultaneous
use of the CMV promoter. On the other hand, protocols that only
require high levels of expression could exploit stress activation of the
CMV promoter to generate even more transgenic protein and
perhaps p38 kinase inhibitors could be used to switch the CMV
promoter on and off.
ACKNOWLEDGEMENTS
This work was supported by grants from the ALS Society of
Canada, the Muscular Dystrophy Association of Canada and the
Medical Research Council of Canada. H.D.D. is an MNI Killam
Scholar. We thank M.Karin for his kind gifts of expression vectors.
REFERENCES
1 Devary,Y., Gottlieb,R.A., Smeal,T. and Karin,M. (1992) Cell, 71, 1081–1091.
2 Kyriakis,J.M. and Avruch,J. (1996) J. Biol. Chem., 271, 24313–24316.
3 Morgan,I.M., Asano,M., Havarstein,L.S., Ishikawa,H., Hiiragi,T., Ito,Y.
and Vogt,P.K. (1993) Oncogene, 8, 1135–1140.
4 Smeal,T., Binetruy,B., Mercola,D.A., Birrer,M. and Karin,M. (1991) Nature,
354, 494–496.
5 Han,J., Lee,J.D., Bibbs,L. and Ulevitch,R.J. (1994) Science, 265, 808–811.
6 Freshney,N.W., Rawlinson,L., Guesdon,F., Jones,E., Cowley,S., Hsuan,J.
and Saklatvala,J. (1994) Cell, 78, 1039–1049.
7 Derijard,B., Raingeaud,J., Barrett,T., Wu,I.H., Han,J., Ulevitch,R.J. and
Davis,R.J. (1995) Science, 267, 682–685.
8 Zervos,A.S., Faccio,L., Kyriakis,J.M. and Brent,R. (1995) Proc. Natl.
Acad. Sci. USA, 92, 10531–10534.
9 Giasson,B. and Mushynski,W. (1996) J. Biol. Chem., 271, 30404–30409.
10 Meier,R., Rouse,J., Cuenda,A., Nebreda,A.R. and Cohen,P. (1996) Eur. J.
Biochem., 236, 796–805.
11 Yan,M., Dai,T., Deak,J.C., Kyriakis,J.M., Zon,L.I., Woodgett,J.R. and
Templeton,D.J. (1994) Nature, 372, 798–800.
12 Cuenda,A., Rouse,J., Doza,Y.N., Meier,R., Cohen,P., Gallagher,T.F.,
Young,P.R. and Lee,J.C. (1995) FEBS Lett., 364, 229–233.
13 Sambucetti,L., Cherrington,J.M., Wilkinson,G.W.G. and Mocarski,E.S.
(1989) EMBO J., 8, 4251–4258.
14 Bruggeman,C.A., Li,F. and Stals,F.S. (1995) Scand. J. Infect. Dis., 99 (suppl.),
43–50.
15 Fish,K.N., Stenglein,S.G., Ibanez,C. and Nelson,J.A. (1995) Scand. J. Infect.
Dis., 99 (suppl.), 34–40.
16 Scholz,M., Cinatl,J., Gross,V., Vogel,J.U., Blaheta,R.A., Freisleber,H.J.,
Markus,B.H. and Doerr,H.W. (1996) Transplantation, 61, 1763–1770.
17 Baskar,J.F., Smith,P.P., Ciment,G.S., Hoffmann,S., Tucker,C., Tenney,D.J.,
Colberg-Poley,A.M., Nelson,J.A. and Ghazal,P. (1996) J. Virol., 70,
3215–3226.
18 Stamminger,T., Fickenscher,H. and Fleckenstein,B. (1990) J. Gen. Virol., 71,
105–113.
19 Wesselborg,S., Bauer,M.K.A., Vogt,M., Schmitz,M.L. and Schulze-Osthoff,K.
(1997) J. Biol. Chem., 272, 12422–12429.
... Consistent with our observations, Watson-Levings et al. also reported approximately four fold higher IL-1ra levels in PTOA joints (214 ng/ml) compared to healthy non-PTOA joints (59 ng/ ml) [39]. The cytomegalovirus (CMV) promoter, which drives the expression of the scAAV cassette, is known to be activated by inflammatory mediators and stress-activated chemokines [49][50][51][52]. As PTOA-cartilage is enriched in stress-activated chondrocytes, particularly at the site of injury, we postulate that the drastic upregulation in transgene expression that we observed in an injured joint is a result of inflammation-induced activation of the CMV promoter in response to mediators in the diseased joint environment. ...
Article
Full-text available
Gene therapy approaches using adeno-associated viral vectors have been successfully tested in the equine post-traumatic osteoarthritis (PTOA) model. Owing to differences in the levels of transgene expression and adverse tissue reactions observed in published studies, we sought to identify a safe therapeutic dose of scAAVIL-1ra in an inflamed and injured joint that would result in improved functional outcomes without any adverse events. scAAVIL-1ra was delivered intra-articularly over a 100-fold range, and horses were evaluated throughout and at the end of the 10-week study. A dose-related increase in IL-1ra levels with a decrease in PGE2 levels was observed, with the peak IL-1ra concentration being observed 7 days post-treatment in all groups. Perivascular infiltration with mononuclear cells was observed within the synovial membrane of the joint treated with the highest viral dose of 5 × 10¹² vg, but this was absent in the lower-dosed joints. The second-highest dose of scAAVeqIL-1ra 5 × 10¹¹ vg demonstrated elevated IL-1ra levels without any cellular response in the synovium. Taken together, the data suggest that the 10-fold lower dose of 5 × 10¹¹vg scAAVIL-1ra would be a safe therapeutic dose in an equine model of PTOA.
... We speculate that the increase in HERV-W LQMV Env cell surface expression via transfection in HEK293 cells and transduced murine BMDCs, as well as the increase in the expression levels of both Env and Gag of transduced A549 cells, could be attributed to changes in protein homeostasis, such as increased production, increased surface accumulation, and decreased breakdown. Alternatively, cell stress can induce increased transactivation of the CMV promoter through which the expression of the HERV-W antigens is determined [30]. The increased CMV promoter activation could increase antigen transcription if the HERV-W LQMV mutant induced more cell stress than the HERV-W WT . ...
Article
Full-text available
Human endogenous retrovirus type W (HERV-W) is expressed in various cancers. We previously developed an adenovirus-vectored cancer vaccine targeting HERV-W by encoding an assembled HERV-W group-specific antigen sequence and the HERV-W envelope sequence Syncytin-1. Syncytin-1 is constitutively fusogenic and forms large multinucleated cell fusions when overexpressed. Consequently, immunising humans with a vaccine encoding Syncytin-1 can lead to the formation of extensive syncytia, which is undesirable and poses a potential safety issue. Here, we show experiments in cell lines that restoring an evolutionary lost cleavage site of the fusion inhibitory R-peptide of Syncytin-1 inhibit cell fusion. Interestingly, this modification of the HERV-W vaccine’s fusogenicity increased the expression of the vaccine antigens in vitro. It also enhanced Syncytin-1-specific antibody responses and CD8+-mediated T-cell responses compared to the wildtype vaccine in vaccinated mice, with a notable enhancement in responses to subdominant T-cell epitopes but equal responses to dominant epitopes and similar rates of survival following a tumour challenge. The impairment of cell–cell fusion and the enhanced immunogenicity profile of this HERV-W vaccine strengthens the prospects of obtaining a meaningful immune response against HERV-W in patients with HERV-W-overexpressing cancers.
... If the promoter size is a concern, the RSV and SV40 promoters, which are shorter in length, are typically preferred. Despite providing high transgene expression levels, the CMV promoter is prone to cell linedependent methylation-induced gene silencing, presents cell-cycle dependency, and variable transgene expression levels (Bruening et al., 1998;Hsu et al., 2010;M. Kim et al., 2011). ...
Article
Full-text available
The production of high‐quality recombinant proteins is critical to maintaining a continuous supply of biopharmaceuticals, such as therapeutic antibodies. Engineering mammalian cell factories presents a number of limitations typically associated with the proteotoxic stress induced upon aberrant accumulation of off‐pathway protein folding intermediates, which eventually culminate in the induction of apoptosis. In this review, we will discuss advances in cell engineering and their applications at different hierarchical levels of control of the expression of recombinant proteins, from transcription and translational to posttranslational modifications and subcellular trafficking. We also highlight challenges and unique opportunities to apply modern synthetic biology tools to the design of programmable cell factories for improved biomanufacturing of therapeutic proteins.
... Upon phosphorylation, CREB seems to be able to recruit CBP as a coactivator, stabilizing the pre-initiation complex formed at the promoter, resulting in an even higher transcription increase [33]. Several groups showed elevated CMV activation by the cAMP signaling pathway and the stress-activated MAP protein kinase pathway [34,35]. However, the transcription quantification of CMV mediated by CREB and phosphorylated CREB was unknown so far. ...
Article
Full-text available
In today’s age, plastic waste is a major problem for our environment. The decomposition of plastic waste causes widespread contamination in all types of ecosystems worldwide. Micro-plastics in the lower micrometer size range and especially nano-plastics can become internalized by cells and thus become a threat to human health. To investigate the effects of internalized micro- and nano-plastics on human gene transcription, we used an in vitro assay to quantify CREB (cAMP response element binding protein) mediated transcription. Here we show that CREB mediated gene expression was mainly but not exclusively induced by phosphorylation. In addition, the amount of CREB affected transcription was also studied. We were also able to show that the strong CREB mediated stimulation of transcription was diminished by micro- and nano-plastics in any chosen setting. This indicates a threat to human health via the deregulation of transcription induced by internalized micro- and nano-plastics. However, this established quantifiable in vitro transcription test system could help to screen for toxic substances and non-toxic alternatives.
... Interestingly, the vector activity appeared to be significantly higher in the joints with OA pathologies compared to healthy joints as demonstrated by a concentration of GFP fluorescence in the areas of articular cartilage with distinct damage. The investigators postulate that the increase in transgene expression could be a result of proliferation and increased metabolism of the diseased chondrocytes as well as stress induced activation of the cytomegalovirus (CMV) promoter which is normally activated by stress-activated protein kinases (173)(174)(175). These observations present a unique opportunity to target transgene expression to specific diseased regions of the joint and direct site-specific repair. ...
Article
Full-text available
With an intrinsically low ability for self-repair, articular cartilage injuries often progress to cartilage loss and joint degeneration resulting in osteoarthritis (OA). Osteoarthritis and the associated articular cartilage changes can be debilitating, resulting in lameness and functional disability both in human and equine patients. While articular cartilage damage plays a central role in the pathogenesis of OA, the contribution of other joint tissues to the pathogenesis of OA has increasingly been recognized thus prompting a whole organ approach for therapeutic strategies. Gene therapy methods have generated significant interest in OA therapy in recent years. These utilize viral or non-viral vectors to deliver therapeutic molecules directly into the joint space with the goal of reprogramming the cells' machinery to secrete high levels of the target protein at the site of injection. Several viral vector-based approaches have demonstrated successful gene transfer with persistent therapeutic levels of transgene expression in the equine joint. As an experimental model, horses represent the pathology of human OA more accurately compared to other animal models. The anatomical and biomechanical similarities between equine and human joints also allow for the use of similar imaging and diagnostic methods as used in humans. In addition, horses experience naturally occurring OA and undergo similar therapies as human patients and, therefore, are a clinically relevant patient population. Thus, further studies utilizing this equine model would not only help advance the field of human OA therapy but also benefit the clinical equine patients with naturally occurring joint disease. In this review, we discuss the advancements in gene therapeutic approaches for the treatment of OA with the horse as a relevant patient population as well as an effective and commonly utilized species as a translational model.
... Thus, CYGB mRNA and protein induction upon pPBS treatment in CYGB overexpressing A375-hCYGB cells is likely of no biological importance. The CMV promoter has been shown to be induced upon cellular stresses, including oxidative stress [83,84]. ...
Article
Full-text available
Melanoma arises from pigment-producing cells called melanocytes located in the basal layers of the epidermis of the skin. Cytoglobin (CYGB) is a ubiquitously expressed hexacoordinated globin that is highly enriched in melanocytes and frequently downregulated during melanomagenesis. Previously, we showed that non-thermal plasma (NTP)-produced reactive oxygen and nitrogen species (RONS) lead to the formation of an intramolecular disulfide bridge that would allow CYGB to function as a redox-sensitive protein. Here, we investigate the cytotoxic effect of indirect NTP treatment in two melanoma cell lines with divergent endogenous CYGB expression levels, and we explore the role of CYGB in determining treatment outcome. Our findings are consistent with previous studies supporting that NTP cytotoxicity is mediated through the production of RONS and leads to apoptotic cell death in melanoma cells. Furthermore, we show that NTP-treated solutions elicit an antioxidant response through the activation of nuclear factor erythroid 2–related factor 2 (NRF2). The knockdown and overexpression of CYGB respectively sensitizes and protects melanoma cells from RONS-induced apoptotic cell death. The presence of CYGB enhances heme-oxygenase 1 (HO-1) and NRF2 protein expression levels, whereas the absence impairs their expression. Moreover, analysis of the CYGB-dependent transcriptome demonstrates the tumor suppressor long non-coding RNA maternally expressed 3 (MEG3) as a hitherto undescribed link between CYGB and NRF2. Thus, the presence of CYGB, at least in melanoma cells, seems to play a central role in determining the therapeutic outcome of RONS-inducing anticancer therapies, like NTP-treated solutions, possessing both tumor-suppressive and oncogenic features. Hence, CYGB expression could be of interest either as a biomarker or as a candidate for future targeted therapies in melanoma.
... In our transgenic animals, the human HSPB1 gene expression is driven by a cytomegalovirus (CMV) promoter. It is usually referred to as a constitutively active promoter; however, it has been shown to be upregulated in vitro under certain stress or inflammatory conditions [40][41][42]. Accordingly, increased transgene expression was observed in response to ethanol treatment in different brain regions of our transgenic mice and in the primary cells as well. ...
Article
Full-text available
Background Heat-shock protein B1 (HSPB1) is among the most well-known and versatile member of the evolutionarily conserved family of small heat-shock proteins. It has been implicated to serve a neuroprotective role against various neurological disorders via its modulatory activity on inflammation, yet its exact role in neuroinflammation is poorly understood. In order to shed light on the exact mechanism of inflammation modulation by HSPB1, we investigated the effect of HSPB1 on neuroinflammatory processes in an in vivo and in vitro model of acute brain injury. Methods In this study, we used a transgenic mouse strain overexpressing the human HSPB1 protein. In the in vivo experiments, 7-day-old transgenic and wild-type mice were treated with ethanol. Apoptotic cells were detected using TUNEL assay. The mRNA and protein levels of cytokines and glial cell markers were examined using RT-PCR and immunohistochemistry in the brain. We also established primary neuronal, astrocyte, and microglial cultures which were subjected to cytokine and ethanol treatments. TNFα and hHSPB1 levels were measured from the supernates by ELISA, and intracellular hHSPB1 expression was analyzed using fluorescent immunohistochemistry. Results Following ethanol treatment, the brains of hHSPB1-overexpressing mice showed a significantly higher mRNA level of pro-inflammatory cytokines ( Tnf , Il1b ), microglia ( Cd68 , Arg1 ), and astrocyte ( Gfap ) markers compared to wild-type brains. Microglial activation, and 1 week later, reactive astrogliosis was higher in certain brain areas of ethanol-treated transgenic mice compared to those of wild-types. Despite the remarkably high expression of pro-apoptotic Tnf , hHSPB1-overexpressing mice did not exhibit higher level of apoptosis. Our data suggest that intracellular hHSPB1, showing the highest level in primary astrocytes, was responsible for the inflammation-regulating effects. Microglia cells were the main source of TNFα in our model. Microglia isolated from hHSPB1-overexpressing mice showed a significantly higher release of TNFα compared to wild-type cells under inflammatory conditions. Conclusions Our work provides novel in vivo evidence that hHSPB1 overexpression has a regulating effect on acute neuroinflammation by intensifying the expression of pro-inflammatory cytokines and enhancing glial cell activation, but not increasing neuronal apoptosis. These results suggest that hHSPB1 may play a complex role in the modulation of the ethanol-induced neuroinflammatory response.
... Similar stressrelated induction of this promoter serves to re-activate human CMV from latency, and is required for expression of genes necessary for DNA replication [83][84][85][86][87]. In the same manner, inflammation and cellular stress can significantly increase the transcription and expression of transgenes under control of the CMV immediate early promoter [87][88][89][90]. In these respects, OA cartilage is highly enriched with stress-activated chondrocytes [91], especially at sites of cartilage degradation, where GFP expression was greatest. ...
Article
Members of the cap'n'collar (CNC) family of transcription factors, including Nrf1 and Nrf2, heterodimerize with small Maf proteins (MafF, MafG and MafK) and regulate target gene expression through CNC-sMaf binding elements (CsMBEs). We recently developed a unique tethered dimer assessment system combined with small Maf triple knockout fibroblasts, which enabled the characterization of specific CNC-sMaf heterodimer functions. In this study, we evaluated the molecular function of the tethered Nrf1-MafG (T-N1G) heterodimer. We found that T-N1G activates the expression of proteasome subunit genes, well-known Nrf1 target genes, and binds specifically to CsMBEs in the proximity of these genes. T-N1G was also found to activate genes involved in proteostasis-related pathways, including endoplasmic reticulum-associated degradation, chaperone, and ubiquitin-mediated degradation pathways, indicating that the Nrf1-MafG heterodimer regulates a wide range of proteostatic stress response genes. By taking advantage of this assessment system, we found that Nrf1 has the potential to activate canonical Nrf2 target cytoprotective genes when strongly induced. Our results also revealed that transposable SINE B2 repeats harbor CsMBEs with high frequency and contribute to the target gene diversity of CNC-sMaf transcription factors.
Article
The use of cultured cells as a tool for research, precision medicine, biopharmacy, and biomanufacturing is constantly increasing. In parallel, the role of cell-cell and cell-substratum contacts in cell functioning is increasingly validated. Adhesion signalling plays a key role here. The activity of cell fate-regulating signalling molecules is an important factor in determining cell behaviour, as well as their response to treatment, depending on cell phenotypic status and location in the body. Three cellular state models (adherent, single cells in suspension, and aggregated cells) were compared for cell signalling, including focal adhesion (FAK), mitogen-activated (MAPK), as well as Akt protein kinases, and transcription factor cJun, by using lung adenocarcinoma A549, muscle-derived stem Myo, as well as primary lung cancer cell lines. Survival of both A549 and Myo cells was dependent on kinases Akt and ERK in detached conditions. Intercellular contacts in aggregates promoted activation of Akt and ERK, and cell survival. Loss of contacts with the substrate increased phosphorylation of MAP kinases JNK and p38, while decreased Akt phosphorylation by processes involving FAK. Unexpectedly, detachment increased phosphorylation of antiapoptotic kinase ERK in A549, while in Myo stem cells ERK phosphorylation was downregulated. JNK target transcription factor cJun protein level was markedly diminished by contacts between cells possibly involving mechanism of proteasomal degradation. Moreover, studies revealed the opposite dependence of molecules of the same signalling pathway - phospho-cJun and phospho-JNK - on cell culture density. Differences in ERK activation under detachment conditions indicate that targeting of prosurvival kinases during anoikis should be different in different cells. Moreover, the outcome of JNK activation in cells may depend on the amount of cJun, which is determined by cell-cell contacts.
Article
Full-text available
The major immediate early enhancer of human cytomegalovirus (HCMV) is known to exert a strong constitutive transcription stimulation in a broad spectrum of cells. This basal activity can be augmented considerably by elevated levels of intracellular cAMP in a cell type-specific manner. Cyclic AMP induction was observed in several lymphoid cell lines and in HeLa cells. One of the functionally important enhancer sequence modules, the 19 bp repeat element, mediates this effect as a cAMP-responsive element (CRE). It acts more efficiently than the corresponding sequence from the human chorionic gonadotropin gene. It is suggested that protein kinase C is involved in the pathway which leads to the activation of CRE-containing genes in lymphoid cells. Gel retardation assays indicated that similar, but not identical complexes are formed between nuclear protein extracts and the CREs of HCMV and the gonadotropin gene.
Article
Full-text available
The expression of cytomegalovirus alpha (immediate early) genes is under control of an enhancer that carries signals for strong constitutive expression as well as response elements for transactivation by viral proteins. We have used synthetic oligonucleotides representing the 16, 18 and 19 bp repeat elements within the enhancer to investigate the role of virus-induced cellular transcription factors in enhancer activation. We show that the transcription factor NF-kappa B, which binds to the 18 bp repeat, plays a central role in enhancer activation in infected human fibroblasts and that activation is mediated by the product of the viral gene ie1. The simian immunodeficiency virus kappa B site can functionally substitute for the 18 bp element in transient transactivation assays and can also compete efficiently for specific binding to the 18 bp repeat element in vitro. Point mutations in the NF-kappa B site within the 18 bp element disrupt ie1-mediated transactivation and binding. We have found that the characteristics of the 18 bp binding factor from human fibroblasts are indistinguishable from NF-kappa B induced by phorbol ester plus mitogen treatment of T lymphocytes, as determined by gel mobility shift assay as well as protection of the binding site from chemical cleavage. Furthermore, T cell stimulation mediates activation of the viral enhancer via kappa B sites, an observation that may be important in the interaction of cytomegalovirus with the naturally infected human host. Thus, NF-kappa B plays a central role as a target for enhancer activation via viral and cellular factors.
Article
Full-text available
We describe Mxi2, a human protein that interacts with Max protein, the heterodimeric partner of the Myc oncoprotein. Mxi2 encodes a 297-residue protein whose sequence indicates that it is related to extracellular signal-regulated kinases (ERK protein kinases). Mxi2 in yeast interacts with Max and with the C terminus of c-Myc. Mxi2 phosphorylates Max both in vitro and in vivo. The Mxi2 putative substrate recognition region has sequence similarity to the helix-loop-helix region in Max and c-Myc, suggesting that substrate recognition might be mediated via this motif. Phosphorylation by Mxi2 may affect the ability of Max to oligomerize with itself and its partners, bind DNA, or regulate gene expression.
Article
Mitogen-activated protein (MAP) kinases are important mediators of the cellular stress response. Here, we investigated the relationship between activation of the MAP kinase p38 and transcription factor NF-κB. Different forms of cellular stress were found to preferentially trigger either p38 or NF-κB. Arsenite or osmotic stress potently activated p38 but were ineffective in inducing NF-κB activation. Tumor necrosis factor-α and hydrogen peroxide, in contrast, led to NF-κB activation but only modestly stimulated p38. The activation of NF-κB was strongly abolished by antioxidants, while the activity of p38 and transcription factor AP-1 were increased. Inhibition of small GTPases including Rac and Cdc42 prevented p38 and AP-1 activation without interfering with NF-κB. In addition, inhibition of p38 by a pharmacological inhibitor or a dominant-negative mutant of MAP kinase kinase-6, an activator of the p38 pathway, interfered with NF-κB-dependent gene expression but not its DNA binding activity. Our results indicate that activation of p38 and NF-κB are mediated by separate pathways, which may converge further downstream in the cell nucleus. Different forms of cellular stress, however, initially trigger distinct signaling cascades involving either oxidative stress or GTPase-coupled pathways.
Article
Exposure of mammalian cells to DNA-damaging agents induces the ultraviolet (UV) response, involving transcription factor AP-1, composed of Jun and Fos proteins. We investigated the mechanism by which UV irradiation induces the c-jun gene. The earliest detectable step was activation of Src tyrosine kinases, followed by activation of Ha-Ras and Raf-1. The response to UV was blocked by tyrosine kinase inhibitors and dominant negative mutants of v-src, Ha-ras, and raf-1. This signaling cascade leads to increased phosphorylation of c-Jun on two serine residues that potentiate its activity. These results strongly suggest that the UV response is initiated at or near the plasma membrane rather than the nucleus. The response may be elicited by oxidative stress, because it is inhibited by elevation of intracellular glutathione. Using tyrosine kinase inhibitors, we demonstrate that the UV response has a protective function.
Article
Recent advances indicate a link between tumour promoters, transformation, and AP-1 activity. Protein kinase C activation increases AP-1 DNA-binding activity independently of new protein synthesis. AP-1 is also stimulated by transforming oncoproteins and growth factors. These proteins are thought to participate in a signalling cascade affecting the nuclear AP-1 complex composed of the Jun and Fos proteins. Because c-Jun is the most potent transactivator in the AP-1 complex and is elevated in Ha-ras-transformed cells, in which c-Fos is downregulated, we focused on it as a potential target. c-Jun could convert input from an oncogenic signalling cascade into changes in gene expression. Indeed, transformation of rat embryo fibroblasts by c-Jun requires an intact transcriptional activation domain and cooperation with oncogenic Ha-ras. Expression of oncogenic Ha-ras augments transactivation by c-Jun and stimulates its phosphorylation. Here we describe the mapping of the Ha-ras-responsive phosphorylation sites to serines 63 and 73 of c-Jun. Site-directed mutagenesis indicates that phosphorylation of these serines is essential for stimulation of c-Jun activity and for cooperation with Ha-ras in ocogenic transformation.
Article
A class of pyridinyl imidazoles inhibit the MAP kinase homologue, termed here reactivating kinase (RK) [Lee et al. (1994) Nature 372, 739-746]. We now show that one of these compounds (SB 203580) inhibits RK in vitro (IC50 = 0.6 microM), suppresses the activation of MAPKAP kinase-2 and prevents the phosphorylation of heat shock protein (HSP) 27 in response to interleukin-1, cellular stresses and bacterial endotoxin in vivo. These results establish that MAPKAP kinase-2 is a physiological RK substrate, and that HSP27 is phosphorylated by MAPKAP kinase-2 in vivo. The specificity of SB 203580 was indicated by its failure to inhibit 12 other protein kinases in vitro, and by its lack of effect on the activation of RK kinase and other MAP kinase cascades in vivo. We suggest that SB 203580 will be useful for identifying other physiological roles and targets of RK and MAPKAP kinase-2.
Article
Mammalian mitogen-activated protein (MAP) kinases include extracellular signal-regulated protein kinase (ERK), c-Jun amino-terminal kinase (JNK), and p38 subgroups. These MAP kinase isoforms are activated by dual phosphorylation on threonine and tyrosine. Two human MAP kinase kinases (MKK3 and MKK4) were cloned that phosphorylate and activate p38 MAP kinase. These MKK isoforms did not activate the ERK subgroup of MAP kinases, but MKK4 did activate JNK. These data demonstrate that the activators of p38 (MKK3 and MKK4), JNK (MKK4), and ERK (MEK1 and MEK2) define independent MAP kinase signal transduction pathways.
Article
Mammalian cells respond to endotoxic lipopolysaccharide (LPS) by activation of protein kinase cascades that lead to new gene expression. A protein kinase, p38, that was tyrosine phosphorylated in response to LPS, was cloned. The p38 enzyme and the product of the Saccharomyces cerevisiae HOG1 gene, which are both members of the mitogen-activated protein (MAP) kinase family, have sequences at and adjacent to critical phosphorylation sites that distinguish these proteins from most other MAP kinase family members. Both HOG1 and p38 are tyrosine phosphorylated after extracellular changes in osmolarity. These findings link a signaling pathway in mammalian cells with a pathway in yeast that is responsive to physiological stress.
Article
An IL-1-stimulated protein kinase cascade resulting in phosphorylation of the small heat shock protein hsp27 has been identified in KB cells. It is distinct from the p42 MAP kinase cascade. An upstream activator kinase phosphorylated a 40 kDa kinase (p40) upon threonine and tyrosine residues, which in turn phosphorylated a 50 kDa kinase (p50) upon threonine (and some serine) residues. p50 phosphorylated hsp27 upon serine. p40 and p50 were purified to near homogeneity. All three components were inactivated by protein phosphatase 2A, and p40 was inactivated by protein tyrosine phosphatase 1B. The substrate specificity of p40 differed from that of p42 and p54 MAP kinases. The upstream activator was not a MAP kinase kinase. p50 resembled MAPKAPK-2 and may be identical.