ArticlePDF Available

NF-??B promotes leaky expression of adenovirus genes in a replication-incompetent adenovirus vector

Authors:

Abstract and Figures

The replication-incompetent adenovirus (Ad) vector is one of the most promising vectors for gene therapy; however, systemic administration of Ad vectors results in severe hepatotoxicities, partly due to the leaky expression of Ad genes in the liver. Here we show that nuclear factor-kappa B (NF-κB) mediates the leaky expression of Ad genes from the Ad vector genome, and that the inhibition of NF-κB leads to the suppression of Ad gene expression and hepatotoxicities following transduction with Ad vectors. Activation of NF-κB by recombinant tumor necrosis factor (TNF)-α significantly enhanced the leaky expression of Ad genes. More than 50% suppression of the Ad gene expression was found by inhibitors of NF-κB signaling and siRNA-mediated knockdown of NF-κB. Similar results were found when cells were infected with wild-type Ad. Compared with a conventional Ad vector, an Ad vector expressing a dominant-negative IκBα (Adv-CADNIκBα), which is a negative regulator of NF-κB, mediated approximately 70% suppression of the leaky expression of Ad genes in the liver. Adv-CADNIκBα did not induce apparent hepatotoxicities. These results indicate that inhibition of NF-κB leads to suppression of Ad vector-mediated tissue damages via not only suppression of inflammatory responses but also reduction in the leaky expression of Ad genes.
Content may be subject to copyright.
1
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
www.nature.com/scientificreports
NF-κB promotes leaky expression
of adenovirus genes in a replication-
incompetent adenovirus vector
M. Machitani
1
, F. Sakurai
1,2
, K. Wakabayashi
1
, K. Nakatani
1
, K. Shimizu
1,3
, M. Tachibana
1
&
H. Mizuguchi
1,4,5,6,7
The replication-incompetent adenovirus (Ad) vector is one of the most promising vectors for gene
therapy; however, systemic administration of Ad vectors results in severe hepatotoxicities, partly
due to the leaky expression of Ad genes in the liver. Here we show that nuclear factor-kappa B (NF-
κB) mediates the leaky expression of Ad genes from the Ad vector genome, and that the inhibition
of NF-κB leads to the suppression of Ad gene expression and hepatotoxicities following transduction
with Ad vectors. Activation of NF-κB by recombinant tumor necrosis factor (TNF)-α signicantly
enhanced the leaky expression of Ad genes. More than 50% suppression of the Ad gene expression
was found by inhibitors of NF-κB signaling and siRNA-mediated knockdown of NF-κB. Similar results
were found when cells were infected with wild-type Ad. Compared with a conventional Ad vector, an
Ad vector expressing a dominant-negative IκBα (Adv-CADNIκBα), which is a negative regulator of
NF-κB, mediated approximately 70% suppression of the leaky expression of Ad genes in the liver. Adv-
CADNIκBα did not induce apparent hepatotoxicities. These results indicate that inhibition of NF-κB
leads to suppression of Ad vector-mediated tissue damages via not only suppression of inammatory
responses but also reduction in the leaky expression of Ad genes.
e replication-incompetent E1-deleted adenovirus (Ad) vector is the most promising vector for both gene ther-
apy and basic studies due to its advantages as a gene delivery vehicle, which include high titer production and high
transduction eciencies in both dividing and non-dividing cells
1,2
. In a conventional Ad vector, the E1 (E1A,E1B)
gene is replaced by a transgene expression cassette and the Ad vector is propagated in E1-expressing cell lines,
such as HEK293 cells. e E1A gene is the rst transcription unit to be expressed immediately aer wild-type Ad
infection, and transactivates the expression of other Ad genes. us, no Ad genes should be expressed from the
Ad vector genome due to the lack of the E1A and E1B genes in the Ad vector genome. However, expression of Ad
genes, including the E2, E4 and pIX genes, is indeed detected in the Ad vector-transduced cells, indicating that
the Ad genes are expressed from the Ad vector genome in an E1A gene-independent manner
3–6
. Further, the leaky
expression results in immune responses against Ad proteins and Ad protein-induced cellular toxicity, leading to
tissue damages such as hepatotoxicities
7–9
.
Various types of next-generation Ad vectors which can overcome these disadvantages have been developed
3,4,8
.
For example, an Ad vector lacking the E2A gene expression achieved a reduction in the immune responses
and the prolonged transgene expression proles
10
. A helper-dependent Ad (HD-Ad) vector, in which all viral
encoding genes were deleted, also showed a reduction in the production of inammatory cytokines and the
tissue damages
11–13
. In addition, we recently developed an Ad vector that suppresses the leaky expression of Ad
1
Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University,
1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
2
Laboratory of Regulatory Sciences for Oligonucleotide Therapeutics,
Clinical Drug Development Unit, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka,
Suita, Osaka, 565-0871, Japan.
3
Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiorikita, Tondabayashi,
Osaka, 584-8540, Japan.
4
Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health
and Nutrition, 7-6-8 Saito, Asagi, Ibaraki, Osaka 567-0085, Japan.
5
iPS Cell-Based Research Project on Hepatic
Toxicity and Metabolism, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita,
Osaka 565-0871, Japan.
6
Global Center for Advanced Medical Engineering and Informatics, Osaka University, 2-2
Yamadaoka, Suita, Osaka 565-0871, Japan.
7
Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita,
Osaka 565-0871, Japan. Correspondence and requests for materials should be addressed to F.S. (email: sakurai@
phs.osaka-u.ac.jp) or H.M. (email: mizuguch@phs.osaka-u.ac.jp)
Received: 28 August 2015
Accepted: 21 December 2015
Published: 27 January 2016
OPEN
www.nature.com/scientificreports/
2
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
genes by inserting microRNA-targeted sequences in the Ad vector genome
9
. Nonetheless, problems remain with
all of these novel Ad vectors, including the complexity of the Ad vector preparation and some remaining Ad
vector-induced hepatotoxicities. To overcome these drawbacks, the mechanisms of the E1A gene-independent
leaky expression of Ad genes should be claried.
Transduction with Ad vectors causes activation of a number of transcriptional factors. For example, injection
with recombinant Ad vectors immediately induces production of inammatory cytokines associated with innate
immune responses
14,15
, suggesting that several transcriptional factors would be activated in the innate immune
responses. In this study, among these transcriptional factors, we focused on nuclear factor-kappa B (NF-κ B)
because NF-κ B has been demonstrated to play a crucial role in the expression of numerous viral genes
16
. NF-κ B
is a ubiquitous transcriptional factor which promotes the expression of a large number of genes, particularly
those from gene families associated with host immune responses
16
. us, NF-κ B plays a crucial role in immune
responses. Although NF-κ B functions as a homodimer or heterodimer (p50/p50, p50/p65, and p52/RelB), canon-
ical NF-κ B is a heterodimer which is composed of p50 and p65. Under normal conditions, NF-κ B stays in the
cytoplasm via association with Iκ Bα , an NF-κ B-inhibitory factor. Upon the stimulation by cytokines and path-
ogens, Iκ Bα is phosphorylated and degraded, leading to localization of NF-κ B into the nucleus from the cyto-
plasm. In the nucleus, translocated NF-κ B promotes the expression of target genes by binding to the consensus
sequence (5 GGGACT TTCC-3 ) in their promoter region.
In the present study, we have demonstrated that NF-κ B promotes Ad gene expression following transduction
with an Ad vector and a wild-type Ad (WT-Ad). Further, an Ad vector expressing a dominant-negative Iκ Bα
(Adv-CADNIκ Bα ), which is a negative regulator of NF-κ B, mediated the signicant suppression of the leaky
expression of Ad genes, with the result that no apparent hepatotoxicities were induced.
Materials and Methods
Cell lines and animals. HEK293 (a transformed embryonic kidney cell line) and HeLa (a human epithe-
lial carcinoma cell line) cells were cultured in Dulbeccos modied Eagles medium supplemented with 10%
fetal bovine serum (FBS), streptomycin (100 μ g/ml), and penicillin (100 U/ml). H1299 (a non-small cell lung
carcinoma cell line) cells were cultured in RPMI1640 supplemented with 10% FBS, streptomycin (100 μ g/ml),
and penicillin (100 U/ml). Female C57BL/6 mice aged 5–6 weeks were obtained from Nippon SLC (Shizuoka,
Japan). Rag2/Il2rγ double knockout mice of C57BL/6 background, aged 5–7 weeks, were obtained from Taconic
Farms (Hudson, NY)
17,18
. All mouse experimental procedures used in this study were approved by Animal
Experimentation Committee of Osaka University and performed in accordance with institutional guidelines for
animal experiments at Osaka University.
Reagents. Recombinant human and mouse tumor necrosis factor (TNF)-α were purchased from Invivogen
(San Diego, CA) and Peprotech (Rocky Hill, NJ), respectively. NF-κ B inhibitors, BAY11-7082 and MG-132, were
purchased from Invivogen. Recombinant human interferon-α (IFN-α ) was purchased from PBL interferon
source (Piscataway, NJ).
Plasmids. pNF-κ B-Luc, which contains the rey luciferase (FLuc) gene expression cassette driven by a pro-
moter containing a consensus sequence for the NF-κ B binding, was purchased from Agilent Technologies (Santa
Clara, CA). pHMCMV-RLuc, a reporter plasmid carrying a cytomegalovirus (CMV) promoter-driven renilla
luciferase (RLuc) expression cassette, was constructed previously
19
.
e reporter plasmids pGL4-E1A, -E1B, -E2, -E3, and -E4, which have the promoter sequences of the respec-
tive Ad genes upstream of the FLuc gene, were constructed as follows: the fragment containing the E2 promoter
sequence (bp 27061–27661) was amplified by PCR using the primers E2p-F and E2p-R, and ligated with a
HindIII/XhoI fragment of pGL4.10 (Promega, Madison, WI), resulting in pGL4-E2. pGL4-E1A, -E1B, -E3, and -E4,
which contain the E1A (bp 1–546), E1B (bp 1336–1702), E3 (bp 26987–27578), and E4 (bp 35530–35939) pro-
moter region, respectively, were similarly constructed using the corresponding primers. e mutated reporter
plasmids pGL4-E2-del1, -del2, and -del3, which have shortened E2 promoter sequences (bp 27061–27296, 27061–
27197, and 27061–27136, respectively) upstream of the FLuc gene, were constructed as follows. First, pGL4-E2
was digested with EcoRV/XhoI. e resulting fragments were then self-ligated aer the sticky end was converted
to a blunt end, resulting in pGL4-E2-del1. e fragment containing the shortened E2 promoter sequence (bp
27061–27197) was amplied by PCR using the primers E2-del2-F and E2-del2-R, and ligated with a HindIII/XhoI
fragment of pGL4.10, resulting in pGL4-E2-del2. An AII/XhoI fragment of pGL4-E2 was ligated with the oligo-
nucleotides, E2-del3-S and E2-del3-AS, which encode the shortened E2 promoter sequence (bp 27061–27136),
resulting in pGL4-E2-del3. A reporter plasmid, pGL4-del2.1, which has the shortened E2 promoter sequences
(bp 27061–27197) and lacks the predicted NF-κ B binding site in the E2 promoter sequence, was constructed as
follows: the predicted NF-κ B binding site was mutated by using a QuikChange Site-Directed Mutagenesis Kit
(Agilent Technologies) and the primers E2-del2.1-F and E2-del2.1-R, resulting in pGL4-E2-del2.1. e mutated
sequence of pGL4-E2-del2.1 is shown in Fig.1B. Sequences of the primers and oligonucleotides are shown in
Table S1.
pAdHM4-CMV-Luc, the Ad vector plasmid carrying a CMV promoter-driven luciferase expression cas-
sette, was constructed previously
9
. pAdHM4-CA-Luc and -DNIκ B α , the Ad vector plasmid carrying a CA
promoter (a β -actin promoter/CMV enhancer with a β -actin intron)-driven luciferase and dominant-negative
Iκ Bα gene expression cassette, respectively, were constructed as follows: pHMCA5
20
was digested with
NotI/XbaI, and ligated with the NotI/XbaI fragment of pCMVL1
9
, resulting in pHMCA5-Luc. e fragment
containing the dominant-negative Iκ Bα gene was amplied by PCR using pCMX-Iκ Bα M (Addgene plasmid
12329; Addgene, Cambridge, MA), which contains the dominant-negative Iκ Bα gene expression cassette, and
the primers DNIκ Bα -F and DNIκ Bα -R, and ligated with the NotI/XbaI fragment of pHMCA5, resulting in
www.nature.com/scientificreports/
3
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
pHMCA5-DNIκ Bα . pHMCA5-Luc and -DNIκ Bα were digested with I-CeuI/PI-SceI, and then ligated with
I-CeuI/PI-SceI digested pAdHM4
21
, resulting in pAdHM4-CA-Luc and pAdHM4-CA-DNIκ Bα , resp ectively.
Viruses. WT-Ad (human Ad serotype 5) was obtained from the American Type Culture Collection (ATCC).
Recombinant Ad vectors were prepared by an in vitro improved ligation method as follows
22,23
; all Ad vector
plasmids used in this study have PacI-anked Ad vector genome. Ad vector plasmids, pAdHM4-CMV-Luc,
pAdHM4-CA-Luc, and pAdHM4-CA-DNIκ Bα , were digested with PacI to release the recombinant Ad vector
genome, and were each transfected into HEK293 cells using Lipofectamine 2000 (Life Technologies, Carlsbad,
CA), resulting in Adv-CMVLuc, Adv-CALuc, and Adv-CADNIκ Bα , respectively. Adv-CALacZ, an Ad vector
expressing LacZ, was constructed previously
24
. ese Ads were amplied and puried by two rounds of cesium
chloride-gradient ultracentrifugation, dialyzed, and stored at 80 °C
22
. e virus particles (VP) were deter-
mined by a spectrophotometrical method
22
. Determination of infectious units (IFU) was accomplished using an
Adeno-X Rapid Titer Kit (Clontech, Mountain View, CA).
Prediction of NF-κB binding sites in the Ad genome. Transcriptional factors binding to the E2 pro-
moter region and the putative binding sites in the promoter region were predicted using TFSEARCH (http://
www.cbrc.jp/research/db/TFSEARCH.html).
Analysis of the promoter activities using reporter plasmids. Cells were seeded in a 24-well plate at a
density of 5 × 10
4
cells/well. On the following day, cells were co-transfected with FLuc-expressing reporter plas-
mids (500 ng/ml) (constructed as shown above) and pHMCMV-RLuc (160 ng/ml) using Lipofectamine 2000 (Life
Technologies). Following a 6-h incubation, the medium was changed. Luciferase activity in cells was determined
using the Dual Luciferase Reporter Assay System (Promega) 24 h aer transfection.
Figure 1. Reduction in the E2 promoter activity by deletion of an NF-κB binding site. (A,B) HeLa cells were
co-transfected with pHMCMV6-RLuc and reporter plasmids carrying the E2-promoter-driven FLuc expression
cassette (pGL4, pGL4-E2, pGL4-E2-del1, pGL4-E2-del2, pGL4-E2-del2.1, or pGL4-E2-del3). Aer a 24-h
incubation, luciferase activity was determined. e data show FLuc activity normalized by RLuc activity in the
cells. Schematic diagrams of each promoter are shown at the le of the graph. (B) Nucleotide sequence around
the NF-κ B binding site (italic type) in the E2 promoter region. e NF-κ B binding sequence is deleted in pGL4-
E2-del2.1. ese data are expressed as the means ± S.D. (n = 4). ***p < 0.001.
www.nature.com/scientificreports/
4
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
For evaluation of the eects of TNF-α on the promoter activities, cells were treated with TNF-α (100 ng/
ml) 24 h aer transfection with plasmids as described above. Luciferase assay was performed 24 h aer TNF-α
treatment.
Quantitative RT-PCR analysis of Ad gene expression. Cells were seeded in a 24-well plate at a den-
sity of 5 × 10
4
cells/well. On the following day, WT-Ad and Ad vectors were added to the cells at the indicated
titers. Following the indicated incubation periods, total RNA was isolated from cells using ISOGEN (Nippon
Gene, Tokyo, Japan). cDNA was synthesized using 500 ng of total RNA with a Superscript VILO cDNA synthesis
kit (Life Technologies). Real-time RT-PCR analysis was performed using Fast SYBR Green Master Mix (Life
Technologies) and StepOnePlus real-time PCR systems (Life Technologies). Sequences of the primers used in this
study are described in Table S1.
For evaluation of the eects of TNF-α on Ad gene expression, cells were pre-treated with TNF-α at a concen-
tration of 100 ng/ml for 5 h, followed by incubation with WT-Ad and Ad vectors. Total RNA was recovered for
quantitative RT-PCR analysis 12 h aer addition of WT-Ad and Ad vectors.
For inhibition of NF-κ B, cells were pre-treated with BAY11-7082 and MG-132 at 10 μ M and 2.5 μ M, respec-
tively, for 1 h, followed by incubation with WT-Ad and Ad vectors. Total RNA was recovered 12 h aer the addi-
tion of WT-Ad and Ad vectors.
For knockdown of p50, cells were transfected with an siRNA against p50 (sip50) (Gene Design, Osaka, Japan)
using Lipofectamine 2000 (Life Technologies) according to the manufacturer’s instructions. e target sequence
of sip50 was 5 -aaggggctataatcctggact-3 . Control siRNA was purchased from Qiagen (Allstars Negative Control
siRNA; Qiagen, Hilden, Germany). Cells were treated with WT-Ad and Ad vectors 48 h aer siRNA transfection.
Total RNA was recovered 12 h aer addition of WT-Ad and Ad vectors.
Western blotting analysis. Western blotting assay was performed as previously described
13
. Briefly,
whole-cell extracts were prepared and electrophoresed on 10% sodium dodecyl sulfate (SDS)-polyacrylamide
gels under reducing conditions, followed by electrotransfer to PVDF membranes (Millipore, Bedford, MA). Aer
blocking with 5% skim milk prepared in TBS-T (tween-20, 0.1%), the membrane was incubated with a mouse
anti-E1A antibody (clone name) (Abcam, Cambridge, UK), a rabbit anti-p50 antibody (H-119) (Santa Cruz
Biotechnology, Santa Cruz, CA), or mouse anti-β -actin (Sigma-Aldrich, St. Louis, MO), followed by incubation
in the presence of horseradish peroxidase (HRP)-labeled anti-rabbit or anti-mouse IgG antibody (Cell Signaling
Technology, Danvers, MA). e intensity of protein bands was quantied by Image J soware.
Determinaton of Ad genome copy numbers. Cells were treated with WT-Ad and Ad vectors in a man-
ner similar to that described above. Following the indicated incubation periods, total DNA, including Ad genomic
DNA, was isolated from the cells infected with Ads using a DNeasy Blood & Tissue Kit (Qiagen). Aer isolation,
Ad genome copy numbers were quantied using a StepOnePlus real-time PCR system (Life Technologies) as pre-
viously described
25
. Sequences of the primers and probes used in this study are described in Table S1.
Chromatin immunoprecipitation (ChIP) assay. HeLa cells were infected with WT-Ad at 100 VP/cell.
Following a 24-h incubation, cells were treated with formaldehyde at a nal concentration of 1% for crosslinking,
and then genomic DNA was fragmented by sonication. e DNA fragment-protein complexes were immuno-
precipitated using a rabbit anti-p65 antibody (ab7970) (Abcam) or normal rabbit IgG (Cell Signaling). e ChIP
assay kit was purchased from Merck Millipore (Darmstadt, Germany). e precipitated DNA copy numbers were
determined by quantitative PCR using the primers shown in Table S1.
In vivo Ad gene expression analysis. Ad vectors were intravenously administered to mice at a dose of 10
9
IFU/mouse via a tail vein. Total RNA was extracted from the livers 48 h aer administration, and the Ad gene
mRNA levels were determined by quantitative RT-PCR analysis.
Analysis of Ad vector-mediated hepatotoxicities. Ad vectors were intravenously administered to mice
at a dose of 10
9
IFU/mouse via a tail vein. e blood samples were collected via retro-orbital bleeding at the
indicated days, and the serum samples were obtained aer centrifugation. e serum alanine aminotransferase
(ALT) and aspartate aminotransferase (AST) levels were determined using a transaminase-CII-test kit (Wako,
Osaka, Japan).
Statistical analysis. Statistical signicance was determined using Students t-test. Data are presented as the
means ± S.D or S.E.
Results
The NF-κB binding site is essential for the E2 promoter activity. In order to identify the transcrip-
tional factors crucial for leaky expression of the Ad genes, functional analysis of a promoter of the E2 gene, which
shows leaky expression from the Ad vector genome
5
, was performed using reporter plasmids (Fig.1A). e lucif-
erase expression levels from pGL4-E2-del3, which has the shortened promoter sequence (bp 27061–27136) of
the E2 gene upstream of the FLuc gene, was signicantly lower than that from pGL4-E2, which has the promoter
sequence (bp 27061–27661) of the E2 gene upstream of the FLuc gene (Fig.1A), suggesting that the sequence
between bp 27136–27197 in the E2 promoter was signicantly involved in the E2 promoter activity. Next, we
performed in silico analysis using TFSEARCH, a program that searches for transcriptional factor binding sites,
to identify transcriptional factor binding sites in the sequence between bp 27136–27197. is analysis revealed
an NF-κ B binding site (5 -gggaatttcc-3 ) in this sequence (Fig.1B). To examine whether the NF-κ B binding site
is essential for the E2 promoter activity, the NF-κ B binding site in the E2 promoter sequence of pGL4-E2-del2
www.nature.com/scientificreports/
5
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
was deleted. e deletion of the NF-κ B binding site resulted in a signicant reduction in the luciferase activity of
pGL4-E2-del2 (Fig.1B). ese results suggested that the NF-κ B binding site was important for the E2 promoter
activity.
Activation of NF-κB promotes Ad gene expression and Ad replication. In order to examine
whether NF-κ B is involved in the leaky expression of Ad genes, including the E2 gene, HeLa cells were pre-treated
with recombinant human TNF-α (hTNF-α ), followed by infection with WT-Ad. The reporter assay using
pNF-κ B-Luc, which carries the luciferase gene expression cassette driven by a promoter containing a consensus
sequence for NF-κ B binding, showed that hTNF-α treatment signicantly activated NF-κ B signaling (Fig.2A).
Activation of the NF-κ B signaling by hTNF-α signicantly (1.4-2-fold) enhanced the mRNA levels of the Ad
early genes, including the E1A, E2, E3, and E4 genes, in HeLa cells following infection with WT-Ad (Fig.2B). e
E1A protein levels were also elevated by TNF-α treatment in WT-Ad-infected HeLa cells (Fig.2C). WT-Ad repli-
cated more eciently in HeLa cells pre-treated with hTNF-α in a dose-dependent manner (Fig.2D). In addition,
hTNF-α treatment signicantly enhanced the leaky expression of the E2 and E4 genes in HeLa cells following
transduction with Adv-CMVLuc, a replication-incompetent Ad vector expressing FLuc (Fig.2E). ese results
suggested that the activation of NF-κ B by TNF-α promoted Ad gene expression and Ad replication.
Inhibition of NF-κB negatively regulates Ad gene expression and Ad replication. Next, in order
to examine the involvement of NF-κ B in Ad gene expression and Ad replication, HeLa cells were pre-treated
with the NF-κ B inhibitors BAY11-7082 and MG-132, followed by infection with WT-Ad. e cell viabilities
were not signicantly reduced following treatment with BAY11-7082 and MG-132 (Figure S1). e reporter
assay using pNF-κ B-Luc showed that the treatment with BAY11-7082 and MG-132 largely inhibited NF-κ B sig-
naling (Fig.3A). Inhibition of NF-κ B signaling by BAY11-7082 and MG-132 resulted in more than 80% reduc-
tion in the E1A, E2, E3, and E4 gene expressions in WT-Ad-infected HeLa cells (Fig.3B). Copy numbers of the
WT-Ad genome were also suppressed by more than 80% in HeLa cells following treatment with BAY11-7082
and MG-132 (Fig.3C). In addition, treatment with BAY11-7082 and MG-132 reduced the leaky expression of
the E2 and E4 genes by approximately 50% in HeLa cells following transduction with Adv-CMVLuc (Fig.3D).
Next, in order to further examine the eects of NF-κ B on Ad gene expression and Ad replication, p50, which
is a component of NF-κ B, was knocked down in HeLa cells. Transfection with sip50 resulted in a signicant
knockdown of p50 at both the mRNA and protein levels (Fig.4A,B). e E1A, E2, E3, and E4 mRNA levels
following infection with WT-Ad were suppressed by more than 50% in p50-knockdown cells, compared with
those in control cells (Fig.4C). An approximately 90% reduction in copy numbers of the WT-Ad genome was
found in p50-knockdown HeLa cells (Fig.4D). Knockdown of p50 also led to approximately 70% decreases in
the leaky expression of the E2 and E4 genes following transduction with Adv-CMVLuc (Fig.4E). Similar results
were found in H1299 cells (Figure S2). ese results indicated that NF-κ B was essential for Ad gene expression
and replication of WT-Ad, and that NF-κ B induced the leaky expression of the Ad genes following transduction
with Ad vectors.
NF-κB binds to the E2 and E3 promoter. In order to examine whether NF-κ B enhances Ad gene pro-
moter activities, HeLa cells were transfected with reporter plasmids carrying the Ad gene promoter-driven lucif-
erase expression cassette, followed by treatment with hTNF-α . e E2 and E3 promoter activities, but not the
activities of the E1A, E1B, or E4 promoter, were more than 3-fold enhanced by hTNF-α treatment (Fig.5A). Next,
we performed a ChIP assay using an antibody against p65, which is another component of NF-κ B, to examine
whether NF-κ B directly binds to the E2 and E3 promoter region. e E2 and E3 promoter is a bidirectional pro-
moter, which drives transcription of both the E2 and E3 genes. e ChIP assay demonstrated that NF-κ B directly
bound to the E2 and E3 promoter region (Fig.5B), indicating that NF-κ B activates the E2 and E3 gene expression
via direct binding to the promoter region.
IFN-α treatment suppresses Ad gene expression and Ad replication. Next, we investigated the
involvement of other transcriptional factors in Ad gene expression. In order to activate transcriptional factors
involved in innate immune responses, HeLa cells were pre-treated with recombinant human IFN-α , which acti-
vates STAT1, STAT2, and IRF9, followed by infection with WT-Ad. e reporter assay using pNF-κ B-Luc showed
that IFN-α treatment signicantly inhibited NF-κ B signaling (Figure S3A). Contrary to TNF-α treatment, IFN-α
treatment signicantly suppressed the mRNA levels of the Ad early genes, including the E1A, E2, E3, and E4
genes, in HeLa cells following infection with WT-Ad (Figure S3B). WT-Ad replicated less eciently in HeLa cells
pre-treated with IFN-α (Figure S3C). ese results suggested that transcriptional factors activated by IFN-α did
not promote Ad gene expression and Ad replication.
NF-κB is involved in Ad gene expression in the mouse liver. Next, in order to examine the eects of
NF-κ B on the Ad gene expression in vivo, mice were intravenously administered WT-Ad and Ad vectors, followed
by intravenous injection of recombinant mouse TNF-α (mTNF-α ) 24 h aer Ad injection. Ad gene expression
levels in the liver were determined by quantitative RT-PCR analysis. e liver is a main organ which an Ad vector
are distributed to following intravenous administration. Ad gene expression from the WT-Ad genome in the liver
increased by more than 9-fold by mTNF-α administration (Fig.6A). mTNF-α treatment also mediated a signif-
icant increase in the leaky expression of the E2 and E4 genes from the Ad vector genome in the liver (Fig.6B).
Next, to inhibit NF-κ B signaling in the liver, we constructed an Ad vector expressing a dominant-negative Iκ Bα
(Adv-CADNIκ Bα ), which is a negative regulator of NF-κ B
26
. Adv-CADNIκ Bα induced signicant inhibition of
NF-κ B signaling, compared with a conventional Ad vector expressing LacZ (Adv-CALacZ) (Fig.7A). Compared
with the leaky expression levels of the Ad genes from Adv-CALuc, Adv-CADNIκ Bα mediated an approximately
70% suppression of the leaky expression of the Ad genes in HeLa cells (Fig.7B) and in the liver (Fig.7C). ese
www.nature.com/scientificreports/
6
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
results indicated that NF-κ B mediated the leaky expression of the Ad genes following administration with Ad
vectors in not only cultured cells but also the organs.
Inhibition of NF-κB leads to suppression of the leaky expression of Ad genes and reduction
in the hepatotoxicities. Several groups, including ours, previously reported that the leaky expression of
Ad genes from the Ad vector genome induced tissue damages such as hepatotoxicities
7–9
. erefore, in order to
Figure 2. Promotion of Ad early gene expression and replication by TNF-α stimulation. (A) HeLa cells
were transfected with pNF-κ B-Luc, followed by treatment with recombinant hTNF-α at 100 ng/ml. Aer 24-h
incubation, luciferase activity was determined. The data show FLuc activity normalized by RLuc activity.
(B,C) HeLa cells were pre-treated with hTNF-α at 100 ng/ml for 5 h, followed by infection with WT-Ad at
100 VP/cell. Aer 12-h incubation, the E1A, E2, E3, and E4 mRNA levels in the cells were determined by
quantitative RT-PCR (B). Aer 24-h incubation, the E1A protein levels in the cells were determined by western
blotting analysis (C). e intensity of E1A expression was quantied using Image J soware. (D) HeLa cells
were pre-treated with hTNF-α at the indicated concentration for 5 h, followed by infection with WT-Ad at
100 VP/cell. After 24-h incubation, Ad genome copy numbers in the cells were determined by quantitative
PCR. (E) HeLa cells were pre-treated with hTNF-α at 100 ng/ml for 5 h, followed by transduction
with Adv-CMVLuc at 100 VP/cell. Aer 12-h incubation, Ad gene mRNA levels in the cells were similarly
determined. ese data are expressed as the means ± S.D. (n = 3–4). *p < 0.05, **p < 0.01, ***p < 0.001.
www.nature.com/scientificreports/
7
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
Figure 3. Suppression of Ad early gene expression and replication by NF-κB inhibitors. (A) HeLa cells were
pre-treated with BAY11-7082 and MG-132 at 10 mM and 2.5 mM, respectively, for 1 h, followed by transfection
with pNF-κ B-Luc. Aer 24-h incubation, luciferase activity was determined. e data show FLuc activity
normalized by RLuc activity. (B,C) HeLa cells were pre-treated with BAY11-7082 and MG-132 at 10 mM and
2.5 mM, respectively, for 1 h, followed by infection with WT-Ad at 100 VP/cell. Aer 12-h incubation, the E1A,
E2, E3, and E4 mRNA levels in the cells were determined by quantitative RT-PCR (B). Aer 24-h incubation,
Ad genome copy numbers in the cells were determined by quantitative PCR (C). (D) HeLa cells were pre-treated
with BAY11-7082 and MG-132 at 10 mM and 2.5 mM, respectively, for 1 h, followed by infection with Adv-
CMVLuc at 100 VP/cell. Aer 12-h incubation, Ad gene mRNA levels in the cells were similarly determined.
ese data are expressed as the means ± S.D. (n = 3–4). *p < 0.05, **p < 0.01, ***p < 0.001.
Figure 4. Suppression of Ad early gene expression and replication by NF-κB knockdown. (A,B) HeLa and
H1299 cells were transfected with siControl or sip50. Aer 48-h incubation, mRNA and protein levels of p50
in the cells were determined by quantitative RT-PCR (A) and western blotting analysis (B), respectively. p50
mRNA levels in the cells transfected with siControl were normalized to 1. (C,D) HeLa cells were transfected
with sip50 at 50 nM, followed by infection with WT-Ad at 100 VP/cell. Aer 12-h incubation, the E1A, E2,
E3, and E4 mRNA levels in the cells were determined by quantitative RT-PCR (C). Aer 24-h incubation, Ad
genome copy numbers in the cells were determined by quantitative PCR (D). (E) HeLa cells were transfected
with sip50 at 50 nM, followed by transduction with Adv-CMVLuc at 100 VP/cell. Aer 12-h incubation,
Ad gene mRNA levels in the cells were similarly determined. ese data are expressed as the means ± S.D.
(n = 3–4). *p < 0.05, **p < 0.01, ***p < 0.001.
www.nature.com/scientificreports/
8
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
Figure 5. NF-κB-mediated enhancement of the E2 and E3 promoter activity. (A) HeLa cells were
transfected with reporter plasmids carrying the expression cassette of Ad early gene promoter-driven FLuc
(pGL4-E1A, -E1B, -E2, -E3, and -E4) for 24 h, followed by treatment with hTNF-α at 100 ng/ml. Aer 24-h
incubation, luciferase activity was determined. (B) HeLa cells were infected with WT-Ad at 100 VP/cell. Aer
24-h incubation, NF-κ B binding DNA levels were determined by ChIP assay. ese data are expressed as the
means ± S.D. (n = 3–4). **p < 0.01, ***p < 0.001.
Figure 6. Promotion of Ad early gene expression by TNF-α stimulation in mouse liver. (A,B) C57BL/6
mice were intravenously administered 10
9
IFU of WT-Ad (A) or Adv-CALuc (B), followed by intravenous
administration of recombinant mTNF-α at 0.5 mg/mouse. Twenty-four hours aer the administration, the
E1A, E2, E3, and E4 mRNA levels in the mouse liver were determined by quantitative RT-PCR. ese data are
expressed as the means ± S.E. (n = 5–6). *p < 0.05, **p < 0.01, ***p < 0.001.
www.nature.com/scientificreports/
9
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
examine whether inhibition of NF-κ B leads to the suppression of Ad vector-mediated hepatotoxicities via reduc-
tion in the leaky expression of Ad genes, serum ALT and AST levels, which are representative biomarkers of hepa-
totoxicities, were measured at the indicated days aer administration with Adv-CALuc and Adv-CADNIκ Bα
(Fig.8A). ALT and AST levels were not apparently elevated following administration with Adv-CADNIκ Bα ,
while Adv-CALuc showed signicant elevation in serum ALT and AST levels.
Over-expression of DNIκ B α in immune cells might inhibit the activation of immune cells via innate
immune responses, leading to a reduction in the hepatotoxicities. Ad vector-induced hepatotoxicities
are mediated by cytotoxic T cells as well as Ad proteins themselves
7–9
. An Ad vector transduces not only
hepatocytes but also dendritic cells, which are professional antigen presenting cells, in the spleen following
intravenous administration
27
. Next, in order to examine whether the suppression of Ad vector-mediated
hepatotoxicities by DNIκ B α expression is induced even in immune-deficient mice, Adv-CALuc and
Adv-CADNIκ Bα were intravenously administered to Rag2/Il2rγ double knockout mice, which have global
defects in both cellular and humoral immune responses due to the lack of T, B, and natural killer (NK)
cells
17,18
. Compared with the leaky expression levels of the E2 genes in the liver of Rag2/Il2rg double knock-
out mice following intravenous administration with Adv-CALuc, Adv-CADNIκ Bα exhibited significantly
lower expression levels of the E2 genes in the liver (Fig.8B). Rag2/Il2rγ double knockout mice showed sig-
nificant elevation in serum ALT and AST levels following intravenous administration with Adv-CALuc, but
not with Adv-CADNIκ B α (Fig.8C). These results indicated that NF-κ B-mediated leaky expression of Ad
genes induced hepatotoxicities, and that Ad-CADNIκ Bα did not induce any apparent hepatotoxicity due to
significant suppression of the leaky expression of Ad genes.
Figure 7. Suppression of Ad early gene expression by expression of a dominant-negative IκBα gene.
(A) HeLa cells were transfected with pNF-κ B-Luc, followed by transduction with Adv-CALuc or
Adv-CADNIκ Bα at an MOI of 5. Aer 48-h incubation, luciferase activity was determined. e data show FLuc
activity normalized by RLuc activity. (B) HeLa cells were transduced with Adv-CALuc or Adv-CADNIκ Bα at
an MOI of 5. Aer 48-h incubation, the E2 and E4 mRNA levels in the cells were determined by quantitative
RT-PCR. ese data (A,B) are expressed as the means ± S.D. (n = 4). (C) C57BL/6 mice were intravenously
administered 10
9
IFU of Adv-CALuc or Adv-CADNIκ Bα . Forty-eight hours aer the administration, the E2,
E4, and pIX mRNA levels in the mouse liver were determined by quantitative RT-PCR. e data are expressed
as the means ± S.E. (n = 5–6). **p < 0.01, ***p < 0.001.
www.nature.com/scientificreports/
10
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
Discussion
Several studies have demonstrated that the leaky expression of Ad genes from a replication-incompetent
Ad vector genome leads to tissue damages such as hepatotoxicities
7–9
; however, the mechanism of the E1A
gene-independent expression of Ad genes following transduction has remained unclear. e aim of this study
was to identify cellular factors involved in the E1A gene-independent expression of Ad genes and to provide
insights into the development of a safer Ad vector. e results showed that NF-κ B promoted Ad gene expression
following treatment with WT-Ad and an Ad vector (Figs2–4 and 6). Adv-CADNIκ Bα , which eciently inhibited
NF-κ B sig naling via overexpression of DNIκ Bα , also induced a signicant suppression of the leaky expression
of Ad genes (Fig.7). In addition, Adv-CADNIκ Bα did not lead to any apparent hepatotoxicity following admin-
istration (Fig.8).
is study demonstrated that NF-κ B is largely responsible for Ad gene expression. Several types of viruses,
including CMV and herpes simplex virus type-1 (HSV-1), utilize NF-κ B signaling to facilitate their replication
16
.
ese viruses possess NF-κ B binding sites in their viral gene promoters, and thus the NF-κ B signaling triggers the
enhancement of their viral gene expressions, leading to promotion of the viral replication. For example, infection
with CMV results in up-regulation of Sp1 activity, which induces NF-κ B activation. e activated NF-κ B triggers
the expression of CMV genes, leading to productive CMV infection
28
. HSV-1 also exploits NF-κ B signaling for
their ecient replication. HSV-1 infection-induced activation of NF-κ B signicantly promotes the HSV-1 rep-
lication via up-regulation of expression of the viral late proteins VP16 and gC
29,30
. On the other hand, there has
been no study reporting the relationship between NF-κ B and Ad replication, although Deryckere et al. reported
that NF-κ B up-regulated E3 gene expression. Rather, previous reports demonstrated that the adenoviral E1A
31
and E3/19K gene products
32
activated NF-κ B in Ad infection. Our present study showed that NF-κ B activation
enhanced Ad gene expression and Ad replication. ese ndings suggest that, like other viruses, Ad utilizes
NF-κ B, which is activated by Ad-mediated innate immunity, for its ecient replication.
Gribaudo et al. has previously demonstrated that IFN-α treatment inhibited enhancer activities of murine
CMV early genes via down-regulation of NF-κ B activities
33
. On the other hand, as shown in Figure S3, IFN-α
treatment signicantly inhibited NF-κ B signaling, Ad gene expression, and Ad replication. ese results suggest
that similarly to murine CMV, Ad infection would be also inhibited by IFN-α signaling via down-regulation of
NF-κ B activities. Further studies will be needed to clarify the mechanism underlying IFN-α -mediated suppres-
sion of Ad gene expression.
This study provided evidence that NF-κ B is largely involved in not only the gene expression of WT-Ad
but also leaky expression of the Ad genes from Ad vectors. Leaky expression of Ad genes is responsible for
Ad vector-mediated tissue damages, including hepatotoxicities. Several groups previously reported that Ad
vector-mediated hepatotoxicities were signicantly reduced by treatment with immunosuppressive agents such
as cyclosporine
34
and FK506
35
, permitting long-term expression of the transgene. In these studies, the diminished
Figure 8. Suppression of Ad vector-mediated hepatotoxicities by expression of a dominant-negative
IκBα gene. C57BL/6 mice were intravenously administered 10
9
IFU of Adv-CALuc or Adv-CADNIκ Bα . At
the indicated time points, the serum ALT and AST levels were determined. ese data are expressed as the
means ± S.E. (n = 5–6). (B,C) Rag2/IL2rgc double knockout mice were intravenously administered 10
9
IFU of
Adv-CALuc or Adv-CADNIκ Bα . (B) Forty-eight hours aer the administration, the E2 and E4 mRNA levels
in the mouse liver were determined by quantitative RT-PCR. ese data are expressed as the means ± S.E.
(n = 4). (C) At the indicated time points, the serum ALT and AST levels were determined. ese data (B,C)
are expressed as the means ± S.E. (n = 6–8). *p < 0.05, **p < 0.01, ***p < 0.001 (Adv-CALuc versus Adv-
CADNIκ Bα ).
www.nature.com/scientificreports/
11
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
hepatotoxicities by the immunosuppressive agents were considered to be mainly attributable to the suppression
of Ad vector-induced immune responses; however, immunosuppressive agents also inhibit NF-κ B activation,
leading to the suppression of leaky expression of Ad genes and the subsequent hepatotoxicities. is study showed
that the inhibition of NF-κ B via overexpression of DNIκ Bα resulted in a reduction of the Ad vector-mediated
hepatotoxicities (Fig.8).
As shown in Fig.5A, stimulation by recombinant TNF-α did not activate the E1A, E1B, and E4 promoters,
although the E2 and E3 promoter activities were signicantly enhanced by TNF-α treatment. e E2 and E3
promoter regions possess the NF-κ B binding site (Figs1B and 5B). On the other hand, in silico analysis predicted
no NF-κ B binding site in the E1A, E1B, and E4 promoter regions, suggesting that NF-κ B indirectly induced
the enhancement of the E1A, E1B, and E4 gene expressions. Lusky et al. reported that the DNA binding protein
encoded by the E2A gene is involved in the leaky expression of other Ad genes in Ad vector-transduced cells
4
. It
would be interesting to examine whether NF-κ B-induced E2 gene products are involved in other Ad gene expres-
sion. Alternatively, cellular factors which are activated by NF-κ B signaling would be involved in up-regulation
of Ad gene expression. Further studies will be needed to clarify the mechanism underlying NF-κ B-mediated
enhancement of Ad gene expression.
Although we demonstrated that Adv-CADNIκ Bα achieved a reduction in the expression levels of Ad genes,
and thereby induced no evident hepatotoxicity (Fig.8), previous studies have reported that an Ad vector-mediated
expression of Iκ Bα eciently inhibited NF-κ B activities, resulting in signicant apoptosis of hepatocytes fol-
lowed by severe hepatotoxicities
26,36
. Because NF-κ B functions as an inhibitor of apoptosis
37
, ecient inhibition
of NF-κ B by overexpression of DNIκ Bα induced the apoptosis of hepatocytes in these previous reports. In the
present study, 10
9
IFU of Adv-CADNIκ Bα was intravenously administered to mice, while the previous studies
evaluated the hepatotoxicities following administration with Ad vectors expressing Iκ Bα at doses of more than
4 × 10
9
IFU
26,36
. ese dierences in the injection doses were likely responsible for the dierences in hepatotox-
icity levels.
In order to suppress or eliminate the leaky expression of Ad genes, various types of replication-incompetent Ad
vectors have been developed
3,4,8
. Following transduction with E2 and/or E4-deleted Ad vectors, the leaky expres-
sion of other Ad proteins was signicantly suppressed, leading to decreases of cytotoxic T lymphocyte (CTL)
responses and hepatotoxicities
3,4,8
. In addition, an HD-Ad vector, in which almost all viral coding sequences were
deleted, has been developed
11–13
. is HD-Ad vector achieved a reduction in inammatory cytokine production
and tissue damages, leading to persistent transgene expression in vivo. However, in vivo application of HD-Ad
vectors has been limited due to the low titers of HD-AD vectors and complicated production method, which
requires special packaging cells and a helper virus for the propagation of HD-Ad vectors. As shown in Fig.8
Adv-CADNIκ Bα , which was propagated to a high titer in a conventional method, mediated the reduced leaky
expression of Ad genes and hepatotoxicities. In addition, Ad vector-induced inammatory immune responses are
suppressed by the overexpression of DNIκ Bα . An Ad vector carrying an expression cassette of DNIκ Bα would be
a promising framework for safer and more ecient Ad vectors, although we should pay attention to the injection
dose, as described above.
In summary, we have demonstrated that NF-κ B induces the leaky expression of Ad genes, and that inhibition
of NF-κ B leads to suppression of Ad vector-mediated hepatotoxicities by not only Ad vector-induced immune
responses but also the Ad proteins themselves. is study provides important clues toward the development of
promising Ad vectors.
References
1. Volpers, C. & ochane, S. Adenoviral vectors for gene transfer and therapy. J Gene Med 6, S164–171 (2004).
2. McConnell, M. J. & Imperiale, M. J. Biology of adenovirus and its use as a vector for gene therapy. Hum Gene er 15, 1022–1033
(2004).
3. Dedieu, J. F. et al. Long-term gene delivery into the livers of immunocompetent mice with E1/E4-defective adenoviruses. J Virol 71,
4626–4637 (1997).
4. Lusy, M. et al. In vitro and in vivo biology of recombinant adenovirus vectors with E1, E1/E2A, or E1/E4 deleted. J Virol 72,
2022–2032 (1998).
5. Shimizu, ., Saurai, F., Machitani, M., atayama, . & Mizuguchi, H. Quantitative analysis of the leay expression of adenovirus
genes in cells transduced with a replication-incompetent adenovirus vector. Mol Pharm 8, 1430–1435 (2011).
6. Waabayashi, . et al. Quantitative analysis of virus-associated NAI expression following transduction with a replication-
incompetent adenovirus vector in vitro and in vivo. J Mol Genet Med 9, 1000169 (2015).
7. Yang, Y. et al. Cellular immunity to viral antigens limits E1-deleted adenoviruses for gene therapy. Proc Natl Acad Sci USA 91,
4407–4411 (1994).
8. Gao, G. P., Yang, Y. & Wilson, J. M. Biology of adenovirus vectors with E1 and E4 deletions for liver-directed gene therapy. J Virol 70,
8934–8943 (1996).
9. Shimizu, . et al. Suppression of leay expression of adenovirus genes by insertion of microNA-targeted sequences in the
replication-incompetent adenovirus vector genome. Mol er Methods Clin Dev 1, 14035 (2014).
10. Zhou, H., O’Neal, W., Morral, N. & Beaudet, A. L. Development of a complementing cell line and a system for construction of
adenovirus vectors with E1 and E2a deleted. J Virol 70, 7030–7038 (1996).
11. eddy, P. S. et al. Sustained human factor VIII expression in hemophilia A mice following systemic delivery of a gutless adenoviral
vector. Mol er 5, 63–73 (2002).
12. Mian, A. et al. Toxicity and adaptive immune response to intracellular transgenes delivered by helper-dependent vs. rst generation
adenoviral vectors. Mol Genet Metab 84, 278–288 (2005).
13. Machitani, M. et al. Improving adenovirus vector-mediated NAi eciency by lacing the expression of virus-associated NAs.
Viruses 178, 357–363 (2013).
14. Saurai, H., awabata, ., Saurai, F., Naagawa, S. & Mizuguchi, H. Innate immune response induced by gene delivery vectors. Int
J Pharm 354, 9–15 (2008).
15. Chen, D., Murphy, B., Sung, . & Bromberg, J. S. Adaptive and innate immune responses to gene transfer vectors: role of cytoines
and chemoines in vector function. Gene er 10, 991–998 (2003).
16. Hiscott, J., won, H. & Genin, P. Hostile taeovers: viral appropriation of the NF-appaB pathway. J Clin Invest 107, 143–151 (2001).
www.nature.com/scientificreports/
12
Scientific RepoRts | 6:19922 | DOI: 10.1038/srep19922
17. Shinai, Y. et al. AG-2-decient mice lac mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell 68, 855–867
(1992).
18. Cao, X. et al. Defective lymphoid development in mice lacing expression of the common cytoine receptor gamma chain. Immunity
2, 223–238 (1995).
19. Suzui, T. et al. mi-122a-regulated expression of a suicide gene prevents hepatotoxicity without altering antitumor eects in suicide
gene therapy. Mol er 16, 1719–1726 (2008).
20. Saurai, F., awabata, ., Yamaguchi, T., Hayaawa, T. & Mizuguchi, H. Optimization of adenovirus serotype 35 vectors for ecient
transduction in human hematopoietic progenitors: comparison of promoter activities. Gene er 12, 1424–1433 (2005).
21. Machitani, M. et al. Development of an adenovirus vector lacing the expression of virus-associated NAs. J Control elease 154,
285–289 (2011).
22. Mizuguchi, H. & ay, M. A. Ecient construction of a recombinant adenovirus vector by an improved in vitro ligation method.
Hum Gene er 9, 2577–2583 (1998).
23. Mizuguchi, H. & ay, M. A. A simple method for constructing E1- and E1/E4-deleted recombinant adenoviral vectors. Hum Gene
er 10, 2013–2017 (1999).
24. awabata, ., Saurai, F., Yamaguchi, T., Hayaawa, T. & Mizuguchi, H. Ecient gene transfer into mouse embryonic stem cells with
adenovirus vectors. Mol er 12, 547–554 (2005).
25. Matsui, H. et al. A targeted adenovirus vector displaying a human bronectin type III domain-based monobody in a ber protein.
Biomaterials 34, 4191–4201 (2013).
26. Lieber, A. et al. Inhibition of NF-appaB activation in combination with bcl-2 expression allows for persistence of rst-generation
adenovirus vectors in the mouse liver. J Virol 72, 9267–9277 (1998).
27. Zhang, Y. et al. Acute cytoine response to systemic adenoviral vectors in mice is mediated by dendritic cells and macrophages. Mol
er 3, 697–707 (2001).
28. Yurocho, A. D., Mayo, M. W., Poma, E. E., Baldwin, A. S., Jr. & Huang, E. S. Induction of the transcription factor Sp1 during human
cytomegalovirus infection mediates upregulation of the p65 and p105/p50 NF-appaB promoters. J Virol 71, 4638–4648 (1997).
29. Patel, A. et al. Herpes simplex type 1 induction of persistent NF-appa B nuclear translocation increases the eciency of virus
replication. Virology 247, 212–222 (1998).
30. Gregory, D., Hargett, D., Holmes, D., Money, E. & Bachenheimer, S. L. Ecient replication by herpes simplex virus type 1 involves
activation of the IappaB inase-IappaB-p65 pathway. J Virol 78, 13582–13590 (2004).
31. Shurman, L., Sen, . & Bergman, Y. Adenovirus E1A products activate the Ig -chain enhancer in broblasts. A possible involvement
of the NF-B binding site. J Immunol 143, 3806–3812 (1989).
32. Pahl, H. L., Sester, M., Burgert, H. G. & Baeuerle, P. A. Activation of transcription factor NF-appaB by the adenovirus E3/19
protein requires its E retention. J Cell Biol 132, 511–522 (1996).
33. Gribaudo, G. et al. Interferon-alpha inhibits the murine cytomegalovirus immediate-early gene expression by down-regulating NF-
appa B activity. Virology 211, 251–260 (1995).
34. Engelhardt, J. F., Ye, X., Doranz, B. & Wilson, J. M. Ablation of E2A in recombinant adenoviruses improves transgene persistence
and decreases inammatory response in mouse liver. Proc Natl Acad Sci USA 91, 6196–6200 (1994).
35. Ilan, Y. et al. Transient immunosuppression with F506 permits long-term expression of therapeutic genes introduced into the liver
using recombinant adenoviruses in the rat. Hepatology 26, 949–956 (1997).
36. Iimuro, Y. et al. NFappaB prevents apoptosis and liver dysfunction during liver regeneration. J Clin Invest 101, 802–811 (1998).
37. Luo, J. L., amata, H. & arin, M. I/NF-appaB signaling: balancing life and death–a new approach to cancer therapy. J Clin
Invest 115, 2625–2632 (2005).
Acknowledgements
e authors declare no conict of interest. We thank Sayuri Okamoto and Eri Hosoyamada (Graduate School of
Pharmaceutical Sciences, Osaka University, Osaka, Japan) for their help. is work was supported by grants-in-
aid for Scientic Research (A) and (B) from the Ministry of Education, Culture, Sports, Science, and Technology
(MEXT) of Japan, and by a grant-in-aid from the Ministry of Health, Labor, and Welfare (MHLW) of Japan and
Japan Agency for Medical Research and Development (AMED).
Author Contributions
M.M. designed and performed the experiments, analyzed data, and wrote the manuscript; F.S. designed and
supervised the projects, analyzed data, and wrote the manuscript; K.W., K.N. and K.S. supported the experiments;
M.T. analyzed data. H.M. supervised the projects, interpreted data, and wrote the manuscript.
Additional Information
Supplementary information accompanies this paper at http://www.nature.com/srep
Competing nancial interests: e authors declare no competing nancial interests.
How to cite this article: Machitani, M. et al. NF-κB promotes leaky expression of adenovirus genes in a
replication-incompetent adenovirus vector. Sci. Rep. 6, 19922; doi: 10.1038/srep19922 (2016).
is work is licensed under a Creative Commons Attribution 4.0 International License. e images
or other third party material in this article are included in the article’s Creative Commons license,
unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license,
users will need to obtain permission from the license holder to reproduce the material. To view a copy of this
license, visit http://creativecommons.org/licenses/by/4.0/

Supplementary resource (1)

... We recently demonstrated that nuclear factor-kB (NF-kB) promotes not only the leaky expression of Ad genes following transduction with a replication-incompetent Ad vector, but also Ad gene expression following infection with a wild-type Ad. 17 NF-kB is a ubiquitous transcriptional factor that promotes the expression of a large number of genes, particularly gene families associated with host immune responses. 18 NF-kB also plays a crucial role in the expression of numerous viral genes. ...
... In our earlier study mentioned above, we constructed a replicationincompetent Ad vector expressing a dominant-negative IkBa (DNIkBa), which is a negative regulator of NF-kB. 17 An Ad vector expressing DNIkBa showed the significant inhibition of NF-kB signaling and suppressed the NF-kB-mediated leaky expression of Ad genes, leading to a significant suppression of Ad vector-mediated hepatotoxicity following systemic administration. These results led us to hypothesize that TRAD-mediated hepatotoxicity is also circumvented by suppressing the Ad gene expression in the liver via the liver-specific expression of DNIkBa. ...
... We previously demonstrated that NF-kB promotes a leaky expression of Ad genes following transduction with recombinant Ads. 17 In addition, NF-kB binds to an hTERT promoter. 19 To determine whether NF-kB enhances Ad gene expression and the replication of TRAD in hepatocytes, in the present study we pretreated PH5CH8 cells 20 (a non-neoplastic human hepatocyte cell line) with an NF-kB inhibitor BAY11-7082 and a proteasome inhibitor MG-132, followed by infection with the conventional TRAD. ...
Article
Full-text available
Telomerase-specific replication-competent adenoviruses (Ads), i.e., TRADs, which possess an E1 gene expression cassette driven by the human telomerase reverse transcriptase promoter, are promising agents for cancer treatment. However, even though oncolytic Ads, including TRAD, are intratumorally administered, they are disseminated from the tumor to systemic circulation, causing concern about oncolytic Ad-mediated hepatotoxicity (due mainly to leaky expression of Ad genes in liver). We reported that inhibition of nuclear factor-κB (NF-κB) leads to the suppression of replication-incompetent Ad vector-mediated hepatotoxicity via reduction of the leaky expression of Ad genes in liver. Here, to develop a TRAD with an improved safety profile, we designed a TRAD that carries a liver-specific promoter-driven dominant-negative IκBα (DNIκBα) expression cassette (TRAD-DNIκBα). Compared with a conventional TRAD, TRAD-DNIκBα showed hepatocyte-specific inhibition of NF-κB signaling and significantly reduced Ad gene expression and replication in the normal human hepatocyte cell line. TRAD-induced hepatotoxicity was largely suppressed in mice following intravenous administration of TRAD-DNIκBα. However, the replication profiles and oncolytic activities of TRAD-DNIκBα were comparable with those of the conventional TRAD in human non-hepatic tumor cells. These results indicate that oncolytic Ads containing the liver-specific DNIκBα expression cassette have improved safety profiles without inhibiting oncolytic activities.
... Importantly, Adv-CADNIκBα did not induce apparent hepatotoxicities. These results indicate that the inhibition of NF-κB results in the suppression of adenoviral vector-mediated tissue damage by both suppression of inflammatory responses and reduction in the leaky expression of adenoviral genes [127]. ...
... Although originally being locally applied, a limited local dissemination of adenoviral vaccines, e.g., via binding to endothelial cells [134][135][136] together with the described NF-κB-triggered leaky expression of adenovirus genes in originally replication-incompetent adenoviral vectors [127] may result in rare cases to self-amplifying cascades resulting in activated or damaged endothelial cells, activated and aggregated platelets, and activation of the coagulation system at sites distant to the application site, i.e., systemic prothrombotic procoagulation events together with a corresponding (consumption) thrombocytopenia as observed in rare cases of adenovirus vector-based SARS-CoV-2 vaccines [39][40][41][42][43][44]. ...
Article
Full-text available
Novel coronavirus SARS-CoV-2 has resulted in a global pandemic with worldwide 6-digit infection rates and thousands of death tolls daily. Enormous efforts are undertaken to achieve high coverage of immunization to reach herd immunity in order to stop the spread of SARS-CoV-2 infection. Several SARS-CoV-2 vaccines based on mRNA, viral vectors, or inactivated SARS-CoV-2 virus have been approved and are being applied worldwide. However, the recent increased numbers of normally very rare types of thromboses associated with thrombocytopenia have been reported, particularly in the context of the adenoviral vector vaccine ChAdOx1 nCoV-19 from Astra Zeneca. The statistical prevalence of these side effects seems to correlate with this particular vaccine type, i.e., adenoviral vector-based vaccines, but the exact molecular mechanisms are still not clear. The present review summarizes current data and hypotheses for molecular and cellular mechanisms into one integrated hypothesis indicating that coagulopathies, including thromboses, thrombocytopenia, and other related side effects, are correlated to an interplay of the two components in the vaccine, i.e., the spike antigen and the adenoviral vector, with the innate and immune systems, which under certain circumstances can imitate the picture of a limited COVID-19 pathological picture.
... Another concern pertains to the leaky expression from the donor DNA template plasmid, which ideally should be completely suppressed in the absence of Cas ( 8 ). Furthermore, residue viral elements in vectors may contribute to cellular toxicity through leaky expression ( 9 ). Despite attracting significant attention, the challenge of leaky expression from viral-delivered HDR donor templates persists. ...
Article
Full-text available
Precise genomic editing through the combination of CRISPR/Cas systems and recombinant adeno-associated virus (rAAV)-delivered homology directed repair (HDR) donor templates represents a powerful approach. However, the challenge of effectively suppressing leaky transcription from the rAAV vector, a phenomenon associated to cytotoxicity, persists. In this study, we demonstrated substantial promoter activities of various homology arms and inverted terminal repeats (ITR). To address this issue, we identified a novel rAAV variant, Y704T, which not only yields high-vector quantities but also effectively suppresses in cis mRNA transcription driven by a robust promoter. The Y704T variant maintains normal functionality in receptor interaction, intracellular trafficking, nuclear entry, uncoating, and second-strand synthesis, while specifically exhibiting defects in transcription. Importantly, this inhibitory effect is found to be independent of ITR, promoter types, and RNA polymerases. Mechanistic studies unveiled the involvement of Valosin Containing Protein (VCP/p97) in capsid-mediated transcription repression. Remarkably, the Y704T variant delivers HDR donor templates without compromising DNA replication ability and homologous recombination efficiency. In summary, our findings enhance the understanding of capsid-regulated transcription and introduce novel avenues for the application of the rAAV-CRISPR/Cas9 system in human gene therapy.
... NF-κB activation was equally important for the expression of the chemokine CXCL10 induced by HAdV5 infection in kidney epithelium-derived cells [38]. Interestingly, NF-κB was also found to mediate the leaky expression of adenovirus genes from the HAdV5 vector genome and the inhibition of NF-κB leads to the suppression of HAdV5 gene expression and hepatotoxicity following transduction with HAdV5-based vectors [39]. In this study, NF-κB inhibition decreased HAdV26-induced IL-6 expression, which could potentially correlate to decreased toxicity. ...
Article
Full-text available
The low seroprevalent human adenovirus type 26 (HAdV26)-based vaccine vector was the first adenovirus-based vector to receive marketing authorization from European Commission. HAdV26-based vaccine vectors induce durable humoral and cellular immune responses and, as such, represent a highly valuable tool for fighting infectious diseases. Despite well-described immunogenicity in vivo, the basic biology of HAdV26 still needs some refinement. The aim of this study was to determine the pro-inflammatory cytokine profile of epithelial cells infected with HAdV26 and then investigate the underlying molecular mechanism. The expression of studied genes and proteins was assessed by quantitative polymerase chain reaction, western blot, and enzyme-linked immunosorbent assay. Confocal microscopy was used to visualize HAdV26 cell uptake. We found that HAdV26 infection in human epithelial cells triggers the expression of pro-inflammatory cytokines and chemokines, namely IL-6, IL-8, IL-1β, and TNF-α, with the most pronounced difference shown for IL-6. We investigated the underlying molecular mechanism and observed that HAdV26-induced IL-6 gene expression is αvβ3 integrin dependent and NF-κB mediated. Our findings provide new data regarding pro-inflammatory cytokine and chemokine expression in HAdV26-infected epithelial cells, as well as details concerning HAdV26-induced host signaling pathways. Information obtained within this research increases our current knowledge of HAdV26 basic biology and, as such, can contribute to further development of HAdV26-based vaccine vectors.
... This supports previous research which implies heat shock (and thus increased expression of heat shock proteins) does not overcome the lack of E1A expression [39] as in MRC5 cells we do not see increasing amounts of ChA-dOx1 vector backbone transcription. In addition, NFκB has been implicated in enabling HuAd5-based vector backbone expression in non-permissive cell lines [40]. However, no marked differences were observed between the two cell lines in the transcriptomic abundance data for NFKB1 in our datasets (Additional file 15: Table S12 and Additional file 16: Table S13). ...
Article
Full-text available
Background ChAdOx1 nCoV-19 is a recombinant adenovirus vaccine against SARS-CoV-2 that has passed phase III clinical trials and is now in use across the globe. Although replication-defective in normal cells, 28 kbp of adenovirus genes is delivered to the cell nucleus alongside the SARS-CoV-2 S glycoprotein gene. Methods We used direct RNA sequencing to analyse transcript expression from the ChAdOx1 nCoV-19 genome in human MRC-5 and A549 cell lines that are non-permissive for vector replication alongside the replication permissive cell line, HEK293. In addition, we used quantitative proteomics to study over time the proteome and phosphoproteome of A549 and MRC5 cells infected with the ChAdOx1 nCoV-19 vaccine. Results The expected SARS-CoV-2 S coding transcript dominated in all cell lines. We also detected rare S transcripts with aberrant splice patterns or polyadenylation site usage. Adenovirus vector transcripts were almost absent in MRC-5 cells, but in A549 cells, there was a broader repertoire of adenoviral gene expression at very low levels. Proteomically, in addition to S glycoprotein, we detected multiple adenovirus proteins in A549 cells compared to just one in MRC5 cells. Conclusions Overall, the ChAdOx1 nCoV-19 vaccine’s transcriptomic and proteomic repertoire in cell culture is as expected. The combined transcriptomic and proteomics approaches provide a detailed insight into the behaviour of this important class of vaccine using state-of-the-art techniques and illustrate the potential of this technique to inform future viral vaccine vector design.
... This supports previous research which implies heat shock (and thus increased expression of heat shock proteins) does not overcome the lack of E1A expression 36 as in MRC5 cells we do not see increasing amounts of ChAdOx1 vector backbone transcription. In addition, NFκB has been implicated in enabling HuAd5 based vector backbone expression in non-permissive cell lines 37 . However, no marked differences were observed between the two cell lines in the transcriptomic abundance data for NFKB1 in our datasets (Additional les 15 and 16). ...
Preprint
Full-text available
Background: ChAdOx1 nCoV-19 is a recombinant adenovirus vaccine candidate against SARS-CoV-2. Although replication defective in normal cells, 28kbp of adenovirus genes are delivered to the cell nucleus alongside the SARS-CoV-2 S glycoprotein gene. Methods: We used direct RNA sequencing to analyse transcript expression from the ChAdOx1 nCoV-19 genome in human MRC-5 and A549 cell lines that are non-permissive for vector replication alongside the replication permissive cell line, HEK293. In addition, we used quantitative proteomics to study over time the proteome and phosphoproteome of A549 and MRC5 cells infected with the ChAdOx1 nCoV-19 vaccine candidate. Results: The expected SARS-CoV-2 S coding transcript dominated in all cell lines. We also detected rare S transcripts with aberrant splice patterns or polyadenylation site usage. Adenovirus vector transcripts were almost absent in MRC-5 cells but in A549 cells there was a broader repertoire of adenoviral gene expression at very low levels. Proteomically, in addition to S glycoprotein, we detected multiple adenovirus proteins in A549 cells compared to just one in MRC5 cells. Conclusions: Overall the ChAdOx1 nCoV-19 vaccine’s transcriptomic and proteomic repertoire is as expected. The combined transcriptomic and proteomics approaches provide an unparalleled insight into the behaviour of this important class of vaccine candidate and illustrate the potential of this technique to inform future viral vaccine vector design.
... Total DNA, including Ad genomic DNA, was isolated from cells infected with Ad by use of a DNeasy blood and tissue kit (Qiagen). After isolation, the copy number of Ad genomic DNA was quantified using the StepOnePlus real-time PCR system (Life Technologies) as previously described (59). The sequences of the primers and probes used in this study are provided in Table 2. ...
Article
Full-text available
Recent studies have reported that host microRNAs (miRNAs) regulate infections by several types of viruses via various mechanisms and that inhibition of the miRNA processing factors enhances or prevents viral infection. However, it has not been clarified whether these effects of miRNAs extend to adenovirus (Ad) infection. Here we show that miR-27a and -b efficiently inhibit infection with an Ad via the downregulation of SNAP25 and TXN2, which are members of the SNARE proteins and the thioredoxin family, respectively. Approximately 80% reductions in Ad genomic copy number were found in cells transfected with miR-27a/b mimics, whereas there were approximately 2.5- to 5-fold larger copy numbers of the Ad genome following transfection with miR-27a/b inhibitors. Microarray gene expression analysis and in silico analysis demonstrated that SNAP25 and TXN2 are target genes of miR-27a/b. A reporter assay using plasmids containing the 3′ untranslated regions of the SNAP25 and TXN2 genes showed that miR-27a/b directly suppressed SNAP25 and TXN2 expression through posttranscriptional gene silencing. Knockdown of SNAP25 led to a significant inhibition of Ad entry into cells. Knockdown of TXN2 induced cell cycle arrest at G1 phase, leading to a reduction in Ad replication. In addition, overexpression of Ad-encoded small noncoding RNAs (VA-RNAs) restored the miR-27a/b-mediated reduction in infection level with a VA-RNA-lacking Ad mutant due to the VA-RNA-mediated inhibition of miR-27a/b expression. These results indicate that miR-27a and -b suppress SNAP25 and TXN2 expression via posttranscriptional gene silencing, leading to efficient suppression of Ad infection. IMPORTANCE Adenovirus (Ad) is widely used as a platform for replication-incompetent Ad vectors (Adv) and replication-competent oncolytic Ad (OAd) in gene therapy and virotherapy. Regulation of Ad infection is highly important for efficient gene therapies using both Adv and OAd. In this study, we demonstrate that miR-27a and -b, which are widely expressed in host cells, suppress SNAP25 and TXN2 expression through posttranscriptional gene silencing. Suppression of SNAP25 and TXN2 expression leads to inhibition of Ad entry into cells and to cell cycle arrest, respectively, leading to efficient suppression of Ad infection. Our findings provide important clues to the improvement of gene therapies using both Adv and OAd.
Article
Reovirus induces tumor cell death efficiently and specifically, and thus is currently undergoing clinical testing as an anticancer agent. In the intracellular trafficking of reovirus, proteolytic disassembly of reovirus capsid-proteins and subsequent penetration of viral particles into the cytosol are crucial steps. Cathepsins B and L are largely responsible for the proteolytic disassembly of reovirus. Reovirus efficiently lyses tumor cells exhibiting relatively high activities of cathepsins B and L, while tumor cells with low activities of cathepsins B and L are often refractory to reovirus, probably due to inefficient endo/lysosomal escape. In this study, in order to enhance the tumor cell-killing efficiencies of reovirus by promoting endo/lysosomal escape, especially in reovirus-resistant tumor cells, reovirus was complexed with a cationic liposome transfection reagent. Reovirus alone and reovirus-cationic liposome complex (reoplex) exhibited similar levels of tumor cell-killing efficiencies in reovirus-susceptible tumor cells, while reoplex mediated more than 30% higher levels of tumor cell-killing activities in reovirus-resistant tumor cells than reovirus alone. Reoplex-mediated tumor cell death was efficiently induced in the tumor cells pretreated with cathepsin inhibitors. The mRNA levels of interferon (IFN)-β and apoptotic genes were significantly elevated following reoplex treatment. These results suggest that cationic liposomes efficiently promoted delivery of reovirus to the cytosol, leading to induction of apoptosis.
Article
Full-text available
Leaky expression of adenovirus (Ad) genes occurs following transduction with a conventional replication-incompetent Ad vector, leading to an induction of cellular immunity against Ad proteins and Ad protein-induced toxicity, especially in the late phase following administration. To suppress the leaky expression of Ad genes, we developed novel Ad vectors by incorporating four tandem copies of sequences with perfect complementarity to miR-122a or miR-142-3p into the 3'-untranslated region (UTR) of the E2A, E4, or pIX gene, which were mainly expressed from the Ad vector genome after transduction. These Ad vectors easily grew to high titers comparable to those of a conventional Ad vector in conventional 293 cells. The leaky expression of these Ad genes in mouse organs was significantly suppressed by 2- to 100-fold, compared with a conventional Ad vector, by insertion of the miRNA-targeted sequences. Notably, the Ad vector carrying the miR-122a-targeted sequences into the 3'-UTR of the E4 gene expressed higher and longer-term transgene expression and more than 20-fold lower levels of all the Ad early and late genes examined in the liver than a conventional Ad vector. miR-122a-mediated suppression of the E4 gene expression in the liver significantly reduced the hepatotoxicity which an Ad vector causes via both adaptive and non-adaptive immune responses.
Article
Full-text available
Several studies have reported that short hairpin RNA (shRNA)-mediated RNA interference (RNAi) was competitively inhibited by the expression of adenovirus (Ad)-encoded small RNAs (VA-RNAs), which are expressed from a replication-incompetent Ad vector, as well as a wild-type Ad; however, it remained to be clarified whether an shRNA-expressing Ad vector-mediated knockdown was inhibited by VA-RNAs transcribed from the same Ad vector genome. In this study, we demonstrated that a lack of VA-RNA expression from the Ad vector leads to an increase in knockdown efficiencies of Ad vector-mediated RNAi. In the cells transduced with a first-generation Ad vector (FG-Ad) expressing shRNA (FG-Ad-shRNA), the copy numbers of shRNA and VA-RNAs incorporated into the RNA-induced silencing complex (RISC) was comparable. In contrast, higher amounts of shRNA were found in the RISC when the cells were transduced with an shRNA-expressing helper-dependent Ad (HD-Ad) vector, in which all viral genes, including VA-RNAs, were deleted (HD-Ad-shRNA), compared with FG-Ad-shRNA. HD-Ad vectors expressing shRNA against luciferase and p53 showed 7.4% and 37.3% increases in the knockdown efficiencies compared to the corresponding FG-Ad-shRNA, respectively, following in vitro transduction. Furthermore, higher levels of knockdown efficiencies were also found by the transduction with shRNA-expressing Ad vectors lacking VA-RNA expression (AdΔVR-shRNA) than by transduction with FG-Ad-shRNA. These results indicate that VA-RNAs expressed from an Ad vector inhibit knockdown by the shRNA-expressing Ad vector and that HD-Ad-shRNA and AdΔVR-shRNA are a powerful framework for shRNA-mediated knockdown.
Article
Full-text available
Hematopoietic stem cells (HSCs) have the potential for self-renewal and multilineage differentiation into all mature blood cells. Hence, efficient transduction into HSCs would afford the opportunity to treat a number of diseases that result from abnormal blood cell function, and would be a powerful tool for study of the regulation of proliferation, differentiation, and trafficking of HSCs. We have previously reported that an adenovirus (Ad) vector composed of whole Ad serotype 35 (Ad35), which belongs to subgroup B, shows efficient gene transfer into human bone marrow CD34+ cells (Gene Ther., 2003, 10, 1041-1048). However, Ad35 vector-mediated transduction into human hematopoietic stem cells/progenitors has not yet been fully optimized. In the present study, we have systemically examined promoter activity in the context of Ad35 vectors in human bone marrow CD34+ cells and primitive CD34+ subsets to optimize the transduction efficiency in human hematopoietic stem/progenitor cells. In the first of the transduction experiments, the improved in vitro ligation method, which was developed by Mizuguchi and Kay (Hum.Gene Ther., 1998, 9, 2577-2583, Hum.Gene Ther., 1999, 10, 2013-2017), was applied to Ad35 vector construction to allow for simple and efficient production of an E1/E3-deleted Ad35 vector. Using this method, we constructed a series of Ad35 vectors encoding the enhanced green fluorescence protein (GFP) under the control of a variety of strong viral and cellular promoters, including the human cytomegalovirus immediate early region promoter/enhancer (CMV promoter), the human elongation factor 1α promoter (EF1α promoter), the CMV immediate early enhancer/the chicken β-actin promoter with the β-actin intron sequence (CA promoter), the mouse phosphoglycerate kinase 1 promoter (PGK promoter), the murine stem cell virus (MSCV) long terminal repeat (LTR) promoter (MSCV promoter), and the CMV promoter/enhancer containing the largest intron of CMV (intron A) (CMVi promoter). Of the six types of promoters tested, significantly higher transduction efficiencies were achieved with the EF1α promoter, the CA promoter, and the CMVi promoter in the human CD34+ cells and the immature subsets (CD34+CD38low/- and CD34+AC133+ subsets). In particular, the CA promoter was found to allow for the highest transduction efficiencies in both the whole human CD34+ cells (53.8 % GFP-positive) and the immature hematopoietic subsets (57 % GFP-positive for CD34+CD38low/- subsets, 51 % GFP-positive for CD34+AC133+ subsets). Furthermore, a colony-forming assay demonstrated that the CA promoter-mediated GFP-expressing cells differentiated into progenitor cells of all lineages. These results provide an optimized Ad35 vector backbone for efficient transduction into hematopoietic stem cells/progenitors.
Article
Full-text available
We investigated the cellular basis for secretion of inflammatory cytokines in mice following intravenous administration of adenoviral vectors (Ad). Serum inflammatory cytokines including interleukin-6 (IL-6), IL-12, and tumor necrosis factor-alpha (TNF-) were detected as early as 6 h following intravenous injection of Ad-expressing Escherichia coli -galactosidase (Ad-lacZ). Ad-lacZ readily accumulated in the splenic marginal zone 1 h after intravenous infusion, where both dendritic cells (DCs) and macrophages were transduced and activated within 6 h. Flow cytometric analyses showed that the expression of Ia and CD86 antigens was markedly enhanced on splenic DCs indicating their activation in vivo by Ad-lacZ. Upon ex vivo culture, these early-activated splenic DCs spontaneously produced high levels of IL-6 and IL-12. By contrast, activated splenic macrophages spontaneously secreted only IL-6. Elimination of tissue macrophages and splenic DCs in vivo considerably reduced the early release of IL-12, IL-6, and TNF- and significantly blocked the specific cellular immune response to Ad and the transgene product in vivo. Our findings indicate that preferential activation of DCs and macrophages may account for Ad-triggered acute inflammatory response in vivo in mice. Moreover, DCs and macrophages may play different roles in this process in terms of their abilities to produce distinct patterns of inflammatory cytokines.Keywords: adenovirus, gene therapy, innate immunity, inflammatory cytokines, antigen-presenting cells
Article
Full-text available
Isogenic, E3-deleted adenovirus vectors defective in E1, E1 and E2A, or E1 and E4 were generated in complementation cell lines expressing E1, E1 and E2A, or E1 and E4 and characterized in vitro and in vivo. In the absence of complementation, deletion of both E1 and E2A completely abolished expression of early and late viral genes, while deletion of E1 and E4 impaired expression of viral genes, although at a lower level than the E1/E2A deletion. The in vivo persistence of these three types of vectors was monitored in selected strains of mice with viral genomes devoid of transgenes to exclude any interference by immunogenic transgene-encoded products. Our studies showed no significant differences among the vectors in the short-term maintenance and long-term (4-month) persistence of viral DNA in liver and lung cells of immunocompetent and immunodeficient mice. Furthermore, all vectors induced similar antibody responses and comparable levels of adenovirus-specific cytotoxic T lymphocytes. These results suggest that in the absence of transgenes, the progressive deletion of the adenovirus genome does not extend the in vivo persistence of the transduced cells and does not reduce the antivirus immune response. In addition, our data confirm that, in the absence of transgene expression, mouse cellular immunity to viral antigens plays a minor role in the progressive elimination of the virus genome.
Article
We have recently shown that the accumulation of diverse viral and cellular membrane proteins in the ER activates the higher eukaryotic transcription factor NF-kappaB. This defined a novel ER-nuclear signal transduction pathway, which is distinct from the previously described unfolded protein response (UPR). The well characterized UPR pathway is activated by the presence of un- or malfolded proteins in the ER. In contrast, the ER stress signal which activates the NF-kappaB pathway is not known. Here we used the adenovirus early region protein E3/19K as a model to investigate the nature of the NF-kappaB-activating signal emitted by the ER. E3/19K resides in the endoplasmic reticulum where it binds to MHC class I molecules, thereby preventing their transport to the cell surface. It is maintained in the ER by a retention signal sequence in its carboxy terminus, which causes the protein to be continuously retrieved to the ER from post-ER compartments. Mutation of this sequence allows E3/19K to reach the cell surface. We show here that expression of E3/19K potently activates a functional NF-kappaB transcription factor. The activated NF-kappaB complexes contained p50/p65 and p50/c-rel heterodimers. E3/19K interaction with MHC class I was not important for NF-kappaB activation since mutant proteins which no longer bind MHC molecules remained fully capable of inducing NF-kappaB. However, activation of both NF-kappaB DNA binding and kappaB-dependent transactivation relied on E3/19K ER retention: mutants, which were expressed on the cell surface, could no longer activate the transcription factor. This identifies the NF-kappaB-activating signal as the accumulation of proteins in the ER membrane, a condition we have termed "ER overload." We show that ER overload-mediated NF-kappaB activation but not TNF-stimulated NF-kappaB induction can be inhibited by the intracellular Ca2+ chelator TMB-8. Moreover, treatment of cells with two inhibitors of the ER-resident Ca(2+) -dependent ATPase, thapsigargin and cyclopiazonic acid, which causes a rapid release of Ca2+ from the ER, strongly activated NF-kappaB. We therefore propose that ER overload activates NF-kappaB by causing Ca2+ release from the ER. Because NF-kappaB plays a key role in mounting an immune response, ER overload caused by viral proteins may constitute a simple antiviral response with broad specificity.
Article
The host immune response limits the duration of expression of adenovirally transduced genes and precludes long-term gene expression upon re-administration of the virus. In this study we wished to evaluate whether short-term immunosuppression of the host, at the time of recombinant virus administration, would allow expression of the therapeutic gene product upon virus reinjection. Gunn rats were used as recipients of recombinant adenoviruses expressing human BUGT (Ad-hBUGT) or E. coli beta-galactosidase (Ad-LacZ). Rats were treated with FK506 (1-1.5 mg/kg, per OS daily) for three days beginning 24 hours before each virus injection. Control groups did not receive any immunosuppressant. The serum bilirubin level was reduced from 7.1 +/- 0.75 mg/dL to 2.0 +/- 0.7 mg/dL within two days of viral injection in both FK506 treated and control groups, and then gradually increased in 6 weeks. FK506-treated rats had low or undetectable antibody titers against the recombinant adenovirus and minimal or no cytotoxic T lymphocyte (CTL) response against adenovirus-infected cells. The tolerized rats received two subsequent injections 42 and 98 days after the first injection, which reduced the bilirubin levels again to 2.0 +/- 0.56 and 2.2 +/- 0.61 mg/dL, respectively. In contrast, control rats developed high titer neutralizing antibodies and a CTL response, and their serum bilirubin levels were not reduced following subsequent injections. We conclude that short-term FK506 treatment around the time of virus administration prevents the host immune response, permitting long-term gene therapy by repeated administration of the recombinant virus. (Hepatology 1997 Oct;26(4):949-56)
Article
A major drawback of adenovirus (Ad) vectors is their nonspecific transduction into various types of cells or tissue after in vivo application, which might lead to unexpected toxicity and tissue damage. To overcome this problem, we developed a fiber-mutant Ad vector displaying a monobody specific for epidermal growth factor receptor (EGFR) or vascular endothelial growth factor receptor 2 (VEGFR2) in the C-terminus of the knobless fiber protein derived from T4 phage fibritin. A monobody, which is a single domain antibody mimic based on the tenth human fibronectin type III domain scaffold with a structure similar to the variable domains of antibodies, would be suitable as a targeting molecule for display on the Ad capsid proteins because of its highly stable structure even under reducing conditions and low molecular weight (approximately 10 kDa). Surface plasmon resonance (SPR) analysis revealed that the monobody-displaying Ad vector specifically bound to the targeted molecules, leading to significant increases in cellular binding and transduction efficiencies in the targeted cells. Transduction with the monobody-displaying Ad vectors was significantly inhibited in the presence of the Fc-chimera protein of EGFR and VEGFR2. This monobody-displaying Ad vector would be a crucial resource for targeted gene therapy.
Article
A major limitation of the use of adenovirus (Ad) vectors is the innate immune response, which causes inflammatory cytokine production and tissue damages. To overcome this limitation, it is necessary to develop safer Ad vectors that are less likely to induce innate immunity. The Ad genome encodes two non-coding small RNAs, virus-associated (VA)-RNA I and VA-RNA II, which are transcribed by RNA polymerase III and promote Ad replication. Recently, we reported that VA-RNAs are produced in the cells transduced with a conventional first-generation (E1-deleted) Ad vector (FG-Ad) and trigger innate immune responses through intracellular nucleic acid sensors. In the present study, we have developed a VA-RNA-deleted Ad (AdΔVR) vector, in which the transcriptional control elements of the VA-RNA-expression were deleted. Although conventional HEK293 cells did not support the propagation of the AdΔVR vectors, HEK293 transformants inducibly expressing VA-RNA I (VR293 cells) with appropriate induction of VA-RNA I expression allowed the propagation of the AdΔVR vector. The AdΔVR vector showed high transduction efficiency comparable to that of the conventional FG-Ad vector in the cultured cells. The AdΔVR vector may be a safer alternative to the FG-Ad vector.