ArticlePDF Available

Adenovirus serotype 5 hexon is critical for virus infection of heptocytes in vivo

Authors:

Abstract and Figures

Human species C adenovirus serotype 5 (Ad5) is the most common viral vector used in clinical studies worldwide. Ad5 vectors infect liver cells in vivo with high efficiency via a poorly defined mechanism, which involves virus binding to vitamin K-dependent blood coagulation factors. Here, we report that the major Ad5 capsid protein, hexon, binds human coagulation factor X (FX) with an affinity of 229 pM. This affinity is 40-fold stronger than the reported affinity of Ad5 fiber for the cellular receptor coxsackievirus and adenovirus receptor, CAR. Cryoelectron microscopy and single-particle image reconstruction revealed that the FX attachment site is localized to the central depression at the top of the hexon trimer. Hexon-mutated virus bearing a large insertion in hexon showed markedly reduced FX binding in vitro and failed to deliver a transgene to hepatocytes in vivo. This study describes the mechanism of FX binding to Ad5 and demonstrates the critical role of hexon for virus infection of hepatocytes in vivo. • gene transfer • virus targeting
CryoEM visualization of the Ad5-FX complex and FX-binding site on hexon. 3D single-particle reconstruction of the Ad5-FX complex. Ad capsid is shown in blue. FX density is shown in red. The view in A is along an icosahedral twofold axis. The view in B is along an icosahedral fivefold axis. (Scale bar, 100 Å.) ( C ) CryoEM structure of the Ad5-FX complex (blue) together with the strongest FX density (red). The FX density was generated by subtracting a cryoEM reconstruction of the Ad5 capsid from that of the Ad5-FX complex. The strongest FX density appears in the central depression of each hexon trimer. (Scale bar, 100 Å.) ( D ) Crystal structure of Ad5 hexon (PDB ID code 1P30) shown in a ribbon representation and as a density map filtered to 30-Å resolution (transparent gray). The hexon HVR7 residues 422– 424 (TET) are shown in a space-filling representation in yellow. The position of HVR3 TDT residues found in Ad3, Ad21, and Ad50 hexons is shown by the corresponding Ad5 residues 212–214 in cyan. Both 45° tilt and top views are shown. ( E ) Amino acid sequence alignment of the hexon HVR3 and HVR7 regions for the 11 Ad serotypes tested for FX binding (Table 1). The positions of the two alternative sites proposed for FX binding are indicated by asterisks. The sequences TDT and TET that correlate with FX binding are highlighted in cyan within HVR3 and in yellow within HVR7. Nearby positively charged Arg residues that may ablate or reduce FX-binding affinity are highlighted in green. The red lines separate Ad serotypes that bind FX (above the line) from those that do not (below the line).
… 
Content may be subject to copyright.
Adenovirus serotype 5 hexon is critical for virus
infection of hepatocytes
in vivo
O. Kalyuzhniy*, N. C. Di Paolo*, M. Silvestry
, S. E. Hofherr
, M. A. Barry
§
, P. L. Stewart
, and D. M. Shayakhmetov*
*Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195-7720; Department of Molecular Physiology and
Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston,
TX 77030; and §Departments of Internal Medicine and Immunology, Mayo Clinic, Rochester, MN 55902
Edited by Thomas E. Shenk, Princeton University, Princeton, NJ, and approved February 19, 2008 (received for review December 14, 2007)
Human species C adenovirus serotype 5 (Ad5) is the most common
viral vector used in clinical studies worldwide. Ad5 vectors infect
liver cells in vivo with high efficiency via a poorly defined mech-
anism, which involves virus binding to vitamin K-dependent blood
coagulation factors. Here, we report that the major Ad5 capsid
protein, hexon, binds human coagulation factor X (FX) with an
affinity of 229 pM. This affinity is 40-fold stronger than the
reported affinity of Ad5 fiber for the cellular receptor coxsackievi-
rus and adenovirus receptor, CAR. Cryoelectron microscopy and
single-particle image reconstruction revealed that the FX attach-
ment site is localized to the central depression at the top of the
hexon trimer. Hexon-mutated virus bearing a large insertion in
hexon showed markedly reduced FX binding in vitro and failed to
deliver a transgene to hepatocytes in vivo. This study describes the
mechanism of FX binding to Ad5 and demonstrates the critical role
of hexon for virus infection of hepatocytes in vivo.
gene transfer virus targeting
Gene delivery systems based on human species C adenovirus
serotype 5 (Ad5) are among the most frequently used in
clinical studies, which aim to correct human genetic and acquired
diseases, including cancer (1). The extreme propensity of the
virus for hepatocyte infection after its intravascular delivery has
made Ad5 the vector of choice for applications requiring high-
level transgene expression in hepatocytes in vivo (2). However,
the efficient interaction between Ad5 and liver cells, which
sequester a large proportion of the delivered vector dose (3),
represents a significant hindrance if gene delivery to extrahe-
patic cells and tissues is required. From in vitro analyses, it was
found that Ad5 infection is initiated when the minor capsid
protein, fiber, binds to the coxsackievirus and adenovirus re-
ceptor (CAR) on the cell surface (4, 5). Subsequent binding of
the penton base protein to cellular integrins facilitates internal-
ization of the attached particle into the cell (6). Although binding
of both CAR and integrin is critical for cell infection in vitro,
neither of these interactions is essential for Ad5 entry into
hepatocytes in vivo (7–9).
We and others recently identified a pathway of hepatocyte
infection by Ad5, which is mediated by the vitamin K-dependent
blood coagulation factors, including factors VII, IX, X, and
protein C (10, 11). When Ad5 particles are delivered intrave-
nously, they interact with blood coagulation factors, which
subsequently bind low-density lipoprotein receptor-related pro-
tein (LRP) and heparan sulfate proteoglycans (HSPGs) on
hepatocytes to mediate virus entry. Although rather complex,
this mechanism is very efficient and can be specifically blocked
by lactoferrin, which competes for blood factor binding to LRP
and HSPGs on hepatic cells (10). It is noteworthy that, although
all of these blood factors support CAR-independent cell infec-
tion by Ad5 vectors to varying degrees, coagulation factor X
(FX) appears the most efficient at mediating Ad5 entry into
hepatic cells in vivo (11).
Based on numerous experiments that demonstrate reduced
hepatocyte infection after i.v. delivery of fiber-modified vectors
in mice, it was suggested that coagulation factors might bind to
the Ad5 fiber to mediate hepatocyte infection. However, there
is still no definitive evidence that a fiber– coagulation factor
interaction is ultimately required to mediate CAR-independent
Ad5 entry into hepatocytes in vivo.
In this study, we analyzed the interactions of human FX with
human adenoviruses from species B–F. Using surface plasmon
resonance (SPR), we found that FX binds with picomolar
affinity to hexon, the major Ad capsid protein. Cryoelectron
microscopy (cryoEM) and single-particle image reconstruction
have localized the FX-binding area to the central depression at
the top of each Ad5 hexon trimer. This cryoEM result, combined
with sequence analysis of hexons that bind FX and those that do
not, indicates there are likely two alternative sites for FX binding.
One site is within hypervariable region 3 (HVR3), and the other
is within hypervariable region 7 (HVR7). Both sites are pre-
dicted to form similar binding pockets within the central de-
pression of the hexon trimer. An adenovirus mutant that binds
FX in vitro with 10,000-fold reduced affinity compared with
unmodified vector failed to deliver the red fluorescent protein
(RFP) transgene in vivo. Thus, our study describes the mecha-
nism of FX binding to Ad5 and demonstrates the critical role of
hexon for virus infection of hepatocytes in vivo.
Results
Ad5 Hexon Binds FX with Picomolar Affinity. To quantitatively
characterize binding between blood coagulation factors and the
adenovirus capsid, we used SPR analysis (BIAcore). To avoid
complications with interpretation of the results caused by the
multivalent nature of the virus particle, Ad5 particles were
immobilized on the sensor chip by cross-linking, and varying
concentrations of blood factors were injected over sensor sur-
faces and binding responses were recorded and analyzed. Fig. 1
(Ad5 column) shows the representative datasets obtained from
the analysis of blood–factor interactions together with global fits
to a 1:1 interaction model (orange lines). Among all blood
factors tested [Fig. 1 and supporting information (SI) Fig. S1],
human FX (hFX) and mouse FX (mFX) demonstrated the
highest-affinity binding to Ad5. The association constant of the
Ad5-hFX-binding reaction was determined to be 3.37 10
5
M
1s
1
, the dissociation constant 7.71 10
5
s
1
and K
D
equal
to 228.7 pM. We detected no binding of FXI, FXII, and
FX-desGla to Ad5 under the conditions used (Fig. S1). Ad5-hFX
Author contributions: O.K., N.C.D.P., and M.S. contributed equally to this work; O.K.,
N.C.D.P., P.L.S., and D.M.S. designed research; O.K., N.C.D.P., M.S., S.E.H., P.L.S., and D.M.S.
performed research; O.K., S.E.H., M.A.B., and P.L.S. contributed new reagents/analytic
tools; O.K., N.C.D.P., M.S., S.E.H., M.A.B., P.L.S., and D.M.S. analyzed data; and O.K., P.L.S.,
and D.M.S. wrote the paper.
The authors declare no conflict of interest.
This article is a PNAS Direct Submission.
To whom correspondence should be addressed. E-mail: dshax@u.washington.edu.
This article contains supporting information online at www.pnas.org/cgi/content/full/
0711757105/DCSupplemental.
© 2008 by The National Academy of Sciences of the USA
www.pnas.orgcgidoi10.1073pnas.0711757105 PNAS
April 8, 2008
vol. 105
no. 14
5483–5488
MEDICAL SCIENCES
binding strongly depends on the presence of both Ca
2
and Mg
2
ions and could be completely eliminated in a buffer containing
3 mM EDTA (data not shown).
Because of the very high affinity of FX for Ad5 and its strong
dependence on the presence of calcium and magnesium ions, we
hypothesized that the FX-binding protein within the Ad5 capsid
could be purified from virus-infected cells using affinity chro-
matography. Indeed, using the hFX affinity column, a 98% pure
protein with an apparent molecular mass of 100 kDa (after
separation by SDS/PAGE under denaturating conditions) was
obtained from the lysate of cells infected with Ad5 (Fig. S2 and
SI Text,Materials and Methods). The affinity-purified protein
was digested with trypsin and analyzed by using data-dependent
tandem mass spectrometry. Numerous peptides corresponding
to the Ad5 hexon were readily identified by Mascot software.
The trimeric state of the purified hexon was further confirmed
by size-exclusion chromatography. The experimentally deter-
mined molecular mass of the protein was estimated to be 337
kDa, which corresponds well with the expected molecular mass
of 324 kDa for the Ad5 hexon trimer (Fig. S2 A–C).
Using the SPR technique, we next analyzed the kinetic
parameters of the blood factor interaction with purified Ad5
hexon and purified Ad5 fiber knob domain, expressed in Esch-
erichia coli (Fig. S2 D–F and SI Text,Materials and Methods). The
data obtained (Fig. 1, Ad5-Hex and Ad5-FKn columns, and
Table S1) showed that, for all analyzed blood factors, interaction
with purified Ad5 hexon trimer, but not the Ad5 fiber knob
domain, closely follows the kinetics for the FX interaction with
the whole virus (Fig. 1). In addition, we analyzed blood factor
binding to another group C Ad serotype, Ad2, and commercially
available purified Ad2 hexon protein. This analysis demon-
strated that both human and mouse FX bind Ad2 particles and
purified Ad2 hexon with identical kinetics and af finity (Fig. 1 and
Table S1). Collectively, these data indicate that the Ad hexon
possesses a high-affinity binding site for vitamin K-dependent
blood factors. To further assess this possibility, we analyzed
blood factor interaction with an Ad5-based vector, Ad5BAP,
that possesses an insertion of a 71-aa biotin acceptor peptide
(BAP) in the exposed hexon loop of the HVR5 (12). Our analysis
showed that FX binding to Ad5BAP was reduced 10,000-fold
compared with unmodified Ad5 vector, and the 71-aa insertion
in HVR5 region of the hexon completely eliminated binding of
FIX and Protein C (Fig. 1 and Table S1).
FX Interaction with Different Human Ad Serotypes and Reovirus. We
next analyzed the affinity of FX binding to various human Ad
serotypes from species B–F and human reovirus T3D (13) by
using the same SPR protocol. This analysis revealed that human
Ad serotypes greatly vary in their ability to bind FX (Table 1).
The highest-affinity FX binding was observed for species C Ad5,
followed by species B Ad16, and then species C Ad2. Except for
the observation that two species C serotypes (Ad2 and Ad5)
exhibited high affinity for FX, no other correlation was noted
between the Ad group and FX affinity among the 11 serotypes
tested. Micromolar affinity binding was observed for species B
(Ad3 and Ad21), E (Ad4), and F (Ad41) serotypes. No FX
binding was observed for species B Ad35 or Ad50, species D Ad9
or Ad51 serotypes, or reovirus T3D. It is noteworthy that, in
agreement with these data, vectors based on Ad5 and Ad2
serotypes were shown to infect hepatocytes with high efficiency
(14, 15); however, Ad35-based vectors transduce hepatocytes
poorly after i.v. delivery (16, 17).
CryoEM Analysis of the Ad5-FX Complex. CryoEM and single-
particle image reconstruction were performed to visualize the
interaction of FX with Ad5. Comparison of cryoEM images of
the Ad5-FX complex with those of Ad5 alone revealed that FX
coats the virus with a layer of extra density. The FX coating
effect was also clearly observed in a 3D reconstruction of the
Ad5-FX complex (FX density is shown in red in Fig. 2 Aand B).
The reconstructed Ad5 virion appears to be covered with a
70-Å-thick mesh of FX density over the entire capsid, except
within the vicinity of the pentons at the icosahedral fivefold
symmetry axes.
The Ad5-FX reconstruction compared with a previous recon-
struction of the Ad5 capsid (18) revealed that the point of
contact between FX and the Ad5 hexon is within the central
depression of each hexon trimer (Fig. 2C). Fitting of the Ad5
hexon crystal structure [PDB ID code 1P30 (19)] into the
cryoEM hexon density showed that the exposed hypervariable
Fig. 1. Kinetic response data for different blood coagulation factor binding
to Ad5, Ad2, and adenovirus capsid proteins. Experimentally obtained data
are shown by black lines. Global fits of these data to 1:1 single-site interaction
model are shown in orange. The responses are shown in instrument response
units (RU) vs. time in seconds (s). Columns represent six different ligands that
were immobilized on the CM5 sensor chip. Ad5, adenovirus type 5; Ad5,
Hex-Ad5 hexon; Ad5-FKn, Ad5 fiber knob domain; Ad2, adenovirus type 2;
Ad2-Hex, Ad2 hexon; Ad5BAP, hexon-mutated Ad5-based vector. Rows rep-
resent eight different blood factors used as analytes in Biacore experiments.
hFX, human factor X; mFX, mouse factor X; FIX, human factor IX; FVII, human
factor VII; protein C, human protein C.
Table 1. Affinity of FX binding to different viruses
Virus Immobilized RU K
d
,nM
Adenovirus
Ad5 384 0.229
Ad16 470 1.67
Ad2 352 20.9
Ad21 615 410
Ad41 347 630
Ad4 2,900 2,480
Ad3 2,973 3,000
Ad35 315 No binding
Ad51 667 No binding
Ad9 311 No binding
Ad50 256 No binding
Reovirus
T3D 486 No binding
Ad5-sCAR 7.9
5484
www.pnas.orgcgidoi10.1073pnas.0711757105 Kalyuzhniy et al.
regions HVR3, HVR5, and HVR7 are in close proximity to FX
density (Fig. 2Dand Fig. S3). Amino acid sequence alignment of
the hexons from the 11 Ad serotypes tested (Table 1) indicated
that FX binding could be associated with the presence of amino
acids TET or TDT within HVR7 (Fig. 2 E). In addition, there are
two serotypes (Ad3 and Ad21) that have the sequence TDT
within HVR3. Both the HVR7 TET site in the Ad5 hexon and
the HVR3 TDT site mapped onto the Ad5 hexon point into the
central depression of hexon with a similar geometric arrange-
ment (Fig. 2D). Although HVR5 is spatially adjacent to HVR7
and is also localized in the central depression of the hexon trimer
(Fig. S3), its amino acid composition is highly diverse, and there
is no apparent correlation between the presence of particular
amino acids within HVR5 and the serotype affinity for FX.
FX-Binding-Ablated Virus Failed to Transduce Hepatocytes
in Vivo
.
Recent studies demonstrated that vitamin K-dependent blood
coagulation factors mediate efficient hepatocyte transduction by
Ad5-based vectors in vivo (10, 11). To analyze the role of the
FX–hexon interaction in Ad hepatocyte transduction, we took
advantage of a hexon-mutated vector, Ad5BAP (12). Analysis of
blood factor binding to Ad5BAP by using SPR demonstrated a
4 order-of-magnitude loss in affinity of FX binding to Ad5BAP
virions (Fig. 1 and Table S1). This suggested that this large
insertion in HVR5 near the putative FX attachment site on
hexon might sterically block FX binding or induce a conforma-
tional change within the FX attachment site that could inhibit
binding. We first analyzed Ad5BAP infectivity in vitro. The virus
particle-to-plaque forming unit ratio, determined on 293 cells,
was 75 23 and 79 10 for control unmodified Ad5RFP vector
and Ad5BAP, respectively. To further investigate whether a
large insertion in the HVR5 area of hexon may adversely affect
early steps of virus infection, we analyzed virus particle attach-
ment by using quantitative Southern blot analysis (20) (Fig.
S4A). Our analysis showed that both unmodified Ad5RFP vector
and Ad5BAP attached to cells with similar efficacy. Next, we
analyzed internalization rates for these vectors using a neutral-
izing antibody escape assay (21). The data obtained showed that
internalization rates were identical for both Ad5RFP and
Ad5BAP vectors (Fig. S4B). Confocal microscopy studies of
intracellular virus trafficking revealed that both vectors rapidly
migrated to the nuclear periphery. However, unlike the Ad5RFP
vector, infection of cells with Ad5BAP induces significant rear-
rangement of Cathepsin D-positive late endosomal/lysosomal
compartment and Ad5BAP virus particles colocalized with late
endosomes/lysosomes (Fig. S4C).
Analysis of virus infectivity on human lung epithelial (A549)
or hepatoma (HepG2) cells revealed that, although no differ-
ence in infectivity between Ad5RFP and Ad5BAP was found in
A549 cells, there was a marked reduction in infectivity of
Ad5BAP, compared with Ad5RFP, in HepG2 cells (Fig. S4 D
and E). When we analyzed virus infectivity on these cell lines in
the presence of blood coagulation factors, we found that the
infectivity of Ad5BAP was significantly lower compared with
Ad5RFP when blood factors were added to the infection media
(Fig. 3 Aand B). This difference was most dramatic in hepatoma
HepG2 cells, compared with A549 cells. Collectively, all these in
Fig. 2. CryoEM visualization of the Ad5-FX complex and FX-binding site on hexon. 3D single-particle reconstruction of the Ad5-FX complex. Ad capsid is shown
in blue. FX density is shown in red. The view in Ais along an icosahedral twofold axis. The view in Bis along an icosahedral fivefold axis. (Scale bar, 100 Å.) (C)
CryoEM structure of the Ad5-FX complex (blue) together with the strongest FX density (red). The FX density was generated by subtracting a cryoEM reconstruction
of the Ad5 capsid from that of the Ad5-FX complex. The strongest FX density appears in the central depression of each hexon trimer. (Scale bar, 100 Å.) (D) Crystal
structure of Ad5 hexon (PDB ID code 1P30) shown in a ribbon representation and as a density map filtered to 30-Å resolution (transparent gray). The hexon HVR7
residues 422– 424 (TET) are shown in a space-filling representation in yellow. The position of HVR3 TDT residues found in Ad3, Ad21, and Ad50 hexons is shown
by the corresponding Ad5 residues 212–214 in cyan. Both 45° tilt and top views are shown. (E) Amino acid sequence alignment of the hexon HVR3 and HVR7
regions for the 11 Ad serotypes tested for FX binding (Table 1). The positions of the two alternative sites proposed for FX binding are indicated by asterisks. The
sequences TDT and TET that correlate with FX binding are highlighted in cyan within HVR3 and in yellow within HVR7. Nearby positively charged Arg residues
that may ablate or reduce FX-binding affinity are highlighted in green. The red lines separate Ad serotypes that bind FX (above the line) from those that do not
(below the line).
Fig. 3. Transduction of A549 and HepG2 cells by Ad5RFP and Ad5BAP vectors
in the presence of human blood factors. Cells were infected with a multiplicity
of infection of 25 pfu per cell for each indicated virus in the presence of 8
g/ml
of FIX, FX, or protein C or 0.5
g/ml of FVII (physiological concentration of
these factors in human plasma). Two hours after addition of virus to cells, cells
were washed with new media, and RFP gene expression was analyzed 24 h
later by flow cytometry. In control settings (Saline), viruses were added to cells
in growth media only (n6).
Kalyuzhniy et al. PNAS
April 8, 2008
vol. 105
no. 14
5485
MEDICAL SCIENCES
vitro studies showed that virus attachment to cells and internal-
ization into cells were identical for Ad5RFP and hexon-mutated
Ad5BAP vectors. However, cell-type-specific factors may affect
intracellular trafficking and infectivity of hexon-mutated vector.
Importantly, addition of blood factors to infection media signif-
icantly increased infectivity of Ad5RFP vector and had no effect
on infectivity of Ad5BAP in vitro.
When equal doses of unmodified Ad5RFP and Ad5BAP
vectors were injected i.v. into mice, the amounts of Ad5BAP
vector trapped in the liver1haftervirusadministration were
50% lower than for Ad5RFP (Fig. 4Aand Fig. S5A). Pretreat-
ment of mice with warfarin, which blocks
-carboxylation of all
vitamin K-dependent zymogens and has been shown to reduce
liver cell transduction by Ad5-based vectors (11), did not signif-
icantly reduce Ad vector trapping in the liver tissue at this time.
Analysis of Ad5BAP DNA in the liver 48 h after virus injection
revealed that these levels were 75% lower, compared with DNA
levels of Ad5RFP. Treatment of mice with warfarin reduced
levels of Ad5RFP DNA in the liver by 85%. Preinjection of
warfarin-treated mice with FX before Ad5RFP injection re-
sulted in a remarkable 24-fold increase in vector DNA levels in
the liver at this time point (Fig. 4A). Administration of FX
before Ad5BAP injection in warfarin-treated mice did not
increase low levels of Ad5BAP DNA in the liver, confirming our
findings on reduced affinity of blood factor binding to Ad5BAP
(Fig. 1) and cell transduction in the presence of blood factors in
vitro (Fig. 3).
Evaluation of hepatocyte transduction by fluorescent micros-
copy 48 h after i.v. injection of Ad5RFP or Ad5BAP revealed
(Fig. 4B) that RFP levels in hepatocytes mimicked vector
genomic DNA levels, determined by Southern blot analysis.
The Ad5BAP vector failed to express RFP after intravascular
delivery. Warfarin treatment of mice ablated RFP transgene
expression in hepatocytes after injection of Ad5RFP vector, and
this ablation could be completely reverted by restoration of
physiological levels of FX in blood (Fig. 4B). Importantly, the
Ad5BAP vector failed to transduce hepatocytes in warfarin-
treated mice even in the presence of FX. Western blot analysis
for RFP protein levels in hepatic tissue demonstrated high levels
of RFP expression in livers of mice injected with Ad5RFP (Fig.
S5 Band C). The levels of hepatic RFP expression were at the
limit of detection by Western blot analysis in mice injected with
Ad5BAP. The median of RFP band intensity was 68-fold
stronger for Ad5RFP, compared with Ad5BAP.
Discussion
Virus binding to cellular receptors is a critical early step in
initiation of infection. This interaction is under strong evolu-
tionary pressure and is often mediated by specialized virus
capsid proteins. For species C Ad serotype 5, CAR was shown
to be the high-affinity attachment receptor at the cell surface (4,
5). The presence of CAR on epithelial cells determines the
susceptibility of these cells to Ad5 infection both in vitro and in
vivo (22, 23). Although the interactions of Ad5 with cells in vitro
are known in great detail (24, 25), the pathways of cell-specific
infection by Ad in vivo remain poorly understood. Specifically,
after i.v. delivery, the vast majority of Ad particles are trapped
in the liver (26–28). Unsuccessful attempts to prevent hepato-
cyte transduction by Ad5-based vectors through ablation of CAR
and/or integrin binding implied that another mechanism must
exist to allow Ad5 virus particles to enter hepatic cells in a
CAR-independent manner (29, 30).
Recently, we and others identified a blood factor-dependent
pathway that allows Ad5 to infect hepatocytes independently of
virus binding to CAR (10, 11). These studies also demonstrated
that FX is the most efficient at supporting Ad5 entry into
hepatocytes. Despite this advance in our understanding of
pathways of hepatocyte infection with Ad5 in vivo, the precise
mechanism of blood coagulation factor interaction with Ad
particles remained unclear.
In this study, we analyzed, in a systematic way, Ad capsid
protein interaction with vitamin K-dependent coagulation fac-
tors and found that the Ad5 hexon possesses a high-affinity
binding site for human FX. The determined picomolar affinity
of FX binding to Ad5 implies that, when delivered via an
intravascular route, Ad5 will form a ver y stable complex with FX
and not other vitamin K-dependent zymogens, which bind Ad5
with lower affinity (Table S1) and are less abundant in plasma.
CryoEM visualization of the Ad5-FX complex revealed a mesh
of FX molecules covering almost the entire adenovirus capsid.
Importantly, analysis of the contact point between FX and hexon
showed that FX binds within the central depression at the top of
each hexon trimer.
The hexon-mutated Ad5BAP vector failed to transduce he-
patocyte when injected into wild-type mice or warfarin-treated
mice with physiological levels of FX in circulation. Our in vitro
data also showed that Ad5BAP infectivity cannot be increased
by addition of blood factors into infection media (Fig. 3);
however, unmodified Ad5RFP vector infectivity increases dra-
matically when FVII, FIX, or FX is added to the media during
virus infection.
Hexon-mutated Ad5BAP virus could be efficiently propa-
gated in 293 cells, and our in vitro analyses showed it is attached
to and internalized into cells with high efficiency, which is equal
to the unmodified Ad5RFP vector (Fig. S4). However, unlike
Ad5RFP, Ad5BAP demonstrated colocalization with late endo-
Fig. 4. In vivo analysis of hexon-mutated adenovirus vector. (A) Ad DNA deposition and persistence in the liver after i.v. injection of Ad5RFP or hexon-mutated
Ad5BAP vectors. Amounts of vector genomic DNA in the liver were analyzed by Southern blot analysis and quantified after processing of band signal intensity
by phosphorimager. The intensity of Ad5RFP bands at 1 and 48 h after virus injection was designated as 100. The graphs show the averaged data obtained from
two independent experiments with four to six biological replicates per each group. *,P0.01. The representative image of Southern blot hybridization is shown
in Fig. S6A.(B) Histological analysis of Ad-encoded RFP gene expression in mouse hepatocytes 48 h after i.v. injection of indicated vectors. Representative fields
are shown. RFP expression is observed as red fluorescence on fixed liver sections. Corresponding fields in DAPI channel (nucleus-specific staining) are shown.
5486
www.pnas.orgcgidoi10.1073pnas.0711757105 Kalyuzhniy et al.
somal/lysosomal cathepsin D-positive compartments and vari-
able infectivity on human lung epithelial and hepatoma cells.
These data indicate that such postinternalization steps of infec-
tion as release from the endosomes or viral DNA transport into
the nucleus may be more significantly affected by cell-type-
specific factors for hexon-modified Ads when compared with
unmodified vectors.
Our earlier data suggested that FIX could efficiently support
CAR-independent liver cell infection both in vitro and in vivo,
and that the fiber knob domain is the likely moiety that binds
FIX in the Ad capsid (10). Moreover, numerous studies of
hepatocyte transduction by capsid-modified Ad vectors
showed markedly reduced transgene expression, when vectors
with short-shafted fibers were applied in vivo (29, 31, 32). Our
SPR studies demonstrated that FIX binding to Ad5 follows a
complex kinetic that cannot be described by a 1:1 single-site
interacting model (Fig. 1). Moreover, the affinity of FIX
binding to purified hexon is reduced, compared with its
binding to complete Ad5 particles (Table S1). The observed
kinetics of FIX binding to Ad5 particles fit well into a two-site
interacting model (data not shown; O.K. and D.M.S., unpub-
lished work). Although the precise localization of these sites
remained to be determined, the hexon-mutated Ad5BAP
vector failed to bind FIX in SPR assay and transduce cells in
a FIX-dependent manner in vitro (Fig. 3). These data suggest
that, similar to FX, for FIX, Ad5 hexon possesses the dominant
binding site on the Ad capsid.
Our finding is critical for understanding the interactions of Ad
with host cells in vivo and has important practical implications
for development of safe and efficient cell- and tissue-type-
specific adenovirus vectors for gene therapy applications.
Materials and Methods
Cells and Viruses. The first-generation Ad5-based vectors, expressing the
dsRED2 RFP, Ad5RFP, and Ad5BAP, were constructed and described in detail
(12, 33). Ad5BAP is described as Ad-Hexon-BAP (33). Wild-type human Ad
serotypes Ad2, Ad3, Ad4, Ad5, Ad9, Ad16, Ad21, Ad35, Ad41, Ad50, and Ad51
were obtained from American Type Culture Collection. Human reovirus T3D
was kindly provided by Terence Dermody (Vanderbilt University, Nashville,
TN). Viruses were banded in CsCl gradients, dialyzed, and stored in aliquots, as
described (34). Adenovirus genome titers were determined by quantitative
real-time PCR (for Ad5RFP and Ad5BAP) and OD260 measurement (for wild-
type viruses).
Proteins. All coagulation blood factors were purchased from Haematologic
Technologies. All supplied blood factors were at 95% purity: human FX,
HCX-0050, lot T1206; mouse FX, MCX-5050, lot R0903; human FIX, HCIX-0040,
lot U1206; human FVII, HCVII-0030, lot U1206–0.02; human protein C, HCPC-
0070, lot W0822; human FXI, HCXI-0150, lot U0607; human FXII, HCXII-0155,
lot U1020; and human Gla-domainless FX, HCX-GD, lot U0629. Ad2 hexon
(catalog no. R14800) was purchased from Meridian Life Sciences. Ad5 hexon
and Ad5 fiber knob domain purification is described in detail in SI Text,
Materials and Methods. BSA, catalog no. A0281-5G, was purchased from
Sigma.
SPR Analyses. All analyses were carried out on a Biacore 2000 machine.
Research grade CM5 sensor chips, N-hydroxysuccinimide, N-ethyl-N-(3-
diethylaminopropyl)carbodiimide, ethanolaminehydrochloride, HBSP run-
ning buffer, and HBSEP regeneration buffer were purchased from the man-
ufacturer (Biacore). All data were collected at 1 Hz by using two or three
replicate injections for each concentration of analyte. Data processing and
kinetic analysis were performed by using Scrubber software (version 2.0,
BioLogic Software). Data processing included double referencing (35). Pro-
cessed data were globally fit to a simple 1:1 interaction model.
Cryoelectron Microscopy. An Ad5-FX complex was prepared by mixing 100
l
of the Ad5S vector (34), which has a short-shafted fiber (1 1013 virus particles
per ml) with 1
l of FX (2 mg/ml). Six-microliter aliquots of the Ad5-FX complex
were applied to Quantifoil R2/4 holey carbon grids (Quantifoil Micro Tools)
and a Vitrobot cryofixation device (FEI) was used for blotting and sample
vitrification in liquid ethane. Data collection was performed on an FEI Tecnai
12 (120 kV, LaB6) transmission cryoelectron microscope equipped with a Gatan
cryoholder and Gatan UltraScan 2kx2k charge-coupled device camera (Gatan).
Twenty-six cryoelectron micrographs were collected at a nominal magnifica-
tion of 67,000, from which 40 particle images were selected by using the
program VIRUS (36). Initial estimates for the defocus and astigmatism param-
eters were determined with CTFFIND3 (37). The FREALIGN package (38) was
used for refinement of the orientational, defocus, and astigmatism parame-
ters of each particle image and to calculate 3D reconstructions. The resolution
of the icosahedral capsid is estimated to be 38 Å by the Fourier Shell Corre-
lation 0.5 threshold with applied inner and outer spherical masks (radii 300
and 463 Å). The resolution of the reconstruction including FX and fiber density
(outer radius 613 Å) is 40 Å.
Adenovirus Infection
in Vivo
.All experimental procedures involving animals
were conducted in accordance with the institutional guidelines set forth by
the University of Washington. C57BL/6 mice (Charles River) were housed in
specific pathogen-free facilities. For analysis of Ad-mediated gene transfer
into liver cells, 1011 Ad particles in 200
l of PBS were injected by tail-vein
infusion. For in vivo transduction studies, mice were killed 1 or 48 h after virus
infusion, and livers were processed for DNA, protein, and histological analy-
ses. For analysis of Ad genome accumulation in the liver tissue 1 h after Ad
vector administration, the blood was flushed from the liver by a cardiac saline
perfusion, livers were harvested, and total DNAs were purified as described
(39). Where applicable, mice were injected with warfarin and FX, as described
in ref. 11.
Histological Analysis. Liver samples from mice injected with Ad vectors were
fixed, dehydrated in sucrose, and frozen in OCT compound. RFP gene expres-
sion in hepatic cells was analyzed on frozen sections using conventional UV
fluorescent microscopy.
ACKNOWLEDGMENTS. We thank Daniel Stone for critical reading of this
manuscript and Terence Dermody for providing reovirus T3D. This work was
supported by funding from the National Institutes of Health (Grants AI062853,
AI064882, and AI065429, to D.M.S.; Grant AI042929, to P.L.S.) and by grants to
M.A.B. from the Muscular Dystrophy Association and the Propionic Acidemia
Foundation.
1. (2008) J Gene Med Gene Therapy Clinical Trials Worldwide, www.wiley.co.uk/genmed/
clinical. Accessed March 20, 2008.
2. Thomas CE, Ehrhardt A, Kay MA (2003) Progress and problems with the use of viral
vectors for gene therapy. Nat Rev Genet 4:346–358.
3. Wickham TJ (2003) Ligand-directed targeting of genes to the site of disease. Nat Med
9:135–139.
4. Bergelson JM, Cunningham JA, Droguett G, Kurt-Jones EA, Krithivas A, et al. (1997)
Isolation of a common receptor for Coxsackie B viruses and adenoviruses 2 and 5.
Science 275:1320–1323.
5. Tomko RP, Xu R, Philipson L (1997) HCAR and MCAR: the human and mouse cellular
receptors for subgroup C adenoviruses and group B coxsackieviruses. Proc Natl Acad Sci
USA 94:3352–3356.
6. Wickham TJ, Mathias P, Cheresh DA, Nemerow GR (1993) Integrins alpha v beta 3 and
alpha v beta 5 promote adenovirus internalization but not virus attachment. Cell
73:309–319.
7. Alemany R, Curiel DT (2001) CAR-binding ablation does not change biodistribution and
toxicity of adenoviral vectors. Gene Ther 8:1347–1353.
8. Smith T, Idamakanti N, Kylefjord H, Rollence M, King L, et al. (2002) In vivo hepatic
adenoviral gene delivery occurs independently of the coxsackievirus-adenovirus re-
ceptor. Mol Ther 5:770–779.
9. Smith TA, Idamakanti N, Marshall-Neff J, Rollence ML, Wright P, et al. (2003) Receptor
interactions involved in adenoviral-mediated gene delivery after systemic administra-
tion in non-human primates. Hum Gene Ther 14:1595–1604.
10. Shayakhmetov DM, Gaggar A, Ni SH, Li ZY, Lieber A (2005) Adenovirus binding to blood
factors results in liver cell infection and hepatotoxicity. J Virol 79:7478–7491.
11. Parker AL, Waddington SN, Nicol CG, Shayakhmetov DM, Buckley SM, et al. (2006)
Multiple vitamin K-dependent coagulation zymogens promote adenovirus-mediated
gene delivery to hepatocytes. Blood 108:2554–2561.
12. Campos SK, Parrott MB, Barry MA (2004) Avidin-based targeting and purification of a
protein IX-modified, metabolically biotinylated adenoviral vector. Mol Ther 9:942–
954.
13. Stehle T, Dermody TS (2003) Structural evidence for common functions and ancestry of
the reovirus and adenovirus attachment proteins. Rev Med Virol 13:123–132.
14. Morral N, O’Neal W, Rice K, Leland M, Kaplan J, et al. (1999) Administration of
helper-dependent adenoviral vectors and sequential delivery of different vector se-
rotype for long-term liver-directed gene transfer in baboons. Proc Natl Acad Sci USA
96:12816–12821.
15. Parks RJ, Evelegh CM, Graham FL (1999) Use of helper-dependent adenoviral vectors of
alternative serotypes permits repeat vector administration. Gene Ther 6:1565–1573.
Kalyuzhniy et al. PNAS
April 8, 2008
vol. 105
no. 14
5487
MEDICAL SCIENCES
16. Seshidhar Reddy P, Ganesh S, Limbach MP, Brann T, Pinkstaff A, et al. (2003) Devel-
opment of adenovirus serotype 35 as a gene transfer vector. Virology 311:384–393.
17. Sakurai F, Mizuguchi H, Yamaguchi T, Hayakawa T (2003) Characterization of in vitro
and in vivo gene transfer properties of adenovirus serotype 35 vector. Mol Ther
8:813–821.
18. Saban SD, Silvestry M, Nemerow GR, Stewart PL (2006) Visualization of alpha-helices in
a 6-angstrom resolution cryoelectron microscopy structure of adenovirus allows re-
finement of capsid protein assignments. J Virol 80:12049–12059.
19. Rux JJ, Kuser PR, Burnett RM (2003) Structural and phylogenetic analysis of adenovirus
hexons by use of high-resolution X-ray crystallographic, molecular modeling, and
sequence-based methods. J Virol 77:9553–9566.
20. Di Paolo NC, Kalyuzhniy O, Shayakhmetov DM (2007) Fiber shaft-chimeric adenovirus
vectors lacking the KKTK-motif efficiently infect liver cells in vivo.J Virol 81:12249–
12259.
21. Shayakhmetov DM, Eberly AL, Li ZY, Lieber A (2005) Deletion of penton RGD motifs
affects the efficiency of both the internalization and the endosorne escape of viral
particles containing adenovirus serotype 5 or 35 fiber knobs. J Virol 79:1053–1061.
22. Walters RW, Freimuth P, Moninger TO, Ganske I, Zabner J, et al. (2002) Adenovirus fiber
disrupts CAR-mediated intercellular adhesion allowing virus escape. Cell 110:789–799.
23. Walters RW, van’t Hof W, Yi SMP, Schroth MK, Zabner J, et al. (2001) Apical localization
of the coxsackie-adenovirus receptor by glycosyl-phosphatidylinositol modification is
sufficient for adenovirus-mediated gene transfer through the apical surface of human
airway epithelia. J Virol 75:7703–7711.
24. Goosney DL, Nemerow GR (2003) Adenovirus infection: Taking the back roads to viral
entry. Curr Biol 13:R99–R100.
25. Meier O, Greber UF (2003) Adenovirus endocytosis. J Gene Med 5:451– 462.
26. Brunetti-Pierri N, Palmer DJ, Beaud, Carey KD, Finegold M, et al. (2004) Acute toxicity
after high-dose systemic injection of helper-dependent adenoviral vectors into non-
human primates. Hum Gene Ther 15:35–46.
27. Hodges BL, Taylor KM, Chu QM, Scull SE, Serriello RG, et al. (2005) Local delivery of a
viral vector mitigates neutralization by antiviral antibodies and results in efficient
transduction of rabbit liver. Mol Ther 12:1043–1051.
28. Lievens J, Snoeys J, Vekemans K, Van Linthout S, de Zanger R, et al. (2004) The size of
sinusoidal fenestrae is a critical determinant of hepatocyte transduction after adeno-
viral gene transfer. Gene Ther 11:1523–1531.
29. Nicol CG, Graham D, Miller WH, White SJ, Smith TAG, et al. (2004) Effect of adenovirus
serotype 5 fiber and penton modifications on in vivo tropism in rats. Mol Ther
10:344–354.
30. Akiyama M, Roelvink P, Einfeld D, Kovesdi I, King CR (2003) Effect of Ablating CAR and
integrin binding on the biodistribution and cellular localization of adenovirus vectors
following intravenous or intraperitoneal delivery. Mol Ther 7:S173.
31. Ni SH, Bernt K, Gaggar A, Li ZY, Kiem HP, et al. (2005) Evaluation of biodistribution and
safety of adenovirus vectors containing group B fibers after intravenous injection into
baboons. Hum Gene Ther 16:664– 677.
32. Schoggins JW, Gall JGD, Falck-Pedersen E (2003) Subgroup B and F fiber chimeras
eliminate normal adenovirus type 5 vector transduction in vitro and in vivo.J Virol
77:1039–1048.
33. Campos SK, Barry MA (2006) Comparison of adenovirus fiber, protein IX, and hexon
capsomeres as scaffolds for vector purification and cell targeting. Virology 349:453–
462.
34. Shayakhmetov DM, Lieber A (2000) Dependence of adenovirus infectivity on length of
the fiber shaft domain. J Virol 74:10274–10286.
35. Myszka DG (1999) Improving biosensor analysis. J Mol Recognit 12:279 –284.
36. Adiga U, Baxter WT, Hall RJ, Rockel B, Rath BK, et al. (2005) Particle picking by
segmentation: A comparative study with SPIDER-based manual particle picking. J
Struct Biol 152:211–220.
37. Mindell JA, Grigorieff N (2003) Accurate determination of local defocus and specimen
tilt in electron microscopy. J Struct Biol 142:334–347.
38. Grigorieff N (2007) FREALIGN: High-resolution refinement of single particle structures.
J Struct Biol 157:117–125.
39. Shayakhmetov DM, Li ZY, Ni S, Lieber A (2004) Analysis of adenovirus sequestration in
the liver, transduction of hepatic cells, and innate toxicity after injection of fiber-
modified vectors. J Virol 78:5368–5381.
5488
www.pnas.orgcgidoi10.1073pnas.0711757105 Kalyuzhniy et al.
... It is widely used as a gene delivery vector of transgenes in gene therapy and basic science research. It was shown [49] that hexon protein is critical for virus infection of hepatocytes in vivo and human coagulation factor (FX) binding to Ad5. The evolution of species HAdV-C was studied by generating 51 complete genome sequences from circulating strains. ...
... As early gene region sequences are not considered in the type definition of HAdVs, the evolution of HAdV-C remains on the subtype level. Nonetheless, recombination of the E1 and E4 gene regions may influence the virulence of HAdV-C strains [49]. ...
Article
Full-text available
Simple Summary Current cancer therapies are insufficient to cure advanced malignancies and cause off-target toxicity. Scientists have focused on developing new, more efficacious, and selective therapies to overcome these issues. This review describes a modern approach based on oncotherapy, a type of cancer treatment that helps the immune system fight cancer. Thus, genetic modifications of adenoviruses to increase the anticancer properties of the therapy (benefits, critical points, and risk assessment) were reviewed. We analyzed clinical trial databases and highlighted significant achievements in adenovirus oncotherapy in recent years. Abstract In clinical trials, adenovirus vectors (AdVs) are commonly used platforms for human gene delivery therapy. High genome capacity and flexibility in gene organization make HAdVs suitable for cloning. Recent advancements in molecular techniques have influenced the development of genetically engineered adenovirus vectors showing therapeutic potential. Increased molecular understanding of the benefits and limitations of HAdVs in preclinical research and clinical studies is a crucial point in the engineering of refined oncolytic vectors. This review presents HAdV species (A–G) used in oncotherapy. We describe the adenovirus genome organizations and modifications, the possibilities oncolytic viruses offer, and their current limitations. Ongoing and ended clinical trials based on oncolytic adenoviruses are presented. This review provides a broad overview of the current knowledge of oncolytic therapy. HAdV-based strategies targeting tumors by employing variable immune modifiers or delivering immune stimulatory factors are of great promise in the field of immune oncologyy This approach can change the face of the fight against cancer, supplying the medical tools to defeat tumors more selectively and safely.
... Also, biosensors containing covalently immobilized ligands usually need harsh regeneration procedures after each analysis, that are scarcely suitable for fragile virion structure. Finally, to avoid heterogeneous surfaces that could make difficult the interpretation of the binding data [12], amine coupling is usually performed with purified virion preparations [22][23][24]. We then decided to adopt the ligand-capture approach, anchoring the intact RHDV2 virions to MAb-4H12 covalently immobilized to the sensorchip. ...
Preprint
Full-text available
Biosensing technologies and monoclonal antibodies (MAbs) are gaining increasing importance as powerful tools in the field of virology. Surface plasmon resonance (SPR) is an optical biosensing technology already used in virus detection and in the screening of MAbs of diagnostic and therapeutic value. Rabbit haemorrhagic disease virus 2 (RHDV) and foot-and-mouth disease virus (FMDV) are top veterinary issues for whom, the development of novel methods for their detection in biological samples represents a priority with important livestock healthcare and economic implications. With these premises, here we prepared a series of SPR biosensors containing RHDV2 or its 6S subunit immobilized to the surface by different strategies. The biosensors were then used to characterize the binding capacity of a panel of anti-RHDV2 MAbs. From the comparison of the results obtained, the biosensor composed of intact RHDV2 captured with catcher-MAb covalently immobilized to the surface showed the best analytical performances, that were retained also when the same strategy was adopted to prepare a biosensor containing a different virus (namely, FMVD). The results obtained are discussed in view of the exploitation of SPR in the rapid, sensitive and resilient detection of viruses in biological materials and in the screening of antiviral MAbs libraries.
... Transgene expression in the liver after Ad5 immunization (intravenous and intramuscular) of mice and rats has been previously reported [10,24], but not after Ad26 immunization (intravenous or intramuscular) of mice [44]. Ad5 has been shown to transduce liver cells through factors IX [45] and X [44,46] mediated CAR interactions, whereas these interactions have not been shown for Ad26 so far, explaining the low or undetectable signal in the liver of the mice immunized with Ad26 compared with Ad5. Interestingly, Ad5 has been reported to distribute to the liver and spleen but not to draining lymph nodes in rabbits [47] indicating either a lower sensitivity of the method used in this report (qPCR) or differences in the tropism of the transduced trafficked cells between species. ...
Article
Full-text available
Non-replicating adenovirus-based vectors have been broadly used for the development of prophylactic vaccines in humans and are licensed for COVID-19 and Ebola virus disease prevention. Adenovirus-based vectored vaccines encode for one or more disease specific transgenes with the aim to induce protective immunity against the target disease. The magnitude and duration of transgene expression of adenovirus 5- based vectors (human type C) in the host are key factors influencing antigen presentation and adaptive immune responses. Here we characterize the magnitude, duration, and organ biodistribution of transgene expression after single intramuscular administration of adenovirus 26-based vector vaccines in mice and evaluate the differences with adenovirus 5-based vector vaccine to understand if this is universally applicable across serotypes. We demonstrate a correlation between peak transgene expression early after adenovirus 26-based vaccination and transgene-specific cellular and humoral immune responses for a model antigen and SARS-CoV-2 spike protein, independent of innate immune activation. Notably, the memory immune response was similar in mice immunized with adenovirus 26-based vaccine and adenovirus 5-based vaccine, despite the latter inducing a higher peak of transgene expression early after immunization and a longer duration of transgene expression. Together these results provide further insights into the mode of action of adenovirus 26-based vector vaccines.
... In addition to IgM and complement proteins, blood coagulation factors also bind HAdv and influence its tropism [18]. Coagulation factor X (FX) binds to the hexon proteins of the viral capsid with picomolar affinity [19][20][21]. This is in contrast to IgM, which binds via high avidity rather than affinity, due to its multivalent interaction with the capsid surface through its Fab arms [5]. ...
Article
Full-text available
Adenovirus has strong therapeutic potential as an oncolytic virus and gene therapy vector. However, injecting human species C serotype 5 adenovirus, HAdv-C5, into the bloodstream leads to numerous interactions with plasma proteins that affect viral tropism and biodistribution, and can lead to potent immune responses and viral neutralization. The HAdv/factor X (FX) interaction facilitates highly efficient liver transduction and protects virus particles from complement-mediated neutralization after intravenous delivery. Ablating the FX interaction site on the HAdv-C5 capsid leaves the virus susceptible to neutralization by natural IgM followed by activation of the complement cascade and covalent binding of complement components C4b and C3b to the viral capsid. Here we present structural models for IgM and complement components C1, C4b, and C3b in complex with HAdv-C5. Molecular dynamics simulations indicate that when C3b binds near the vertex, multiple stabilizing interactions can be formed between C3b, penton base, and fiber. These interactions may stabilize the vertex region of the capsid and prevent release of the virally encoded membrane lytic factor, protein VI, which is packaged inside of the viral capsid, thus effectively neutralizing the virus. In a situation where FX and IgM are competing for binding to the capsid, IgM may not be able to form a bent conformation in which most of its Fab arms interact with the capsid. Our structural modeling of the competitive interaction of FX and IgM with HAdv-C5 allows us to propose a mechanistic model for FX inhibition of IgM-mediated virus neutralization. According to this model, although IgM may bind to the capsid, in the presence of FX it will likely retain a planar conformation and thus be unable to promote activation of the complement cascade at the virus surface.
... HAdV group C5 has a good affinity for hepatocytes. It has shown that coagulation factors containing GLA domain can mediate the entry of the virus into hepatocytes by binding to major viral capsid proteins, such as FX, FIX, and FVI, among which FX has the highest affinity with the capsid protein of the HAdV group C5 [5,6]. This may be part of the reason for the severe coagulopathy. ...
Article
Fibroblast activation protein (FAP) is a cell surface serine protease that is highly expressed on reactive stromal fibroblasts, such as cancer-associated fibroblasts (CAFs), and generally absent in healthy adult tissues. FAP expression in the tumor stroma has been detected in more than 90% of all carcinomas, rendering CAFs excellent target cells for a tumor site-specific adenoviral delivery of cancer therapeutics. Here, we present a tropism-modified human adenovirus 5 (Ad5) vector that targets FAP through trivalent, designed ankyrin repeat protein (DARPin)-based retargeting adapters. We describe the development and validation of these adapters via cell-based screening assays and demonstrate adapter-mediated Ad5 retargeting to FAP+ fibroblasts in vitro and in vivo. We further show efficient in vivo delivery and in-situ production of a therapeutic payload by CAFs in the tumor microenvironment (TME), resulting in attenuated tumor growth. We thus propose using our FAP-Ad5 vector to convert CAFs into a 'biofactory', secreting encoded cancer therapeutics into the TME to enable a safe and effective cancer treatment.
Article
Full-text available
Objective To know the status quo of research on vitamins and depression. Method The precise search was conducted in the Web of Science Database, and the target literature assemblies of researches on "vitamins" and "depression" were identified. Use the database online analysis tool and NoteExpress software analysis module to carry out general quantitative analysis, and use CiteSpace software to carry out network clustering co-occurrence analysis. A systematic review was carried out against the closely related high-quality literature, combining the results of the bibliometric analysis. Result The search finally yielded 2817 articles which were mainly published in the journals of nutrition and psychiatry, and there was 3 timepoint with relatively significant growth. The literature from the USA and Europe accounted for the vast majority. Nine of the top 20 most-cited articles were from the USA (45%) , while the most prolific authors and high-frequency citations came from Australia and European countries. Keywords such as vitamin D, oxidative stress, vitamin B 12 , folic acid, vitamin E, vitamin A, and meta-analysis have received much attention; the research frontier includes biomarkers, gut microbiota, serotonin, systematic review, etc. Most-cited citations (7 / 20) , frontier citations (9 / 14) , highest intensity citations (13 / 20) , and longest duration citations (18 / 22) are studies on vitamin D and mental health (depression-like). The keyword time-zone shows a wave of high-frequency words emerging around 2012, including " stress", "inflammation", " serum 25 hydroxyvitamin D ", " vitamin D supplementation " and " C-reactive protein". The citation time-zone shows three-wave gatherings emerging with the last one being denser, while the citation clustering timeline shows a distinct temporal trend for different clusters. Conclusion Research on vitamins and depression underwent a paradigm shift in 2013, with vitamin D and oxidative stress dominating the field, gut microbiota, serotonin, biomarker, and systematic review being research frontiers. Research is unevenly distributed regionally, with the USA leading the way; however, the most prolific and influential authors and literature are from Australia and Eu-rope countries. Meta-analysis and systematic review are still trusted by researchers.
Preprint
Human norovirus (HuNoV) is a leading cause of acute viral gastroenteritis, but despite high impact on public health and health care, the mechanisms of viral attachment to and entry into target cells are not yet fully understood. It is well known that body fluids such as blood can transmit unrelated viruses, but recent reports also indicate that saliva and bile contribute to transmission of HuNoV. For example, human bile acids increase cell surface ceramide levels in human enteroids, which improves norovirus entry into cells resulting in enhanced replication. Bile acids can also interact directly with the norovirus capsid, but it is not known whether bile or other gastrointestinal body fluids directly affect HuNoV attachment to host cells. In this study, we investigated the effects of patient-derived gastric juice, pancreatic juice, and bile on HuNoV GII.4 virus-like particle (VLP) attachment to and entry into a human duodenal cell line, HuTu-80. We show that while gastric juice and pancreatic juice do not affect viral attachment or entry, bile – in particular hydrophobic bile acids – significantly enhance cellular attachment and subsequent entry of GII.4 VLPs into cells. In addition, we show that hydrophobic bile acids induce accumulation of viral particles in the vicinity of cells. Our results suggest the presence of a new en masse infection mechanism, where bile acids aggregate virions, and allow direct and more efficient attachment to and entry into target cells. Importance Viruses transmitted by the fecal-oral route encounter secreted host factors in gastrointestinal fluids. Some host factors can be exploited by the virus to facilitate infection. Human bile acids indirectly promote norovirus entry into and infection of human enteroids, but the direct effect of bile acids on attachment and uptake, along with the impact of other gastrointestinal fluids, remain unknown. Here, we investigated the direct effects of human body fluids on cellular attachment of norovirus VLPs. We show that human bile and hydrophobic bile acids induce an accumulation of norovirus VLPs, which is associated with significantly enhanced attachment and entry into human duodenal cell lines. These results highlight the differential effects of gastrointestinal body fluids on viral attachment and entry, while providing useful information into the complex HuNoV-host interactions that facilitate infection.
Article
Full-text available
Vectors containing group B adenovirus (Ad) fibers have been shown to efficiently transduce gene therapy targets that are refractory to infection with standard Ad serotype 5 (Ad5) vectors, including malignant tumor cells, hematopoietic stem cells, and dendritic cells. Preliminary studies in mice indicate that B-group fiber containing Ads do not efficiently transduce most organs and cause less acute toxicity than Ad5 vectors after intravenous injection. However, biodistribution and safety studies in mice are of limited value because the mouse analogue of the B-group Ad receptor, CD46, is expressed only in the testis, while in humans, CD46 is expressed on all nucleated cells. Unlike mice, baboons have CD46 expression patterns and levels that closely mimic those in humans. We conducted a biodistribution and toxicity study of group B Ad vectors in baboons. Animals received intravenous injection of PBS, a standard Ad5 vector, or chimeric vectors containing B-group Ad11 or Ad35 fibers (Ad5/35 or Ad5/11) at dose of 2×1012 vp/kg and biodistribution and safety was analyzed over 3 days. Analysis of Ad5/35 and Ad5/11 vector genome distribution by quantitative PCR revealed uptake into most tissues at levels 1-3 orders of magnitude below that of Ad5. Significant Ad5/35 and Ad5/11-mediated transgene (b-galactosidase) expression was only seen in the marginal zone of splenic follicles. Compared to the animal that received the Ad5 vector, all animals injected with B-group fiber containing Ad vectors had lower elevations in serum pro-inflammatory cytokine levels. Gross and histopathology were normal in animals that received B-group Ad fiber containing Ads, in contrast to the Ad5 infused animal which showed widespread endothelial damage and inflammation (which is in agreement with studies performed by others). In a further study, a chimeric Ad5/35 vector carrying pro-apoptotic TRAIL and Ad E1A genes under tumor-specific regulation was well tolerated in a 30 day toxicity study. No major clinical, serologic or pathologic abnormalities were noticed in this animal. Overall, this study suggests that Ad vectors possessing B-group Ad fibers have a better safety profile after intravenous injection than conventional Ad5-based vectors. This makes them potential candidates for gene therapy, for example in treatment of metastatic cancer or cardiovascular diseases.
Article
Full-text available
The efficiency of first-generation adenoviral vectors as gene delivery tools is often limited by the short duration of transgene expression, which can be related to immune responses and to toxic effects of viral proteins. In addition, readministration is usually ineffective unless the animals are immunocompromised or a different adenovirus serotype is used. Recently, adenoviral vectors devoid of all viral coding sequences (helper-dependent or gutless vectors) have been developed to avoid expression of viral proteins. In mice, liver-directed gene transfer with AdSTK109, a helper-dependent adenoviral (Ad) vector containing the human α1-antitrypsin (hAAT) gene, resulted in sustained expression for longer than 10 months with negligible toxicity to the liver. In the present report, we have examined the duration of expression of AdSTK109 in the liver of baboons and compared it to first-generation vectors expressing hAAT. Transgene expression was limited to approximately 3–5 months with the first-generation vectors. In contrast, administration of AdSTK109 resulted in transgene expression for longer than a year in two of three baboons. We have also investigated the feasibility of circumventing the humoral response to the virus by sequential administration of vectors of different serotypes. We found that the ineffectiveness of readministration due to the humoral response to an Ad5 first-generation vector was overcome by use of an Ad2-based vector expressing hAAT. These data suggest that long-term expression of transgenes should be possible by combining the reduced immunogenicity and toxicity of helper-dependent vectors with sequential delivery of vectors of different serotypes.
Article
Full-text available
In well-differentiated human airway epithelia, the coxsackie B and adenovirus type 2 and 5 receptor (CAR) resides primarily on the basolateral membrane. This location may explain the observation that gene transfer is inefficient when adenovirus vectors are applied to the apical surface. To further test this hypothesis and to investigate requirements and barriers to apical gene transfer to differentiated human airway epithelia, we expressed CAR in which the transmembrane and cytoplasmic tail were replaced by a glycosyl-phosphatidylinositol (GPI) anchor (GPI-CAR). As controls, we expressed wild-type CAR and CAR lacking the cytoplasmic domain (Tailless-CAR). All three constructs enhanced gene transfer with similar efficiencies in fibroblasts. In airway epithelia, GPI-CAR localized specifically to the apical membrane, where it bound adenovirus and enhanced gene transfer to levels obtained when vector was applied to the basolateral membrane. Moreover, GPI-CAR facilitated gene transfer of the cystic fibrosis transmembrane conductance regulator to cystic fibrosis airway epithelia, correcting the Cl(-) transport defect. In contrast, when we expressed wild-type CAR it localized to the basolateral membrane and failed to increase apical gene transfer. Only a small amount of Tailless-CAR resided in the apical membrane, and the effects on apical virus binding and gene transfer were minimal. These data indicate that binding of adenovirus to an apical membrane receptor is sufficient to mediate effective gene transfer to human airway epithelia and that the cytoplasmic domain of CAR is not required for this process. The results suggest that targeting apical receptors in differentiated airway epithelia may be sufficient for gene transfer in the genetic disease cystic fibrosis.
Article
Full-text available
Systemic administration of adenoviral vectors leads to a widespread distribution of vector. Therefore, targeting of adenoviral vectors to specific tissues or cell types will require methods to ablate the normal tropism of the vector simultaneously with the introduction of new receptor specificities. To inhibit native receptor binding, we mutated residues in the AB loop of the adenovirus type 5 (Ad5) fiber. We genetically incorporated the S408E-P409A mutation, referred to as KO1, into the adenoviral genome alone or in combination with an RGD-targeting ligand in the HI loop of fiber. Transduction experiments confirmed that the KO1 mutation results in a significant reduction in fiber-dependent gene transfer on A549 and primary fibroblast cells that could be restored via the RGD-targeting ligand. Competition transduction experiments verified the receptor-binding properties of each vector on A549 and hepatocytes in vitro. Unexpectedly, in mice systemic delivery of the vector containing the KO1 mutation resulted in efficient liver transduction that was localized specifically to hepatocytes. We confirmed these results in three different mouse strains, indicating that hepatic adenoviral gene transfer may be independent of the coxsackievirus-adenovirus receptor and that in vivo retargeting will require further viral capsid modifications to generate a fully detargeted adenoviral vector upon which to introduce new tropisms.
Article
A complementary DNA clone has been isolated that encodes a coxsackievirus and adenovirus receptor (CAR). When transfected with CAR complementary DNA, nonpermissive hamster cells became susceptible to coxsackie B virus attachment and infection. Furthermore, consistent with previous studies demonstrating that adenovirus infection depends on attachment of a viral fiber to the target cell, CAR-transfected hamster cells bound adenovirus in a fiber-dependent fashion and showed a 100-fold increase in susceptibility to virus-mediated gene transfer. Identification of CAR as a receptor for these two unrelated and structurally distinct viral pathogens is important for understanding viral pathogenesis and has implications for therapeutic gene delivery with adenovirus vectors.
Article
The subgroup C of the adenoviruses (Ad) and the group B coxsackieviruses (CVB) are structurally unrelated viruses that are known to compete for an unidentified cell surface receptor. We now describe the isolation of cDNAs from human and mouse that encode the human CVB and Ad2 and 5 receptor (HCAR) and the mouse CVB Ad2 and 5 receptor (MCAR). Both are 46-kDa glycoproteins whose primary amino acid sequences are highly homologous. Structurally, HCAR and MCAR appear to be transmembrane proteins that contain two extracellular immunoglobulin-like domains and therefore belong to this superfamily. Transfection of either of these cDNA molecules into receptor-negative NIH 3T3 cells conferred susceptibility to CVB infection and permitted the expression of beta-galactosidase from a recombinant Ad5 vector. In addition, HCAR and MCAR mRNAs could be detected on Northern blots of oligo(dT)-selected RNA from receptor-positive HeLa cells and TCMK-1 as well as several tissues of human and mouse origin that are known to be targets for Ad and CVB infections. Finally, Western blots using antibodies that inhibit virus binding to either the human or mouse CVB receptors detected 46-kDa proteins in HCAR- and MCAR-transfected cells, respectively. Taken together, these results confirm that the isolated cDNAs encode the receptors for the subgroup C Ad and CVB.
Article
The quality of optical biosensor data must be improved in order to characterize the mechanism and rate constants associated with molecular interactions. Many of the artifacts associated with binding data can be minimized or eliminated by designing the experiment properly, collecting data under optimum conditions and processing the data with reference surfaces. It is possible to globally fit high-quality biosensor data with simple bimolecular reaction models, which validates the technology as a biophysical tool for interaction analysis.
Article
Intravenous administration of adenoviral vectors results mostly in hepatocyte transduction and subsequent hepatotoxicity. Because hepatocytes express high levels of the primary adenovirus receptor CAR, untargeting hepatocytes requires CAR-binding ablation. The amino acid residues of the viral fiber responsible for CAR-binding are known. We have constructed a mutant adenoviral vector unable to bind CAR and studied vector biodistribution and hepatotoxicity after intravenous administration. In contrast to a vector with wild-type fiber, the infectivity of the CAR-ablated vector is greatly reduced and not susceptible to inhibition with wild-type knob. Biodistribution and hepatotoxicity are, however, not affected by CAR-binding ablation. A possible explanation could be related to an increased blood persistence detected for the CAR-ablated vectors combined with their residual infectivity through other receptors.
Article
Adenovirus binds its receptor (CAR), enters cells, and replicates. It must then escape to the environment to infect a new host. We found that following infection, human airway epithelia first released adenovirus to the basolateral surface. Virus then traveled between epithelial cells to emerge on the apical surface. Adenovirus fiber protein, which is produced during viral replication, facilitated apical escape. Fiber binds CAR, which sits on the basolateral membrane where it maintains tight junction integrity. When fiber bound CAR, it disrupted junctional integrity, allowing virus to filter between the cells and emerge apically. Thus, adenovirus exploits its receptor for two important but distinct steps in its life cycle: entry into host cells and escape across epithelial barriers to the environment.