ArticlePDF Available

Characterization of mAb6-9-1 monoclonal antibody against hemagglutinin of avian influenza virus H5N1 and its engineered derivative, single-chain variable fragment antibody

Authors:

Abstract and Figures

Hemagglutinin (HA), as a major surface antigen of influenza virus, is widely used as a target for production of neutralizing antibodies. Monoclonal antibody, mAb6-9-1, directed against HA of highly pathogenic avian influenza virus A/swan/Poland/305-135V08/2006(H5N1) was purified from mouse hybridoma cells culture and characterized. The antigenic specificity of mAb6-9-1 was verified by testing its cross-reactivity with several variants of HA. The mimotopes recognized by mAb6-9-1 were selected from two types of phage display peptide libraries. The comparative structural model of the HA variant used for antibody generation was developed to further facilitate epitope mapping. Based on the sequences of the affinity- selected polypeptides and the structural model of HA the epitope was located to the region near the receptor binding site (RBS). Such localization of the epitope recognized by mAb6-9-1 is in concordance with its moderate hemagglutination inhibiting activity and its antigenic specificity. Additionally, total RNA isolated from the hybridoma cell line secreting mAb6-9-1 was used for obtaining two variants of cDNA encoding recombinant single-chain variable fragment (scFv) antibody. To ensure high production level and solubility in bacterial expression system, the scFv fragments were produced as chimeric proteins in fusion with thioredoxin or displayed on a phage surface after cloning into the phagemid vector. Specificity and affinity of the recombinant soluble and phage-bound scFv were assayed by suitable variants of ELISA test. The observed differences in specificity were discussed.
Content may be subject to copyright.
Regular paper
Characterization of mAb6-9-1 monoclonal antibody against
hemagglutinin of avian inuenza virus H5N1 and its engineered
derivative, single-chain variable fragment antibody
Róża Sawicka1, Paweł Siedlecki1, Barbara Kalenik1, Jan P. Radomski2, Violetta Sączyńska3,
Anna Porębska3, Bogusław Szewczyk4, Agnieszka Sirko1 and Anna Góra-Sochacka1*
1Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland; 2Interdisciplinary Centre for Mathematical and Computa-
tional Modelling, Warsaw University, Warsaw, Poland; 3Institute of Biotechnology and Antibiotics, Warsaw, Poland; 4Department of Recombinant
Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland
Hemagglutinin (HA), as a major surface antigen of inu-
enza virus, is widely used as a target for production of
neutralizing antibodies. Monoclonal antibody, mAb6-9-1,
directed against HA of highly pathogenic avian inuenza
virus A/swan/Poland/305-135V08/2006(H5N1) was puri-
ed from mouse hybridoma cells culture and character-
ized. The antigenic specicity of mAb6-9-1 was veried
by testing its cross-reactivity with several variants of HA.
The mimotopes recognized by mAb6-9-1 were selected
from two types of phage display peptide libraries. The
comparative structural model of the HA variant used for
antibody generation was developed to further facilitate
epitope mapping. Based on the sequences of the ani-
ty- selected polypeptides and the structural model of HA
the epitope was located to the region near the receptor
binding site (RBS). Such localization of the epitope rec-
ognized by mAb6-9-1 is in concordance with its moder-
ate hemagglutination inhibiting activity and its antigenic
specicity. Additionally, total RNA isolated from the hy-
bridoma cell line secreting mAb6-9-1 was used for ob-
taining two variants of cDNA encoding recombinant sin-
gle-chain variable fragment (scFv) antibody. To ensure
high production level and solubility in bacterial expres-
sion system, the scFv fragments were produced as chi-
meric proteins in fusion with thioredoxin or displayed on
a phage surface after cloning into the phagemid vector.
Specicity and anity of the recombinant soluble and
phage-bound scFv were assayed by suitable variants of
ELISA test. The observed dierences in specicity were
discussed.
Key words: inuenza virus, monoclonal antibody, scFv, hemaggluti-
nin, antigenic epitope
Received: 23 March, 2016; revised: 07 October, 2016; accepted:
14 October, 2016; available on-line: 07 December, 2016
*e-mail: annag@ibb.waw.pl
Abbreviations: CDR, complementarity determining region; HA, he-
magglutinin; HI, hemagglutination inhibition test; HRP, Horseradish
Peroxidase; mAb, monoclonal antibody; PBS, phosphate-buered
saline; pfu, plaque forming unit; scFv, single-chain variable frag-
ment antibody; RBS, receptor binding site; VH, variable region of
the heavy chain; VL, variable region of the light chain
INTRODUCTION
Antibodies are used in many research applications, as
well as in various diagnostic assays and medical treat-
ments. Hybridoma technology enables production of
huge amounts of monoclonal antibodies (mAb) against
the target antigen but it is a laborious, time consuming
and expensive technique. Growing demand for recombi-
nant antibodies and their derivatives prompted the de-
velopment of various expression platforms. Up to date
many attempts of their production in mammalian cells
(Ho et al., 2006), insect cells (Choo et al., 2002), plants
(Dobhal et al., 2013), yeast or bacteria (Rouet et al., 2012)
have been reported. The derivatives of antibodies such
as the antigen-binding fragment (Fab) or the single chain
variable fragment (scFv) are of particular interest. Their
reduced structures and molecular weights facilitate ex-
pression and purication process. The scFv fragment
consists of variable regions of heavy (VH) and light (VL)
chains of immunoglobulin connected by a exible linker
(Kalenik et al., 2014). The scFv can be modied by con-
jugation to drugs or toxins and may penetrate the blood
and tissue more rapidly than a complete antibody (Cao et
al., 2012a; Oriuchi et al., 2005). Easiness of modication
gives many possibilities of direct or indirect detection of
this protein and facilitates manipulations of its activity
(afnity to antigens). These properties are especially im-
portant for such targets as rapidly evolving surface anti-
gens of inuenza virus.
The inuenza viruses belong to Orthomyxoviridae fam-
ily. Their genome consists of a single- stranded negative
sense RNA. Due to the antigenic drift and shift, result-
ing frequently in novel mutants and re-assortants that
are able to cross the species barriers, the inuenza vi-
ruses are still a serious global problem for humans and
their livestock, especially for poultry and pigs. Highly
pathogenic avian H5N1 inuenza viruses are responsible
for serious economic losses to the poultry industry and
have caused numerous threats to public health. Since
1996 when H5N1 virus was isolated for the rst time
in China it has spread rapidly and continued to evolve,
which resulted in a periodic emergence of new phyloge-
netic groups in several regions of the world. Currently,
H5N1 viruses are divided into multiple clades and sub-
clades based on antigenic variation (Smith et al., 2012;
Smith et al., 2015; Li et al., 2013). Hemagglutinin (HA),
a strong surface antigen, is synthesized as a precursor,
HA0 molecule, which is cleaved into two subunits, HA1
and HA2 (Szewczyk et al., 2014). The HA1 subunit en-
compasses the membrane-distal globular head containing
the receptor binding site (RBS) and the vestigial esterase
domain. The HA2 subunit encompasses the stalk region
(Velkov, 2013). The main functions of HA during virus
infection are attachment of the virus to the host cell re-
Epub: No 2016_1292
Vol. 63, 2016
https://doi.org/10.18388/abp.2016_1292
2 2016R. Sawicka and others
ceptor and mediation of the fusion of the virus enve-
lope with the endosomal membrane after endocytosis.
This glycoprotein is a major target for anti-virus antibod-
ies and a subject of high antigenic variability (antigenic
drift). Identication and characterization of its antigenic
epitopes is needed for understanding of the molecu-
lar mechanisms of antigenic drift and viral immune es-
cape, which in turn might result in better selection of
the vaccine strains. The reactivity of selected monoclonal
antibodies with escape mutants of H5 HA were inten-
sively studied but the map of H5 antigenic epitopes is
still incomplete. Most of the characterized mAbs against
HA from H5N1 virus recognize the epitopes localized in
the globular head of HA, most of them overlap the RBS
and are isolate-specic, (e.g. Cao et al., 2012b; Du et al.,
2013; Wu et al., 2014), while antibodies binding to the
stem region have often a broad activity (Tan et al., 2015;
Zuo et al., 2015). Extensive review of H5 HA antigenic
sites was provided by (Velkov et al., 2013) who described
the anti-HA antibodies divided into 3 groups: (i) RBS
selective, (ii) non-RBS, membrane-distal globular domain
selective and (iii) HA2 selective.
The mAb6-9-1 is a monoclonal mouse antibody se-
creted by the hybridoma cell line 6-9-1, which was
generated by us from immune cells of a mouse im-
munized with the recombinant H5 HA from A/swan/
Poland/305-135V08/2006 (H5N1). We described usage
of Fab (antigen-binding fragment) fragment of mAb6-
9-1 in electrochemical immunosensor detecting HA
from H5N1 viruses (Jarocka et al., 2014). It was applied
for detection of HA (17-530 aa) from A/Bar-headed
Goose/Qinghai/12/05(H5N1) and HA1 subunit of HA
from two strains of H5N1 viruses: A/swan/Poland/305-
135V08/2006 and A/Vietnam/1194/2004 (Jarocka et al.,
2014). The strongest binding was observed for the long
HA from A/Bar-headed Goose/Qinghai/12/05(H5N1)
with the detection limit of 2.2 pg/mL. The successful
application of the derivatives of this antibody prompt-
ed us to perform its broaden characterization, especially
mapping of an epitope recognized by mAb6-9-1 on the
structural model of the HA from A/swan/Poland/305-
135V08/2006 (H5N1). Moreover, we described cloning,
expression and characterization of recombinant scFv
fragments derived from the mAb6-9-1. The specicity of
two variants (VLVH and VHVL) of scFv produced in
fusion with thioredoxin were evaluated by indirect and
sandwich ELISA assays and conrmed with the scFv
variant produced by the phage display method.
MATERIALS AND METHODS
Hybridoma propagation and monoclonal antibody
purication. Mouse hybridoma cell line 6-9-1 that se-
creted monoclonal antibody against hemagglutinin from
A/swan/Poland/305-135V08/2006 (H5N1) was gener-
ated from splenocytes obtained after immunization of
mice with the recombinant H5 HA. The hybridoma cells
were maintained in RPMI-1640 medium (20 mM L-glu-
tamine, 10% fetal bovine serum, 5% CO2, 37°C). The
mAb6-9-1 was puried from culture supernatant using
protein G (ThermoScientic).
Comparative modeling of HA from A/swan/
Poland/305-135V08/2006 (H5N1). Structures suitable
for modeling were chosen from a default BLAST search
against the PDB database (accessed February 5th, 2015),
with respect to the serotype (H5), X-ray resolution, and
sequence length. Two structures emerged through these
criteria: 2ibx (A/Vietnam/1194/2004 (H5N1)) and 2fk0
(A/Vietnam/1203/2004 (H5N1)), both having over
90% sequence identity. Preparation of X-ray structures
was done in Chimera (v1.11) and consisted of deletion
of non-standard residues and HOH molecules, N- and
C-terminal x and incomplete side chains repair. Both
structures were aligned with MatchMaker (Meng et al.,
2006) and the most similar chains were used as tem-
plates; 2IBX_A, 2IBX_B, 2FK0_K, 2FK0_L. Modeler
software (Sali & Blundell, 1993) was used throughout
the modeling process. Ten different models were gen-
erated and manually evaluated with respect to hydrogen
bond patterns, AA contact and clashes and Ramachan-
dran plot. The best model was subjected to a minimiza-
tion procedure using AMBER force eld (ff14SB) and
a steepest descent procedure to relieve steric hindrances
(15Å box, TIP3PBOX solvent model and Cl- to neutral-
ize the structure).
Epitopes mapping. Epitope recognition by puri-
ed mAb6-9-1 was determined using two Phage Display
Peptide Libraries, Ph.D. ™-C7C and Ph.D.™-12 (New
England Biolabs) according to the manufacturer’s pro-
tocol (Ph.D Phage Display Libraries, version 1.2, New
England Biolabs). Briey, the 96-well plates (Maxisorb
Nunc) were coated with puried mAb6-9-1 (7.5 µg/
well) and incubated at 4°C overnight. After washing with
TBST (TBS + 0.1% Tween-20) and blocking with BSA
solution (5 µg/ml), the titrated phage peptide library
(Ph.D.™-C7C or Ph.D.™-12) was added to each well
(107–108 plaque forming units (pfu) in 100 µl) and in-
cubated for 60 minutes at room temperature. Unbound
phages were discarded, plates washed 10 times with
TBST and bound phages were eluted with 0.2 M glycine-
HCl, (pH 2.2), 1 mg/ml BSA. In a parallel experiment
the OET protein (see below, Antigens and antibodies
used in ELISA) at concentration 0.8 mg/ml was used
for phage elution. Eluted phages were amplied and
taken to the next panning rounds. After three rounds
of panning several phage clones were randomly select-
ed, subjected to phage ELISA, (performed according to
the protocol described in Ph.D Phage Display Libraries,
New England Biolabs) to conrm their binding capac-
ity with mAb6-9-1. DNA isolated from individual phage
clones was sequenced and amino acid sequence of the
corresponding peptides determined. The Pepitope server
(http://pepitope.tau.ac.il/) (Mayrose et al., 2007a) was
used to map the selected peptides onto the 3D model of
HA from A/swan/Poland/305-135V08/2006 (H5N1).
Using the consensus methodology, both PepSurf (May-
rose et al., 2007b) and Mapitope algorithms (Bublil et al.,
2007) were executed and their results combined into a
single prediction including only residues predicted to be
a part of the epitope in both algorithms. Mapping results
were compared with the surface of the H5 HA structural
model using UCSF Chimera (v1.11).
Synthesis of the scFv cDNA. Primers. Sequences
of primers used in the experiment of scFv synthesis and
cloning are listed in the Supplementary le Table S1 at
www.actabp.pl.
Synthesis of VH and VL regions. Sequences encod-
ing the variable regions of light (VL) and heavy (VH)
chains were obtained according to the method and pro-
cedure described by (Ladiges & Osman, 2001). The main
steps of procedure are described compendiously below.
Total RNA was isolated from the hybridoma cell line
6-9-1 using RNeasy Plus Micro Kit according to the
manufacturer’s instructions (Qiagen). 6.5 µg of isolated
RNA and oligo (dT)15 primer were used to synthesize
blunt-ended cDNA using the cDNA Synthesis System
(Roche). Next, the PCR OG/GO linker was ligated to
Vol. 63 3Monoclonal antibody against hemagglutinin of inuenza virus H5N1
the cDNA by T4 DNA ligase (Invitrogen). The resulting
cDNA was used to amplify VH and VL fragments using
Easy-A High-Fidelity PCR Master Mix (Agilent Technol-
ogies) supplemented with the sets of primers, OG and
IgGkapp or OG and IgG1HC1 for VL and VH synthe-
sis, respectively. IgGkapp primer corresponded to the
constant region of the light chain CL whereas IgG1HCl
corresponded to the CH1 gene (sequences are provided
in Supplementary le 1: Table S1 at www.actabp.pl).The
PCR products were cloned into the pGEM-T easy vec-
tor (Promega). Several clones were sequenced and ana-
lyzed using vbase2 on-line program (http://www.vbase2.
org/vbdnaplot.php) to determine sequences of CDRs of
VH and VL.
Assembling of scFv. Selected functional clones of
light and heavy chain of mouse IgG were used to syn-
thesize scFv construct. First, to obtain scFv in VH-VL
orientation the variable fragments of heavy and light
chains were amplied using two pairs of primers either
VHNdeF and scFvVHR or scFvVKF and VkNotR.
Then VH and VL fragments were assembled into scFv
by overlapping PCR reaction using external primers
VHNdeF and VkNotR. The same method was applied
to synthesize scFv in opposite orientation (VL-VH) us-
ing specic primers: NdeVLF, linkVLR and linkVHF,
VHrevNot. The PCR reactions were performed using
Pfu Polymerase (Fermentas). Finally, PCR products were
digested with NdeI and NotI restriction enzymes and li-
gated into the modied pET26b expression vector (No-
vagen) carrying the sequence encoding His- and Strep-
tags previously cloned by us.
scFv expression in E. coli. The scFv constructs were
re-cloned from the pET26b into the modied pET201
vector carrying the sequence encoding thioredoxin
which resulted in obtaining two recombinant plasmids:
pET201/TVHVL and pET201/TVLVH that were used
for scFv overexpression in E. coli Origami (DE3) strain.
For an efcient production of recombinant TscFvs sev-
eral different conditions of temperature (37°C, 30°C,
25°C, 20°C), IPTG concentration (10 mM, 1 mM,
0.1 mM, 0.05 mM, 0.01 mM) and time of induction (be-
tween 1 h–48 h) were tested. The TscFv expressed by
E. coli were puried on a gravity ow Ni-TED 1000 col-
umn (Marcherey-Nagel) in denaturing conditions accord-
ing to the procedure described in manufacture’s manual
(Marcherey-Nagel). The rst and the second elution frac-
tions were mixed and dialyzed against stepwise decreas-
ing concentration of urea in the range between 6.0–0 M.
Each step lasted at least 3 hours. Purity of recombinant
proteins was evaluated with SDS PAGE.
Phage displayed scFv (PhscFv). The cDNA frag-
ment encoding scFv in VL-VH orientation (without
thioredoxin) was amplied by Easy A polymerase (Easy-
A High-Fidelity PCR Master Mix, Agilent Technologies)
with the RRPvuF and VHrevNot primers (sequences
are provided in Supplementary le 1: Table S1 at www.
actabp.pl) using recombinant plasmid pET26b carrying
scFv cDNA as a template. The obtained PCR prod-
uct was digested with PvuII and NotI and cloned into
pSEX81 phagemid vector (Progen). Small-Scale Phage
Rescue was conducted according to the published proto-
col 154.3.4 by (Dorsam et al., 2002) with minor changes
(rst centrifugation was prolonged to 25 min and 5 h
incubation in 37°C was changed to 18 h in 30°C). The
obtained phages (PhVLVH) were titrated according to
the online protocol ‘Helper Phage Production(Stocking-
er Lab website: http://www.oardc.ohio-state.edu/stock-
ingerlab/Protocols/HelperPhage.pdf) and used in Phage
ELISA. The ELISA was conducted according to (Dor-
sam et al., 2002) with minor modications. Briey, Max-
iSorp plates were coated with 500 ng of the antigen and
incubated overnight at 4°C, washed with PBS, blocked
(2 h, 2% skim milk in PBS), incubated with 107-108 pfu
for 2 h, washed and incubated with anti-M13 antibodies
conjugated with HRP (GE Healthcare).
Sequencing of mAb6-9-1. The amino acid sequence
of heavy and light chains of mAb6-9-1 was estimated by
mass spectrometry of trypsin-digested mAb fragments.
The analysis was performed in Mass Spectrometry Lab
at the Institute of Biochemistry and Biophysics Polish
Academy of Sciences, Warsaw.
Hemagglutination inhibition test. HI tests were
performed as described previously (Stachyra et al., 2014;
Stachyra et al., 2016) using inactivated viruses as antigens,
either the highly pathogenic A/Turkey/Poland/35/2007
(H5N1) (National Veterinary Research Institute,
Pulawy, Poland) or the low pathogenic A/chicken/Bel-
gium/150/1999 (H5N2) (DG Deventer, Netherlands).
HI titer was dened as the reciprocal of the highest dilu-
tion of sera that completely inhibited hemagglutination.
Antigens and antibodies used in ELISA. The fol-
lowing antigens corresponding to HA from A/swan/
Poland/305-135V08/2006 (H5N1) were used: (1) HA/
Nde, a short variant containing region of 17-340 aa with
His-tag at N- terminus produced (in our laboratory) in
E. coli; (2) OET, a long variant containing region of 17-
530 aa derived from baculovirus system (Oxford Expres-
sion Technologies, UK); The following recombinant HA
antigens were purchased from Immune Technology Cor-
poration (all of them are 6x His-tagged and derived from
293 cell culture): Qinghai 17-530 aa from A/Bar-head-
ed Goose/Qinghai/12/05 (H5N1); Vietnam 17-530 aa
from A/VietNam/1203/2004 (H5N1); Anhui 18-530
aa from A/Anhui/1/2005 (H5N1); Cambodia 24–341
aa from A/Cambodia/R0405050/2007 (H5N1); Hubei
18-530 aa from A/Hubei/1/2010 (H5N1); Germany
17-342 aa from A/Turkey/Germany-MV/R2472/2014
(H5N8); Wuhan 17-529 aa from A/Wuhan/359-
95(H3N2); Netherlands 17-527 aa from A/chicken/
Netherlands/1/03 (H7N7); New Caledonia 18-530 aa
from A/New Caledonia/20/99 (H1N1).
Antibodies. IgY 745 – anty-H5 HA (H5N1) poly-
clonal IgY was puried in our laboratory from eggs
collected from laying hens immunized with DNA vac-
cine based on pCI vector with cloned cDNA sequence
encoding 1-568 aa of HA from A/swan/Poland/305-
135V08/2006 (H5N1) (Stachyra et al., 2014). IgY was
puried using Pierce™ Chicken IgY purication kit
(ThermoFisher). Anti-M13 HRP-conjugated monoclo-
nal antibody (HRP/Anti-M13 Monoclonal Conjugate,
GE Healthcare); anti-Strep-tag II monoclonal antibody
(StrepMAB- Classic, IBA, GmbH); anti chicken IgY
(Goat anti-Chicken IgY Fc Secondary Antibody, HRP
conjugate, Thermo Scientic); anti-mouse HRP antibody
(Anti-Mouse IgG (γ-chain specic) Peroxidase anti-
body produced in goat, Sigma Aldrich) were purchased
as indicated.
Indirect and sandwich ELISA. Indirect ELISA.
The 96-well MaxiSorp plates (Nunc, Denmark) were
coated overnight at 4°C with 300 ng of the respective
antigens. Following 4x washing with PBST buffer (0.1 M
PBS with 0.05% Tween-20) plates were blocked with 2%
BSA in PBS for 1.5 h. Next, plates were incubated with
puried antibody (mAb6-9-1 or TscFv) overnight at 4°C
and after washing incubated (1.5 h, 37°C) with the anti-
strep-tag antibody, washed and incubated (1 h, 37°C)
with anti-mouse HRP conjugated IgG. In the case of
mAb6-9-1 the step with anti-strep incubation was omit-
4 2016R. Sawicka and others
ted. Following incubation with TMB (3,3’,5,5’ tetrameth-
ylbenzidine, Sigma-Aldrich) absorbance at 450 nm was
measured. Denatured (reduced) antigens were obtained
after incubation with 10 mM DTT for 1 h at 37°C ac-
cording to (Wu et al., 2014).
Sandwich ELISA. The same plates as in Indirect
ELISA were coated with 500 ng of the TscFv. Wash-
ing and blocking conditions were as above. Next, plates
were incubated with antigens for 1.5 h at room temper-
ature and after washing incubated (1.5 h) with IgY745
anti- H5 polyclonal IgY at various dilutions in the range
1:4 000 to 1:16 000, washed again and incubated (45 min,
room temperature) with HRP-conjugated goat anti-chick-
en IgY Fc (Thermo Scientic) 1:12 000. Following incu-
bation with TMB absorbance at 450 nm was measured.
RESULTS AND DISCUSSION
Prediction of the epitope recognized by mAb6-9-1
In order to facilitate mapping of the antigenic sites
recognized by mAb 6-9-1 the structural model of the
HA variant (A/swan/Poland/305-135V08/2006 (H5N1))
used for antibody generation was created (Fig. 1a) as de-
scribed in Materials and Methods.
Two commercial phage display peptide libraries were
used for selection of peptides that were reactive against
mAb6-9-1. In both of them, the library of random
peptides (linear dodeca- or loop-constrained heptapep-
tides in Ph.D.™-12 Phage Display Peptide Library or
Ph.D.™-C7C Phage Display Peptide Library, respective-
ly) are fused to a minor coat protein pIII of the M13
phage. After three rounds of panning several potentially
positive clones (phage-displayed peptides) were selected
for further analysis. First, they were subjected to Phage
ELISA to verify their binding capacity with mAb6-9-1,
then phage DNA was isolated and sequenced to reveal
the predicted amino acid sequence of the mimotopes
(polypeptides displayed on the surface of the phages).
Phage clones selected from the Ph.D-12 Library were
examined in three concentrations: 109, 108 and 107 pfu/
well, respectively. The absorbance value (OD450) two-
fold higher than the value of the negative control was
considered positive. The rst concentration (109 pfu/
well) was too high, since it resulted in positive signal for
all selected clones. The lower concentrations allowed to
select 7 clones having the highest reactivity among 21
phages examined (Fig. 2). Six of them expressed the
same peptide sequence, VHWDFRQWWQPS, while one
different, FPSDWWSQAWSM.
Similar analyses were performed for phage clones se-
lected from the Ph.D-7C Library and resulted in selec-
tion of four mimotopes (ETDTLTQ, KTFLSSH, PHK-
PAMN and KPYTFVG) which occurred with the similar
frequency.
The Pepitope server was used for epitope mapping
using these afnity-selected peptides and the struc-
tural model of HA from the A/Swan/Poland/305-
135V08/2006 (H5N1). The combined algorithm (con-
sensus results of Mapitope and Pepsurf algorithms) se-
lected one cluster of four HA residues (W138, D140,
F159 and W165) from 6 mimotopes that are shown on
the structural model (Fig. 1c–d). The Pepitope analysis
with only one, (FPSDWWSQAWSM) identied a larger
cluster of 8 HA residues (K135, S136, W138, D140,
A143, V147, S148 and W165) as an epitope. Both clus-
ters shared three residues (W138, D140 and W165). All
of them were localized on the globular head of the HA1
subunit in the proximity of the RBS. Moreover amino
acids W138 and D140 were localized within the anti-
genic epitope called region A, while W165 in the region
B. Antigenic epitopes (A–E) were proposed for H5N1
Figure 1. Model of three-dimensional structure of HA from
A/swan/Poland/305-135V08/2006 (H5N1) (a) and A/Viet-
nam/1194/2004 (H5N1) (b).
Both proteins are represented as Connolly surface, colored by
amino acids hydrophobicity based on Kyte-Doolittle scale (blue
– hydrophilic, red – hydrophobic) (Kyte & Doolittle, 1982). (c) en-
larged head of HA from A/swan/Poland. Yellow surface represents
the six amino acids building the RBS interface (Y107, W165, H195,
E202, L206, and Y207), dark blue depicts the four residues of the
mAb6-9-1 epitope (W138, D140, F159 and W165). W165 is shared
between these two groups and is colored green. (d) ribbon dia-
gram of overlapped structures of the part of HA head from Poland
(cyan color) and Vietnam (pink color). Two residues dening the
binding specicity of mAb6-9-1 are marked on each structure, [1]
D140 (Poland)/S (Vietnam), [2] S145 (Poland)/L (Vietnam).
Figure 2. Reactivity of individual phage clones with mAb6-9-1 in
Phage ELISA.
Plates were coated with mAb6-9-1 or BSA and to each well 108
pfu and 107 pfu were added, plates were read at 450 nm. For
each phage clone signal obtained with target protein (mAb6-9-
1) was compared with that obtained without target protein (e.g.
BSA); presented on the horizontal axis OD value is reduced by the
background (OD value for BSA). For the positive phage clones the
amino acid sequences of the dodecapeptides are provided.
Vol. 63 5Monoclonal antibody against hemagglutinin of inuenza virus H5N1
highly pathogenic avian inuenza viruses based on the
classication of the antigenic domains of H3 (from
H3N2 virus) (Peng et al., 2014). Both epitopes, A and B,
contrary to the epitopes C–E, contribute strongly to the
antigenic variation of the HPAI (highly pathogenic avian
inuenza) H5N1 viruses (Peng et al., 2014).
The conclusions from epitope mapping were veried
by both, the Hemagglutination Inhibition test and ELI-
SA assay. In the HI test ability of mAb6-9-1 to inhibit
binding of H5N1 (by RBS) to receptors on erythrocytes
surface were analyzed. The HI titer was moderately posi-
tive, equal to 32, for the homologous virus, A/Turkey/
Poland/35/2007(H5N1), while for a nonhomologous vi-
rus A/chicken/Belgium/150/1999(H5N2) (DG Deven-
ter, Netherlands) it reached only 8, which was consid-
ered as negative. The HI results suggest that the epitope
recognized by mAb6-9-1 might be localized nearby the
RBS. Similarity between both antigens used in HI test
was 86%, besides HA cleavage sites the differences were
localized around RBS and the mapped epitope (aa posi-
tions: 135, 140, 154, 100, 124, 129, 140, 142, 154, 170,
172, 184, 205, Fig. 3) which may explain the low HI titer
obtained for the nonhomologous antigen.
With ELISA assay we analyzed binding of mAb6-9-1
to various HA antigens from different clades of H5N1
viruses as well as H1, H3 and H7 HA from several se-
lected strains (Fig. 4a). The results showed that mAb6-
9-1 can recognize both, a short (HA/Nde) and a long
(OET) variants of H5 HA from A/swan/Poland/305-
135V08/2006 (H5N1), clade 2.2 as well as H5 HA from
A/Bar-headed Goose/Qinghai/12/05 (H5N1), clade 2.2
and H5 HA from A/Anhui/1/2005 (H5N1), but not the
H5 HA from A/Hubei/1/2010 (H5N1), clade 2.3.2.1,
A/Cambodia/R0405050/2007 (H5N1), clade 1 nor A/
Turkey/Germany- MV/R2472/2014 (H5N8). The data
is in concordance with observation that the most vari-
able inter-clade positions are largely located on the glob-
ular head proximal to the RBS (Velkov et al., 2013). We
also found that mAb6-9-1 does not recognize the H3
HA from A/Wuhan/359/95 (H3N2), the H1 HA from
A/New Caledonia/20/99 (H1N1) nor the H7 HA from
A/chicken/Netherlands/1/03 (H7N7) (Fig. 4a).
Reactivity with HA/Nde antigen containing only the
HA1 part of H5 HA conrmed that the epitope rec-
ognized by mAb6-9-1is located within HA1 subunit
(Fig. 4b). Moreover, ELISA conrmed that mAb6-9-1
binds probably a conformational epitope because dena-
turation of the antigens (reduction of disulde bonds
with 10 mM DTT) resulted in a complete loss of mAb6-
9-1 binding (Fig. 4b).
Comparison of the 3D structures of H5 HA proteins
from A/swan/Poland/305-135V08/2006 (H5N1) and
from A/Vietnam/1194/2004 (H5N1) (Fig. 1a, b and d)
and an alignment of amino acid sequences of the H5
HA antigens tested by ELISA and in HI tests (Fig. 3)
clearly showed that two amino acids, D140 and S145
in the proximity of the predicted epitope might decide
about antigen binding specicity of mAb6-9-1. None of
the analyzed HA proteins with the residue other than as-
partic acid in the position [1] i.e. D140 and other than
serine in the position [2] i.e. S145 was recognized by
mAb6-9-1.
Construction and expression of the scFv – mAb6-9-1
derivative
The cDNA fragments encoding variable regions of
heavy and light chains of mAb6-9-1 were cloned into
the pGEM Teasy vector and sequenced. Each of four
selected clones of VH carried the same (functional) se-
quence. Similarly, one functional sequence was found
among thirty analyzed clones carrying VL, however, ma-
jority of them were classied as nonfunctional due to
an internal stop codon in CDR3. Taking into account
the nature of hybridoma cells such nding is not sur-
prising and was reported previously (Toleikis & Frenzel,
2012). The predicted amino acid sequences of the CDRs
in both variable chains are shown in Table 1. Sequenc-
es encoding VL and VH were assembled in two possi-
ble orientations (VL-VH and VH-VL) with the exible
linker (Gly4Ser)3 into scFv and cloned into the modi-
ed pET201vector downstream the sequence encoding
thioredoxin (Supplementary le Fig. S1 at www.actabp.
pl). The choice of thioredoxin as the fusion partner was
based on the literature data indicating that the formation
of disulde was essential for many scFvs described (Ju-
rado et al., 2006; Sonoda et al., 2010).
Finally, two recombinant variants of scFv in fusion
with thioredoxin (TscFv: TVHVL and TVLVH) were
Table 1 The sequences of complementarity determining regions (CDRs) of scFv6-9-1.
CDR1 CDR2 CDR3
Light chain
Heavy chain
SSVNY
GYSITSDYA
YTS
ISYSGST
QQFTSSPWT
ARSGISYYFGTDY
Amino acids complementarity which determines CDRs of the VH and VL domains were estimated according to the international ImMunoGeneTics
information system® (IMGT, http://www.imgt.org)
Figure 3. Alignment of the fragments of HA antigens in the region adjacent to the mapped epitope of mAb6-9-1.
The residues corresponding to those forming the RBS according to (Al-Majhdi, 2007) and those identied by the Pepitope software
(W138, D140, F159, W165) as the potential target of mAb6-9-1 are marked above the alignment by asterisks and triangles, respectively.
The positions that seem to dene binding specicity of mAb6-9-1 are marked as white letters on a black background and additionally
identied as [1] and [2] below the alignment. Belgium (H5N2) was used only in HI test. For the full names of the viruses refer to the text.
6 2016R. Sawicka and others
Figure 4. Reactivity of mAb6-9-1.
(a) Indirect ELISA with HA antigens in the native form. (b) Indirect ELISA with the selected HA antigens in the native and the reduced
forms. C(-), negative control. The plates were coated with 300 ng of HA from the indicated viruses. Puried mAb6-9-1 and the anti-
mouse IgG conjugated to HRP were used as primary and secondary antibody. The full names of the viruses are provided in the text.
Figure 5. Verication of antigenic specicity of scFv.
(a) Indirect ELISA using TscFv. The plates were coated with 300 ng of the indicated HA antigens or BSA as a negative control, recom-
binant TscFv (TVHVL, TVLVH) were used as primary antibody, while anti-strep IgG served as secondary antibody. (b) Phage ELISA using
phage displayed scFv (PhscFv). Plates were coated with 500 ng of the indicated antigens, incubated with phages (PhVLVH or the nega-
tive control) and with anti-M13 IgG as secondary antibody. (c) and (d) sandwich ELISA using TVHVL and TVLVH, respectively. The plates
were coated with 500 ng of TVHVL or TVLVH, incubated with the indicated HA antigens (250-31 ng/well) or BSA (c-), negative control,
next incubated with anti-H5 HA polyclonal chicken IgY and with anti-chicken IgG-HRP. (e) sandwich ELISA using TVLVH. The plates were
coated with 500 ng/well of TVLVH, concentrations of HA antigens and dilutions of anti-H5 HA polyclonal chicken IgY were as indicated.
C1(-), C2(-) and C3(-) various negative controls: ELISA performed without HA antigen, without IgY 745, and without antigen and IgY745,
respectively.
Vol. 63 7Monoclonal antibody against hemagglutinin of inuenza virus H5N1
produced in E. coli and puried on Ni-TDA agarose.
SDS-PAGE analysis indicated the high purity of TscFvs
obtained and their expected molecular weights to be
about 42 kDa (Supplementary le Fig. S1b at www.act-
abp.pl).
Antigen binding specicity of the recombinant
scFv 6-9-1
Irrespectively of the TscFv variant, in the indi-
rect ELISA the strongest binding was observed with
HA/Nde short H5 HA (17-340aa) from A/swan/
Poland/305-135V08/2006 (H5N1) (Fig. 5a). Surprisingly,
the TscFv failed to recognize H5 HA from A/Bar-head-
ed Goose/Qinghai/12/05 (H5N1), the protein previous-
ly very well recognized by mAb6-9-1 (Fig. 4a and 5a).
Higher plasticity of scFvs in comparison to the full-
size antibodies may affect their specicity and afnity
(Shepelyakovskaya et al., 2011). To verify if this change
of specicity was due to incorrect scFv folding a phage
displayed variant of scFv (PhVLVH) was obtained. Pub-
lished data showed that the conversion of scFv from
phage-bound to soluble form may results in change of
specicity because the phage pIII protein may provide
structural support for antigen- binding site (Kaku et al.,
2012). The results of phage ELISA (Fig. 5b) showed that
indeed PhVLVH was able to specically recognize the
HA antigen from A/Bar-headed Goose/Qinghai/12/05
(H5N1). These results strongly support the conclusion
that the cloned cDNA encoding the variable regions of
heavy and light chains correspond to the heavy and light
chains of mAb6-9-1. Moreover, comparison of the ami-
no acids sequences determined by mass spectroscopy of
trypsin-digested mAb6-9-1 with CDRs sequences of VH
and VL predicted from nucleotide sequences of cloned
scFv showed 87% and 100% identity, respectively (Sup-
plementary le Fig. S2). The alteration of antigen speci-
city observed by indirect ELISA prompted us to search
for a better method of testing the activity of TscFv.
We performed sandwich ELISA using TscFvs (TVH-
VL and TVLVH, 500ng/well) as capture antibodies, an-
ti-H5 HA IgY 745 as detection antibody and various HA
antigens. TscFvs were capable of targeting not only the
short fragment of H5 HA (HA/Nde) but also the long
fragment of H5 HA from A/Bar-headed Goose/Qing-
hai/12/05 (H5N1) (Fig. 5c–d).
The H5 HA from A/Anhui/1/2005 (H5N1; clade
2.3.4) was also weakly recognized by TscFv, while bind-
ing to H5 HA from A/Vietnam/1203/2004 (H5N1;
clade 1) and to H7 HA from A/chicken/Nether-
lands/1/03 (H7N7) was not detected (Fig. 5c–d). Ob-
tained results showed that binding capacity of TscFv to
the long H5 HA version may be stronger than to the
short H5 HA (HA/Nde) (Fig. 5c–d). This observation
was conrmed in detailed analysis when serial two fold
dilutions of both HA antigens in the range of 250–30
ng/well and anti-H5 HA IgY (1:4 000–1:16 000) were
applied (Fig. 5e). All data from sandwich ELISA were
in accordance with the results obtained for mAb6-9-1
(Fig. 4a).
The observed discrepancies of reactivity of TscFv to-
ward the long version of H5 HA, in two kinds of ELI-
SA, indirect and sandwich (Fig. 5a and 5c, d, e), may
be explained by technical inconsistencies. The lack of
binding to the long antigen was observed in the indirect
assay, when HA antigens were used to coat the plates
and the TscFv bound to the HA antigen was detected
by the anti-Strep antibody. However, the binding with
the same antigen was observed in the sandwich ELISA,
when TscFv was used to coat the plates and HA an-
tigens bound to TscFv were recognized by the specic
polyclonal anti-HA IgY antibody, detected subsequently
by secondary anti-chicken IgY antibody conjugated with
HRP. Due to the high plasticity of scFv, in the indirect
ELISA, the Strep-tag was probably hidden in the confor-
mational structure of TscFv and was not accessible for
the anti-Strep antibody.
In conclusion, epitope recognized by the full-length
monoclonal antibody was mapped in the proximity of
the RBS. This assumption is supported by the results of
HI assay. Unfortunately, contrary to mAb6-9-1, TscFv
did not demonstrate HI activity (not shown). However,
such result does not exclude that scFv recognized the
same epitope as mAb. To prevent hemagglutination, an-
tibody must bind to or near RBS and cause steric hin-
drance (Ascione et al., 2009; Zhang et al., 2013). It is
more likely that the reduced size of scFv is not an effec-
tive barrier between virus and the red cell. Both antibod-
ies display narrow specicity to H5 HA from H5N1 in-
uenza viruses, mostly from clade 2.2 and might be used
effectively in immunosensors specically detecting these
type of viruses (Jarocka et al., 2014; Jarocka et al., 2016).
Acknowledgments
The authors thank Dr. Tomasz Sarnowski (IBB PAN)
for the thioredoxin vector, Agata Malinowska for LC-MS
analysis, and Anna Stachyra, Dr Anna Olszewska, Prof.
Zenon Minta and Prof. Krzysztof Śmietanka for sharing
the results of HI test.
Acknowledgements of nancial support
This work was done in frame of the Polish Vaccine
Consortium and it was supported by EC Innovative
Economy Program, POIG.01.01.02-00-007/08.
REFERENCES
Al-Majhdi F (2007) Structure of the sialic acid binding site in inuenza
A virus: hemagglutinin. J Biol Sci 7: 113–122
Ascione A, Capecchi B, Campitelli L, Imperiale V, Flego M, Zamboni
S, Gellini M, Alberini I, Pittiglio E, Donatelli I, Temperton NJ,
Cianfriglia M (2009) Human monoclonal antibodies in single chain
fragment variable format with potent neutralization activity against
inuenza virus H5N1. Antiviral Res 83: 238–244. doi: 10.1016/j.an-
tiviral.2009.05.005
Bublil EM, Freund NT, Mayrose I, Penn O, Roitburd-Berman A, Ru-
binstein ND, Pupko T, Gershoni JM (2007) Stepwise prediction of
conformational discontinuous B-Cell Epitopes using the mapitope
algorithm. Proteins-Structure Function and Bioinformatics 68: 294–304.
doi: 10.1002/prot.21387
Cao Y, Marks JD, Huang Q, Rudnick SI, Xiong C, Hittelman WN,
Wen X, Marks JW, Cheung LH, Boland K, Li C, Adams GP, Ro-
senblum MG (2012a) Single-chain antibody-based immunotoxins
targeting Her2/neu: design optimization and impact of afnity on
antitumor efcacy and off-target toxicity. Mol Cancer Therap 11: 143–
153. doi: 10.1158/1535-7163.MCT-11-0519
Cao Z, Meng J, Li X, Wu R, Huang Y, He Y (2012b) The epitope and
neutralization mechanism of AVFluIgG01, a broad-reactive human
monoclonal antibody against H5N1 inuenza virus. Plos One 7. doi:
10.1371/journal.pone.0038126
Choo AB, Dunn RD, Broady KW, Raison RL (2002) Soluble expres-
sion of a functional recombinant cytolytic immunotoxin in insect
cells. Protein Expr Purif 24: 338–347. doi: 10.1006/prep.2001.1589
Dobhal S, Chaudhary VK, Singh A, Pandey D, Kumar A, Agrawal S
(2013) Expression of recombinant antibody (single chain antibody
fragment) in transgenic plant Nicotiana tabacum cv. Xanthi. Mol Biol
Rep 40: 7027–7037. doi: 10.1007/s11033-013-2822-x
Dorsam H, Braunagel M, Kleist C, Moynet D, Welschof M (2002)
Screening of phage displayed antibody libraries. The Protein Protocols
Handbook 1073–1082
Du L, Jin L, Zhao G, Sun S, Li J, Yu H, Li Y, Zheng B-J, Liddington
RC, Zhou Y, Jiang S (2013) Identication and structural charac-
terization of a broadly neutralizing antibody targeting a novel con-
8 2016R. Sawicka and others
served epitope on the inuenza virus H5N1 hemagglutinin. J Virol
87: 2215–2225
Ho M, Nagata S, Pastan I (2006) Isolation of anti-CD22 Fv with high
afnity by Fv display on human cells. Proc Natl Acad Sci U S A 103:
9637–9642
Jarocka U, Sawicka R, Gora-Sochacka A, Sirko A, Dehaen W, Radecki
J, Radecka H (2016) An electrochemical immunosensor based on
a4,4-thiobisbenzenethiol self-assembled monolayer for the detec-
tionof hemagglutinin from avian inuenza virus H5N1. Sensors and
Acttuators B: Chemical 228: 25–30
Jarocka U, Sawicka R, Góra-Sochacka A, Sirko A, Zagórski-Ostoja W,
Radecki J, Radecka H (2014) An immunosensor based on antibody
binding fragments attached to gold nanoparticles for the detection
of peptides derived from avian inuenza hemagglutinin H5. Sensors
(Basel) 14: 15714–15728. doi: 10.3390/s140915714
Jurado P, de Lorenzo V, Fernandez LA (2006) Thioredoxin fusions in-
crease folding of single chain Fv antibodies in the cytoplasm of Es-
cherichia coli: Evidence that chaperone activity is the prime effect of
thioredoxin. J Mol Biol 357: 49–61. doi: 10.1016/j.jmb.2005.12.058
Kaku Y, Noguchi A, Okutani A, Inoue S, Tanabayashi K, Yamamoto
Y, Hotta A, Suzuki M, Sugiura N, Yamada A (2012) Altered speci-
city of single-chain antibody fragments bound to pandemic H1N1-
2009 inuenza virus after conversion of the phage-bound to the
soluble form. BMC Res Notes 5: 483. doi: 10.1186/1756-0500-5-483
Kalenik B, Sawicka R, Góra-Sochacka A, Sirko A (2014) Inuenza pre-
vention and treatment by passive immunization. Acta Biochim Pol 61:
573–587
Kyte J, Doolittle RF (1982) A simple method for displaying the hydro-
pathic character of a protein. J Mol Biol 157: 105–132. 10.1016/0022-
2836(82)90515-0
Ladiges W, Osman GE (2001)Molecular characterization of immuno-
globulin genes. In Basic Methods in Antibody Production and Characteri-
zation. Howard GC & Bethell D eds, pp 169–192: CRC Press LLC
Li Y, Zhang X, Xu Q, Fu Q, Zhu Y, Chen S, Peng D, Liu X (2013)
Characterisation and haemagglutinin gene epitope mapping of a
variant strain of H5N1 subtype avian inuenza virus. Vet Microbiol
162: 614–622
Mayrose I, Penn O, Erez E, Rubinstein ND, Shlomi T, Freund NT,
Bublil EM, Ruppin E, Sharan R, Gershoni JM, Martz E, Pupko T
(2007a) Pepitope: epitope mapping from afnity-selected peptides.
Bioinformatics 23: 3244–3246. doi: 10.1093/bioinformatics/btm493
Mayrose I, Shlomi T, Rubinstein ND, Gershoni JM, Ruppin E, Sharan
R, Pupko T (2007b) Epitope mapping using combinatorial phage-
display libraries: a graph-based algorithm. Nucleic Acids Research 35:
69–78. doi: 10.1093/nar/gkl975
Meng EC, Pettersen EF, Couch GS, Huang CC, Ferrin TE (2006)
Tools for integrated sequence-structure analysis with UCSF Chime-
ra. Bmc Bioinformatics 7. doi: 10.1186/1471-2105-7-339
Oriuchi N, Higuchi T, Hanaoka H, Iida Y, Endo K (2005) Current
status of cancer therapy with radiolabeled monoclonal antibody.
Ann Nucl Med 19: 355–365
Peng Y, Zou Y, Li H, Li K, Jiang T (2014) Inferring the antigenic
epitopes for highly pathogenic avian inuenza H5N1 viruses. Vac-
cine 32: 671–676. doi: 10.1016/j.vaccine.2013.12.005
Rouet R, Lowe D, Dudgeon K, Roome B, Schoeld P, Langley D,
Andrews J, Whitfeld P, Jermutus L, Christ D (2012) Expression of
high-afnity human antibody fragments in bacteria. Nature Protocols
7: 364–373. doi: 10.1038/nprot.2011.448
Sali A, Blundell TL (1993) Comparative protein modeling by satisfac-
tion of spatial restraints. J Mol Biol 234: 779–815. doi: 10.1006/
jmbi.1993.1626
Shepelyakovskaya AO, Laman AG, Lomonosova AV, Fursova KK,
Savinov GV, Vertiev YV, Brovko FA, Grishin EV (2011) Effect of
the format of antibodies on their specicity. Mol Immunol 49: 433–
440. doi: 10.1016/j.molimm.2011.09.017
Smith GJD, Donis RO, Working WOFHNE (2012) Continued evo-
lution of highly pathogenic avian inuenza A (H5N1): updated
nomenclature. Inuenza and Other Respiratory Viruses 6: 1–5. doi:
10.1111/j.1750-2659.2011.00298.x
Smith GJD, Donis RO, World Health Organization W (2015) No-
menclature updates resulting from the evolution of avian inuenza
A(H5) virus clades 2.1.3.2a, 2.2.1, and 2.3.4 during 2013-2014. Inu-
enza and Other Respiratory Viruses 9: 271–276. doi: 10.1111/irv.12324
Sonoda H, Kumada Y, Katsuda T, Yamaji H (2010) Functional expres-
sion of single-chain Fv antibody in the cytoplasm of Escherichia
coli by thioredoxin fusion and co-expression of molecular chaper-
ones. Protein Expression and Purication 70: 248–253. doi: 10.1016/j.
pep.2009.11.003
Stachyra A, Gora-Sochacka A, Sawicka R, Florys K, Saczynska V,
Olszewska M, Pikula A, Smietanka K, Minta Z, Szewczyk B,
Zagorski W, Sirko A (2014) Highly immunogenic prime-boost
DNA vaccination protects chickens against challenge with homolo-
gous and heterologous H5N1 virus. Trials Vaccinol 3: 40–46
Stachyra A, Redkiewicz P, Kosson P, Protasiuk A, Góra-Sochacka A,
Kudla G, Sirko A (2016) Codon optimization of antigen coding se-
quences improves the immune potential of DNA vaccines against
avian inuenza virus H5N1 in mice and chickens. Virol J 13: 143.
doi: 10.1186/s12985-016-0599-y
Szewczyk B, Bienkowska-Szewczyk K, Krol E (2014) Introduction to
molecular biology of inuenza A viruses. Acta Biochim Pol 61: 397–
401
Tan Y, Ng Q, Jia Q, Kwang J, He F (2015) A novel humanized an-
tibody neutralizes H5N1 inuenza virus via two different mecha-
nisms. J Virol 89: 3712–3722. doi:10.1128/JVI.03014-14
Toleikis L, Frenzel A (2012) Cloning single-chain antibody fragments
(ScFv) from hyrbidoma cells. Methods Mol Biol 907: 59–71
Velkov T, Ong C, Baker MA, Kim H, Li J, Nation RL, Huang JX,
Cooper MA, Rockman S (2013) The antigenic architecture of the
hemagglutinin of inuenza H5N1 viruses. Mol Immunol 56: 705–719.
doi: 10.1016/j.molimm.2013.07.010
Wu R, Li X, Leung H-C, Cao Z, Qiu Z, Zhou Y, Zheng B-J, He
Y (2014) A novel neutralizing antibody against diverse clades of
H5N1 inuenza virus and its mutants capable of airborne transmis-
sion. Antiviral Res 106: 13–23. http://dx.doi.org/10.1016/j.antivi-
ral.2014.03.005
Zhang X, Qi X, Zhang Q, Zeng X, Shi Z, Jin Q, Zhan F, Xu Y, Liu
Z, Feng Z, Jiao Y (2013) Human 4F5 single-chain Fv antibody rec-
ognizing a conserved HA1 epitope has broad neutralizing potency
against H5N1 inuenza A viruses of different clades. Antiviral Res
99: 91–99. http://dx.doi.org/10.1016/j.antiviral.2013.05.001
Zuo T, Sun J, Wang G, Jiang L, Zuo Y, Li D, Shi X, Liu X, Fan S,
Ren H, Hu H, Sun L, Zhou B, Liang M, Zhou P, Wang X, Zhang
L (2015) Comprehensive analysis of antibody recognition in conva-
lescent humans from highly pathogenic avian inuenza H5N1 infec-
tion. Nat Commun 6: 8855–8867. doi: 10.1038/ncomms9855
... Based on the structural data, conserved residues involved in the interactions with glycan receptors in the hemagglutinin revealed the interface for binding sialic acid located at the membrane-distal tips of the RBD domain. The same epitopes are recognized specifically by IgG-type [27] or monoclonal mAb6-9-1 anti-bodies [28]. For the HA3 strain the conserved motif comprised α-helix (residues 190 EQTNLYVQA 199 ) together two loops (residues 130 VTQNGGSNACK 140 , and 220 RPWVGQSGRI 230 ) on the two sides of central helix (numbering according to the sequence of HA3) (Figure 1) [8]. ...
Article
Full-text available
Hemagglutinin (HA), the class I influenza A virus protein is responsible for the attachment of virus particles to the cell by binding to glycan receptors, subsequent virion internalization, and cell entry. Consequently, the importance of HA makes it a primary target for the development of anti-influenza drugs. The natural oligoribonucleotides (ORNs) as well as their derivatives functionalized with D-mannitol (ORNs-D-M) possess anti-influenza properties in vitro and in vivo due to interaction with HA receptor sites. This activity suppresses the viral infection in host cells. In the present work, the complexes of ORNs and ORNs-D-M with HA protein were studied by agglutination assay, fluorescence spectroscopy, as well as molecular docking simulations. Acquired experimental data exhibited a decrease in HA titer by 32 times after incubation with the ORNs-D-M for 0.5–24 h. Quenching fluorescence intensity of the HA suggests that titration by ORNs and ORNs-D-M probably leads to changes in the HA structure. Detailed structural data were obtained with the molecular docking simulations performed for ORNs and ORNs-D-M ligands containing three and six oligoribonucleotides. The results reveal that a majority of the ORNs and ORNs-D-M bind in a non-specific way to the receptor-binding domain of the HA protein. The ligand’s affinity to the hemagglutinin was estimated at the micromolar level. Presented experimental data confirmed that both natural ORNs and functionalized ORNs-D-M inhibit the interactions between HA and glycan receptors and demonstrate anti-influenza activity.
... Therefore, IAV infections are a globally important issue not only in humans but also in their livestock. Avian influenza (AI) viruses are classified by their pathogenic strength based on the feasibility of infection in chickens [5]. The critical genetic difference determining the low pathogenic (LPAI) or highly pathogenic (HPAI) phenotype depends on the HA cleavage site [6]. ...
Article
Full-text available
Influenza A viruses (IAVs) pose a serious global threat to humans and their livestock. This study aimed to determine the ideal irradiation by ultraviolet-light emitting diodes (UV-LEDs) for IAV disinfection. We irradiated the IAV H1N1 subtype with 4.8 mJ/cm2 UV using eight UV-LEDs [peak wavelengths (WL) = 365, 310, 300, 290, 280, 270, and 260 nm)] or a mercury low pressure (LP)-UV lamp (Peak WL = 254 nm). Inactivation was evaluated by the infection ratio of Madin–Darby canine kidney (MDCK) cells or chicken embryonated eggs. Irradiation by the 260 nm UV-LED showed the highest inactivation among all treatments. Because the irradiation-induced inactivation effects strongly correlated with damage to viral RNA, we calculated the correlation coefficient (RAE) between the irradiant spectrum and absorption of viral RNA. The RAE scores strongly correlated with the inactivation by the UV-LEDs and LP-UV lamp. To increase the RAE score, we combined three different peak WL UV-LEDs (hybrid UV-LED). The hybrid UV-LED (RAE = 86.3) significantly inactivated both H1N1 and H6N2 subtypes to a greater extent than 260 nm (RAE = 68.6) or 270 nm (RAE = 42.2) UV-LEDs. The RAE score is an important factor for increasing the virucidal effects of UV-LED irradiation.
Article
Full-text available
Background Highly pathogenic avian influenza viruses are a serious threat to domestic poultry and can be a source of new human pandemic and annual influenza strains. Vaccination is the main strategy of protection against influenza, thus new generation vaccines, including DNA vaccines, are needed. One promising approach for enhancing the immunogenicity of a DNA vaccine is to maximize its expression in the immunized host. Methods The immunogenicity of three variants of a DNA vaccine encoding hemagglutinin (HA) from the avian influenza virus A/swan/Poland/305-135V08/2006 (H5N1) was compared in two animal models, mice (BALB/c) and chickens (broilers and layers). One variant encoded the wild type HA while the other two encoded HA without proteolytic site between HA1 and HA2 subunits and differed in usage of synonymous codons. One of them was enriched for codons preferentially used in chicken genes, while in the other modified variant the third position of codons was occupied in almost 100 % by G or C nucleotides. ResultsThe variant of the DNA vaccine containing almost 100 % of the GC content in the third position of codons stimulated strongest immune response in two animal models, mice and chickens. These results indicate that such modification can improve not only gene expression but also immunogenicity of DNA vaccine. Conclusion Enhancement of the GC content in the third position of the codon might be a good strategy for development of a variant of a DNA vaccine against influenza that could be highly effective in distant hosts, such as birds and mammals, including humans.
Article
Full-text available
Understanding the mechanism of protective antibody recognition against highly pathogenic avian influenza A virus H5N1 in humans is critical for the development of effective therapies and vaccines. Here we report the crystal structure of three H5-specific human monoclonal antibodies bound to the globular head of hemagglutinin (HA) with distinct epitope specificities, neutralization potencies and breadth. A structural and functional analysis of these epitopes combined with those reported elsewhere identifies four major vulnerable sites on the globular head of H5N1 HA. Chimeric and vulnerable site-specific mutant pseudoviruses are generated to delineate broad neutralization specificities of convalescent sera from two individuals who recovered from the infection with H5N1 virus. Our results show that the four vulnerable sites on the globular head rather than the stem region are the major neutralizing targets, suggesting that during natural H5N1 infection neutralizing antibodies against the globular head work in concert to provide protective antibody-mediated immunity.
Article
Full-text available
The divergence of the A(H5) hemagglutinin (HA) gene of highly pathogenic avian influenza (HPAI) viruses (A/goose/Guangdong/96 lineage) was analyzed by phylogenetic and average pairwise distance methods to identify new clades that merit nomenclature changes. Three new clade designations were recommended based on division of clade 2.1.3.2a (Indonesia), 2.2.1 (Egypt) and 2.3.4 (widespread detection in Asia, Europe and North America) that includes newly emergent HPAI virus subtypes H5N2, H5N3, H5N5, H5N6 and H5N8. This article is protected by copyright. All rights reserved. This article is protected by copyright. All rights reserved.
Article
Full-text available
Unlabelled: Highly pathogenic avian influenza virus subtype H5N1 continues to be a severe threat to public health, as well as the poultry industry, because of its high lethality and antigenic drift rate. Neutralizing monoclonal antibodies (MAbs) can serve as a useful tool for preventing, treating, and detecting H5N1. In the present study, humanized H5 antibody 8A8 was developed from a murine H5 MAb. Both the humanized and mouse MAbs presented positive activity in hemagglutination inhibition (HI), virus neutralization, and immunofluorescence assays against a wide range of H5N1 strains. Interestingly, both human and murine 8A8 antibodies were able to detect H5 in Western blot assays under reducing conditions. Further, by sequencing of escape mutants, the conformational epitope of 8A8 was found to be located within the receptor binding domain (RBD) of H5. The linear epitope of 8A8 was identified by Western blotting of overlapping fragments and substitution mutant forms of HA1. Reverse genetic H5N1 strains with individual mutations in either the conformational or the linear epitope were generated and characterized in a series of assays, including HI, postattachment, and cell-cell fusion inhibition assays. The results indicate that for 8A8, virus neutralization mediated by RBD blocking relies on the conformational epitope while binding to the linear epitope contributes to the neutralization by inhibiting membrane fusion. Taken together, the results of this study show that a novel humanized H5 MAb binds to two types of epitopes on HA, leading to virus neutralization via two mechanisms. Importance: Recurrence of the highly pathogenic avian influenza virus subtype H5N1 in humans and poultry continues to be a serious public health concern. Preventive and therapeutic measures against influenza A viruses have received much interest in the context of global efforts to combat the current and future pandemics. Passive immune therapy is considered to be the most effective and economically prudent preventive strategy against influenza virus besides vaccination. It is important to develop a humanized neutralizing monoclonal antibody (MAb) against all of the clades of H5N1. For the first time, we report in this study that a novel humanized H5 MAb binds to two types of epitopes on HA, leading to virus neutralization via two mechanisms. These findings further deepen our understanding of influenza virus neutralization.
Article
Full-text available
Passive immunity is defined as a particular antigen resistance provided by external antibodies. It can be either naturally or artificially acquired. Natural passive immunization occurs during pregnancy and breast-feeding in mammals and during hatching in birds. Maternal antibodies are passed through the placenta and milk in mammals and through the egg yolk in birds. Artificial passive immunity is acquired by injection of either serum from immunized (or infected) individuals or antibody preparations. Many independent research groups worked on selection, verification and detailed characterization of polyclonal and monoclonal antibodies against the influenza virus. Numerous antibody preparations were tested in a variety of in vitro and in vivo experiments for their efficacy to neutralize the virus. Here, we describe types of antibodies tested in such experiments and their viral targets, review approaches resulting in identification of broadly neutralizing antibodies and discuss methods used to demonstrate their protective effects. Finally, we shortly discuss the phenomenon of maternal antibody transfer as a way of effective care for young individuals and as an interfering factor in early vaccination.
Article
Full-text available
This minireview presents an overview of current knowledge on virion structure, genome organization and basic events in the development of Influenza A virus. The processes of entry, transcription/replication and viral release are described. In this context, the roles of viral proteins (including recently discovered minor polypeptides) in the subsequent stages of viral development are also discussed.
Article
Full-text available
This paper concerns the development of an immunosensor for detection of peptides derived from avian influenza hemagglutinin H5. Its preparation consists of successive gold electrode modification steps: (i) modification with 1,6-hexanedithiol and gold colloidal nanoparticles; (ii) immobilization of antibody-binding fragments (Fab') of anti-hemagglutinin H5 monoclonal antibodies Mab 6-9-1 via S-Au covalent bonds; and (iii) covering the remaining free space on the electrode surfaces with bovine serum albumin. The interactions between Fab' fragments and hemagglutinin (HA) variants have been explored with electrochemical impedance spectroscopy (EIS) in the presence of [Fe(CN)6]3-/4- as an electroactive marker. The immunosensor was able to recognize three different His-tagged variants of recombinant hemagglutinin from H5N1 viruses: H1 subunit (17-340 residues) of A/swan/Poland/305-135V08/2006, the long HA (17-530 residues) A/Bar-headed Goose/Qinghai/12/2005 and H1 subunit (1-345 residues) of A/Vietnam/1194/2004. The strongest response has been observed for the long variant with detection limit of 2.2 pg/mL and dynamic range from 4.0 to 20.0 pg/mL.
Article
An electrochemical immunosensor for the detection of hemagglutinin from avian influenza virus H5N1 is presented in this paper. The following steps lead up to the construction of immunosensor: (i) modification of gold electrodes with 4,4′-thiobisbenzenethiol, (ii) modification of self-assembled monolayer of 4,4′-thiobisbenzenethiol with gold colloidal nanoparticles, (iii) immobilization of single chain variable fragments of antibodies (scFv) against hemagglutinin H5 via SAu covalent bonds, (iv) blocking of the remaining free space with bovine serum albumin. The interactions between the scFv and hemagglutinin variants have been explored with electrochemical impedance spectroscopy in the presence of [Fe(CN)6]3−/4− as an electroactive marker. The immunosensor was able to detect two different His-tagged variants of recombinant hemagglutinin from H5N1 viruses: the short fragment (17–340 residues) of A/swan/Poland/305-135V08/2006 and the long (17–530 residues) of A/Bar-headed Goose/Qinghai/12/2005. The strongest response has been observed for the long variant with a detection limit of 0.6 pg/mL and a dynamic range from 4.0 to 20.0 pg/mL. The recombinant hemagglutinin (17–527 residues) from A/chicken/Netherlands/1/03 (H7N7), used as the negative control generated a weak response. This confirms the selectivity of the immunsensor proposed. A miniaturized version of the immunosensor, based on screen-printed gold electrodes, was tested with the same set of recombinant hemagglutinins and it achieved a linear range from 1 to 8 pg/mL with a detection limit of 0.9 pg/mL for the long fragment of hemagglutinin.
Article
Recombinant immunotoxins, consisting of single-chain antibodies (scFv) genetically fused to polypeptide toxins represent potentially effective candidates for cancer therapeutics. Antibody affinity, internalization rates and intracellular fate of molecular constructs play critical roles in the eventual cytotoxic activities of these constructs although these events related to the design characteristics of fusion constructs have not been comprehensively explored. We evaluated the relationship between affinity of various anti-Her2 scFv fused to recombinant gelonin (rGel) and its effect on cytotoxic activity. A series of rGel-based immunotoxins were created from the human anti-Her2/neu scFv C6.5 and various affinity mutants (designated ML3.9, MH3B1 and B1D3-in increasing affinity order). The affinities of the scFv ranged from 10⁻⁸ to 10⁻¹¹ M and the rGel fusion toxins retained the affinities of the scFv from which they were derived. Against BT474M1 human breast tumor cells, the immunotoxins demonstrated IC50 values of 18.9, 10.9, 3.9 and 1.2nM for the C6.5, ML3.9, MH3B1 and B1D3 based immunotoxins respectively. Targeting indices were highest for the B1D3/rGel construct with the highest affinity. The internalization rate of the fusion constructs into target cells directly correlated with the cytotoxic effects. Competition studies showed that addition of soluble Her2/neu extracellular domain (ECD) as the competitive factor significantly reduced the cytotoxic effects of the highest-affinity fusion construct (B1D3/rGel) while the lower-affinity constructs showed lesser impact of Her2/neu ECD addition on the cytotoxic effect. Animal model efficacy studies against several Her2/neu overexpressing tumors are ongoing but appear to suggest that the construct containing the highest affinity antibody was comparatively less effective in vivo than constructs containing antibodies of intermediate affinity. These studies suggest that human MH3B1/rGel fusion construct with an apparent affinity of 10⁻¹⁰ M displayed an overall optimal cell-killing activity to several Her2/neu positive cancer cells especially in the presence of free Her2/neu ECD. These findings demonstrate the necessity to consider antibody affinity in design considerations for targeted therapeutics. In addition, the potential impact of soluble target or truncated receptor molecules in the environment need to be considered when evaluating the efficacy of targeted therapeutics. Research Conducted, in part, by the Clayton Foundation for Research. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2580.
Article
Highly pathogenic avian influenza A virus H5N1 continues to spread among poultry and has frequently broken the species barrier to humans. Recent studies have shown that a laboratory-mutated or reassortant H5N1 virus bearing hemagglutinin (HA) with as few as four or five mutations was capable of transmitting more efficiently via respiratory droplets between ferrets, posing a serious threat to public health and underscoring the priority of effective vaccines and therapeutics. In this study, we identified a novel monoclonal antibody (mAb) named HAb21, that has a broadly neutralizing activity against all tested strains of H5N1 covering clades 0, 1, 2.2, 2.3.4, and 2.3.2.1. Importantly, HAb21 efficiently neutralized diverse H5N1 variants with single or combination forms of mutations capable of airborne transmission. We demonstrated that HAb21 blocked viral entry during the receptor-binding step by targeting a previously uncharacterized epitope at the tip of the HA head. This novel epitope closely neighbors the receptor-binding site (RBS) and the interface of HA trimer and is highly conserved among divergent H5N1 strains. Our studies provide a new tool for use either for therapeutic purposes or as a basis of vaccine development.