Accepted Manuscript Title: Generation of monoclonal antibodies reactive against subtype specific conserved B-cell epitopes on haemagglutinin protein of Influenza virus H5N1 Author: Petra Fiebig Awad A. Shehata Uwe G. Liebert PII: DOI: Reference:

S0168-1702(15)00012-X http://dx.doi.org/doi:10.1016/j.virusres.2015.01.006 VIRUS 96508

To appear in:

Virus Research

Received date: Revised date: Accepted date:

22-9-2014 12-12-2014 10-1-2015

Please cite this article as: Fiebig, P., Shehata, A.A., Liebert, U.G.,Generation of monoclonal antibodies reactive against subtype specific conserved B-cell epitopes on haemagglutinin protein of Influenza virus H5N1, Virus Research (2015), http://dx.doi.org/10.1016/j.virusres.2015.01.006 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1

Generation of monoclonal antibodies reactive against subtype specific

2

conserved B-cell epitopes on haemagglutinin protein of Influenza virus H5N1

3

Petra Fiebiga,1,*, Awad A. Shehataa,b,2, Uwe G. Lieberta,*

5 6

a

7

Diseases Department, Faculty of Veterinary Medicine, Sadat City University, Egypt.

E-mail:

Petra

Fiebig:

[email protected],

Awad

A.

Shehata:

[email protected], Uwe Gerd Liebert: [email protected]

an

10

Author’s

us

8 9

Avian and Rabbit

cr

Institute of Virology, Leipzig University, Leipzig, Germany;

b

ip t

4

11

* Address correspondence to: Petra Fiebig, [email protected], +49 176 387 367

13

49, Windorfer Str. 18, 04229 Leipzig, Germany

14

Post-publication also to: Uwe G. Liebert, [email protected], +49 341

15

9714301, Institute of Virology, Leipzig University, Johannisallee 30, 04103 Leipzig,

16

Germany

17 18

1

20 21

Present address: Petra Fiebig, -

19 2

24 25

Omega Diagnostics GmbH, Reinbek, Germany

Present address: Awad A. Shehata, -

Avian and Rabbit Diseases Department, Faculty of Veterinary Medicine, Sadat City University, Egypt.

22 23

Ac ce pt e

d

M

12

-

Centre for Infectious Medicine, Faculty of Veterinary Medicine, Leipzig University, Germany 1

Page 1 of 43

Abstract

26

H5-specific monoclonal antibodies may serve as valuable tools for rapid diagnosis of

27

H5N1 avian influenza virus. Therefore, conserved H5-specific sequences of the

28

haemagglutinin (HA) protein were expressed in Pichia pastoris and used for

29

generation of monoclonal antibodies (mAbs). The two mAbs, FD6 and HE4, were

30

strongly reactive against native HA protein and exhibited specificity for subtype H5.

31

By epitope mapping linear epitopes of mAbs were identified that are highly conserved

32

among influenza A virus of subtype H5. Additionally no sequence similarities to

33

homologous regions on HA proteins of other influenza A virus subtypes (i.e. H1, H3,

34

H7, H9) were detected by sequence alignment analysis. Both mAbs did not cross

35

react with native or denatured HA proteins of other influenza A virus subtypes.

36

Furthermore, using ELISA and immunofluorescence test mAb FD6 reacted only to

37

the native H5 protein of recently circulating highly pathogenic H5N1 influenza viruses

38

but not to low pathogenic H5N1 isolates. In conclusion, the use of the two mAbs in

39

non-molecular tests like antigen-capture-ELISA appears promising for detecting

40

influenza A H5N1 virus.

42

cr

us

an

M

d

Ac ce pt e

41

ip t

25

43

Keywords: Avian influenza, H5N1, conserved epitopes, monoclonal antibodies,

44

Pichia pastoris, ELISA

45

2

Page 2 of 43

45

1. Introduction Zoonotic Influenza A viruses are endemic in birds and cause sporadic dead-end

47

infections in humans. From the known 18 haemagglutinin (H1-H18) and 11

48

neuraminidase (N1-N11) subtypes only viruses expressing H1, H2 or H3 and N1 or

49

N2 have caused pandemic human respiratory disease like the currently circulated

50

H1N1pdm09 (Fouchier et al., 2005, 2005; Sullivan et al., 2010; Tong et al., 2012;

51

Tong et al., 2013; Webster et al., 1992, 1992; Webster et al., 2013). However,

52

several subtypes of avian influenza A virus e.g. H5N1, H7N7, H9N2, H7N9 and

53

H6N1 have been shown to cross the species barrier and infect humans without prior

54

adaptation to humans in an alternate host (Claas et al., 1998; Gao et al., 2013;

55

Koopmans et al., 2004; Peiris et al., 1999; Wei et al., 2013). Particularly the H5N1

56

influenza virus causes high mortality not only in poultry but also in humans. By end of

57

June

58

(http://www.who.int/influenza/human_animal_interface/HAI_Risk_Assessment/en/,

59

last accessed August 3, 2014). Transmission from human to human occurs

60

sporadically, usually during care of patients (Ungchusak et al., 2005; Wang et al.,

61

2008; WHO, 2008). But as shown in a ferret model only few mutations of the H5N1

62

virus are necessary to acquire airborne transmission (Herfst et al., 2012; Imai et al.,

63

2012; Russell et al., 2012) and the segmented genome additionally favours

64

reassortment and the production of a virus that can cause a human pandemic. These

65

findings emphasize the urgent need for improved easy to perform and sensitive

66

diagnostic tests for emergency diagnosis to timely initiate anti-viral therapy. As RT-

67

PCR requires expertise and technology and antibody detection is not applicable at

68

early infection stages, rapid antigen detection is the method of choice. Results are

69

provided within short time and can be used at the point of care without sophisticated

393

of

667

infected

humans

have

died

Ac ce pt e

d

2014,

M

an

us

cr

ip t

46

3

Page 3 of 43

laboratory equipment by untrained personal. However, at present such tests exhibit a

71

low sensitivity, detect the highly conserved influenza nucleoprotein and thus do not

72

allow influenza virus subtyping (WHO, 2007). New diagnostic tests detecting the viral

73

HA-protein could overcome limitations of existing tests when H5 specific capture and

74

detector antibodies are used. For generating such antibodies, H5N1 virus or full-

75

length HA protein are currently used (Cui and Tong, 2008; He and Kwang, 2013; He

76

et al., 2007; Linke et al., 2011; Yang et al., 2009). The influenza HA mediates

77

receptor binding and penetration of the virus and is its major antigen. Five

78

neutralizing epitopes (site A, B, C, D and E) have been described for HA of subtype

79

H3 (Skehel et al., 1984; Wiley et al., 1981). Antigenic characterization of H5 molecule

80

using escape mutants of low and high pathogenic H5 strains revealed two antigenic

81

sites (Kaverin et al., 2007; Kaverin et al., 2002). Site 1, a loop comprising HA1

82

residues 136-141 (H5 numbering here and throughout the text), corresponds to

83

antigenic site A of H3 and Ca2 of H1 (Caton et al., 1982). Site 2 is composed of HA1

84

residues 151-156 and 182-194 corresponding to site B of H3 and HA1 residues 124-

85

129 that correspond to site Sa in the H1 subtype. As these sites represent

86

immunodominant regions, they are subject to selective pressure of the host immune

87

system. Therefore sequences of the HA protein that are conserved within subtype H5

88

and exhibit significant sequence differences to other HA subtypes (e.g. H1, H3, H7)

89

might be appropriate candidates to generate monoclonal antibodies (mAbs). Based

90

on this assumption, the objectives of this work were to identify such HA regions and

91

to generate and characterise mAbs that allow subtyping of influenza viruses with

92

specific detection of H5N1.

Ac ce pt e

d

M

an

us

cr

ip t

70

4

Page 4 of 43

93

2. Materials and Methods 2.1 Cell lines and viruses

95

Myeloma cells SP2/0-Ag14, cultured in RPMI 1640 Glutamax I (Gibco, Karlsruhe,

96

Germany) supplemented with 10% FCS, 1% (v/v) non-essential amino acids, 1 mM

97

sodium pyruvate, 2 mM L-glutamine, 100 U/ml penicillin and 100 µg/ml streptomycin

98

and Vero cells, cultured in DMEM Glutamax I (Gibco, Karlsruhe, Germany)

99

supplemented with 5% FCS (Biochrom AG, Berlin, Germany), were cultured at 37°C

us

cr

ip t

94

in a 5% CO2 humidified atmosphere.

101

Influenza A virus subtype H5N1 (A/Thailand/1(Kan-1)/2004) was kindly provided by

102

Dr. Puthavathana, Mahidol University, Bangkok (Puthavathana et al., 2005). The

103

virus was grown on Vero cells and supernatant was harvested 3-4 days post

104

infection. All other viruses were obtained as allantoic fluid: provided by Dr.

105

Schweiger, Robert Koch Institute, Berlin, Dr. Nieper, Landesuntersuchungsanstalt

106

Sachsen, Dr. Truyen, Leipzig University and Dr. Harder, Friedrich Löffler Institut, all

107

Germany, and from Dr. Sultan, Avian and Rabbit Diseases Department, Sadat City

108

University, Egypt. Table 1 lists influenza virus isolates used in this study.

109

To prepare whole virus antigen, clarified supernatant (340 × g, 10 min) was layered

110

over a 25% sucrose cushion in NTE buffer (0.1 M NaCl, 10 mM TrisCl pH 7.4, 1 mM

111

EDTA) and concentrated by ultracentrifugation (Surespin Rotor, 100,000 × g, 4°C;

112

Shaw et al., 2008). The pelleted virus was resuspended in NTE-buffer and tested by

113

haemagglutination test. For H5N1 all procedures were performed in BSL-3 facilities.

Ac ce pt e

d

M

an

100

5

Page 5 of 43

2.2 Identification of H5-specific, potential antigenic sites

115

Protein sequence of H5 A/Thailand/1(Kan-1)/2004 was analysed for potential B-cell

116

epitopes using seven algorithms based on physico-chemical properties (Emini et al.,

117

1985; Janin et al., 1978; Karplus and Schulz, 1985; Kolaskar and Tongaonkar, 1990;

118

Parker et al., 1986; Pellequer et al., 1993; Ponnuswamy et al., 1980) provided at

119

www.imtech.res.in/raghava/bcepred (Saha and Raghava). In general, epitopes had

120

to be predicted by at least 3 of the 7 used algorithms, except for exclusively predicted

121

epitopes by method of Kolaskar and Tongaonkar (1990). NCBI's Influenza Virus

122

Resource (http://www.ncbi.nlm.nih.gov/genomes/FLU/FLU.html, 16.10.2013) was

123

used to create HA-protein alignments of 736 H5N1 isolates from 1997-2005 and 206

124

H5 strains other than H5N1 from 1980-2006 (Bao et al., 2008). Consensus

125

sequences were used for determination of sequence similarities of potential B-cell

126

epitopes within subtype H5 using LALIGN Sequence Alignment of ExPASy

127

Proteomics Server (http://www.expasy.ch/, 16.05.2014; Gasteiger et al., 2003). The

128

same approach was used to determine similarities to human influenza viruses of

129

subtype H3N2 (978 isolates, 1990-2006) and H1N1 (252 Isolates, 1990-2006).

130

Finally four regions were chosen designated P1, P2, P3 and P4 for generation of

131

monoclonal antibodies. Also, chain HA1 (rHA1) and ectodomain of H5 (rH5) were

132

cloned and expressed in Pichia pastoris to be used as ELISA antigens.

133

2.3 Expression of recombinant peptides

134

The HA of influenza virus A/Thailand/1(Kan-1)/2004 virus was cloned in pcDNA3

135

plasmid (Invitrogen, Karlsruhe, Germany) and used for amplification of the truncated

136

HA-sequences (P1-P4, rHA1) by PCR. Primers used for PCR amplification are

137

shown in Table 2. Restriction sites of XhoI (sense primer) and NotI (antisense primer)

Ac ce pt e

d

M

an

us

cr

ip t

114

6

Page 6 of 43

were integrated into these primers for cloning into the expression vector pAOX-α mut,

139

as described previously (Shehata et al., 2012). Expression cassettes were

140

transformed into Pichia pastoris strains (GS115 and SMDH1186H, Invitrogen,

141

Karlsruhe, Germany) using an optimized protocol based on the Pichia Easy Comp™

142

Kit (Invitrogen, Karlsruhe, Germany). Positive transformants were selected on zeocin

143

containing media and used for expression of recombinant polypeptides fused with a

144

C-terminal histidin tag. Transformed yeast cells were grown in shake flasks

145

containing yeast peptone glycerol media (YPGly) (1% yeast extract, 2% peptone, 2%

146

glycerol). Expression of

147

methanol (YPM) pH8 (1% yeast extract, 1% peptone, 2% methanol) while expression

148

of P3 and rH5 was induced in buffered methanol-complex medium (BMMY) pH6 (1%

149

yeast extract, 1% peptone, 1,34% yeast nitrogen base, 2% methanol) supplemented

150

with Pepstatin A (2 µg/ml; Sigma Aldrich, Munich, Germany) when required.

151

Supernatants were collected 24 h (P1, P2, P4) or 48 h (P3, rHA1) after induction,

152

clarified by centrifugation (1780 × g, 10 min), supplemented with 1 mM

153

Phenylmethylsalfonylfluorid

154

Pepstatin A. Polypeptides were purified by metal affinity chromatography (IMAC, Ni-

155

NTA Agarose, Qiagen, Hilden, Germany) as described previously (Shehata et al.,

156

2012). Expression and purification was verified by western blot using (His)6-tag-

157

antibody (1:200, Dianova, Hamburg, Germany). The concentration of purified

158

polypeptides was measured using Biorad protein assay kit (Biorad, Hercules, CA,

159

USA).

us

cr

ip t

138

Ac ce pt e

d

M

an

P1, P2, P4 and rHA1 was induced in yeast peptone

(PMSF; Roth, Karlsruhe, Germany) and 2 µg/ml

7

Page 7 of 43

2.4 Generation of monoclonal antibodies

161

Balb/c mice were immunized subcutaneously using 50 µg (in 50 µl PBS) recombinant

162

polypeptide mixed with an equal volume of Gerbu Adjuvant 10 (Gerbu Biochemicals

163

GmbH, Schwabach, Germany) and boosted with identical doses 2 and 3 weeks

164

thereafter. Additional three boosts of 25 µg polypeptide in PBS were given on day 3,

165

2 and 1 before mice were sacrificed. Spleen cells were harvested and fused with

166

SP2/0-Ag14 cells at 2:1 ratio using polyethylene glycol (PEG) 1500 (Roche

167

Diagnostics GmbH, Mannheim, Germany). Hybridoma cells were seeded into 96-well

168

plates and selected in hypoxanthine-aminopterin-thymidine (HAT) supplemented

169

RPMI 1640 Glutamax I medium (Sigma-Aldrich Chemie GmbH, Munich, Germany).

170

After cultivation for 3 weeks aminopterin was omitted from medium and supernatants

171

were screened for antibody reactivity and specificity by ELISA. Cells of positive

172

tested wells were subcloned by limiting dilution method and screened again. MAbs

173

were purified from supernatant using T-Gel Absorbent® (Pierce, Schwerte,

174

Germany). Isotyping was performed using a mouse Immunglobulin Isotyping ELISA

175

Kit (BD Pharmingen™, Heidelberg, Germany) according to the manufacturer.

176

Determination of antibody concentration was performed with Bradford assay.

177

2.5 Indirect ELISA

178

For indirect ELISA, homologous recombinant polypeptides (1 µg/ml) used in mouse

179

vaccination or whole virus antigens (20 HAU/ml) diluted in carbonate buffer (pH 9.6)

180

were coated on Microlon high binding microtiter plates (Greiner Bio-One GmbH,

181

Frickenhausen, Germany) and incubated overnight at 4 °C. ELISA was performed as

182

described previously (Shehata et al., 2012). Briefly, plates were blocked with

183

3% BSA in PBS (PBSA) containing 0.05% Tween 20 and hybridoma supernatant or

Ac ce pt e

d

M

an

us

cr

ip t

160

8

Page 8 of 43

purified mAb diluted in blocking buffer was added (2 h, 37°C). After addition of

185

horseradish peroxidase (HRP)-conjugated rabbit anti-mouse immunoglobulin (diluted

186

1:2000, Dako Cytomation GmbH, Hamburg, Germany) for 90 min, 37 °C and

187

3,3′,5,5′tetramethylbenzidin (TMB) substrate, colour development was stopped after

188

30 min with 1 N sulphuric acid and absorbance was measured at 450/650 nm. Plates

189

were washed 5 times with PBS containing 0.05% Tween 20 after each incubation

190

step. Antibody titers of mice sera were expressed as endpoint titer and the cutoff was

191

determined using sera of mice immunized with PBS plus adjuvant (Frey et al., 1998).

192

2.6 Western blot and Epitope Mapping

193

For Western blot analysis, recombinant peptides (1 µg/lane) and whole virus antigens

194

(4 HAU/lane) of different HA subtypes were separated by SDS-PAGE and transferred

195

onto PVDF membrane (Roti-PVDF, Roth, Nuremberg, Germany). For Epitope

196

Mapping fifteen amino acids long peptides overlapping by 13 amino acid residues

197

covering the sequences of P1, P2, P4, and rHA1, respectively, were spotted onto a

198

cellulose membrane (IZKF-Leipzig, Leipzig, Germany). Membranes were blocked

199

with 4% skim milk in PBS and incubated with hybridoma supernatant overnight at

200

4 °C, followed by incubation with HRP-conjugated rabbit anti-mouse immunoglobulin

201

(Dako, 1:2000) for 90 min at room temperature. Detection was done using

202

chemiluminescence. Membranes were washed 3 times for 10 min with PBS after

203

each incubation step.

204

2.6 Immunofluorescence assay

205

Vero cells (grown on glass cover slips) were infected with Influenza virus at

206

multiplicity of infection (m.o.i.) of 0.01. After 1 h incubation the supernatant was

207

replaced by DMEM supplemented with 5% FCS for viruses exhibiting a multibasic 9

Ac ce pt e

d

M

an

us

cr

ip t

184

Page 9 of 43

HA-cleavage site or 2 µg/ml L-1-tosylamide-2-phenylethyl chloromethyl ketone

209

(TPCK)-treated trypsin (Sigma-Aldrich Chemie GmbH, Munich, Germany) and 0.2%

210

BSA for viruses with a mono basic HA-cleavage site. After 24 h cells were fixed with

211

80% ice cold acetone in water at -20°C for 30 min. Cells were blocked with 5% PBSA

212

for 30 min at 37°C in a humidified chamber. Hybridoma supernatant was added and

213

incubated for 90 min, 37°C. FITC conjugated goat anti-mouse immunoglobulin (Dako,

214

1:100) was added to cells and incubated for 45 min. Cover slips were washed

215

between each step with 0.5% PBSA. After mounting cover slips onto a glass slide

216

using mounting fluid (10% glycerine in PBS) cells were screened for fluorescent

217

staining.

218

2.7 Homology analysis and cross-reaction examination

219

Identified epitopes were analysed for similarities within H1-, H3-, H7- , H9- and H5-

220

subtypes using „Identify Short Peptides in Proteins“ provided by Influenza Research

221

Database (IRD, http://www.fludb.org, 16.05.2014, Squires et al., 2007). Isolates from

222

1990-2013 with complete HA protein sequences were used for sequence analysis

223

exclusively. Doubling sequences of one isolate were excluded as well as viruses

224

obtained by genetic manipulations.

225

2.8 Neutralization assay

226

The neutralizing activity of mouse immune sera as well as mAb were tested by

227

neutralization test according to the WHO manual (WHO, 2002). Briefly, 50 µl mouse

228

serum or mAb (25 µg/50 µl) were mixed with an equal volume of virus H5N1

229

containing 100 TCID50/50 µl and incubated at 37°C. After 1 h the mixture (100 µl/well)

230

was transferred to a flat-bottomed microtiter plate with a Vero cell monolayer (2×106

231

cells/plate). After 1 h incubation at 37°C, supernatant was removed and 200 μl of the

Ac ce pt e

d

M

an

us

cr

ip t

208

10

Page 10 of 43

DMEM containing 5% FCS was added and incubated at 37°C in a 5% CO2 humidified

233

incubator. After 3 days of incubation, plates were read as positive or negative

234

according to presence or absence of cytopathic effect.

235

2.9 Haemagglutination assay and haemagglutination inhibition assay

236

Haemagglutination assay for virus titration and haemagglutination inhibition (HI) for

237

examination of mouse sera and mAbs were performed according to the OIE manual

238

of diagnostic tests and vaccines for terrestrial animals (World Organisation for Animal

239

Health (OIE), 2004).

240

2.10 Antigen capture ELISA for detection of influenza A virus subtype H5 in

241

clinical samples

242

Working concentrations of mAbs were determined by checkerboard titration and

243

comparison of highest signal-to-noise ratio for the detection of H5 and non-H5

244

subtypes. Different concentrations (1 and 2 µg/ml) of mAb FD6 (IgM) or HE4 (IgG)

245

were coated as capture antibody in 50 µl carbonate buffer on Microlon high binding

246

microtiter plates (Greiner Bio-One) and incubated overnight at 4 °C. After removal of

247

unbound antibody, plates were blocked with 3% PBSA 0.05% Tween 20 for 1 h.

248

Afterwards 5 HAU of Influenza virus isolates were added at volumes of 75 µl/well and

249

incubated for 2 hrs. Each sample was tested in duplicate. Detection of bound antigen

250

was carried out by addition of 100 µl/well non-conjugated detection antibody HE4 or

251

FD6, respectively for 90 min, followed by addition of HRP-conjugated heavy chain

252

specific antibody (Dianova, goat anti-mouse IgG1 or goat anti-mouse IgM,

253

respectively, both diluted 1:5000) for 90 min.

254

Thirty tracheal swabs obtained from commercial layer chickens in Egypt were

255

assessed. Chickens were suspicious for infection with avian influenza. The clinical 11

Ac ce pt e

d

M

an

us

cr

ip t

232

Page 11 of 43

symptoms consisted of severe respiratory manifestations, petechial haemorrhages

257

on the shank and severe congestion of internal organs. Six influenza negative

258

tracheal swabs were obtained from healthy chickens to be used as negative control.

259

For detection of H5N1 in clinical samples (tracheal swabs from animals with clinical

260

disease and from healthy controls), mAb FD6 (IgM) was used as capture antibody

261

(2 µg/ml) and mAb HE4 (2 µg/ml) was used as detector antibody. The cut-off value

262

was defined as mean OD450 of negative controls + 3*SD.

263

2.11 RT-PCR

264

H5N1 infection of chickens was verified by RT-PCR, for which RNA was extracted

265

from specimens using Viral Gene-spin™Viral DNA/RNA Extraction kit (Intron

266

Biotechnology, Freiburg, Germany) according to manufacturer’s protocol. Reverse

267

transcription of H5 and N1 gene was performed using AMV Reverse Transcriptase kit

268

(Promega, Mannheim, Germany) with CU-H5F and CU-N1F primers (Barbazan et al.,

269

2008), respectively. To amplify a 189 bp fragment of H5 and 131 bp fragment of N1

270

primer pair of CU-H5F/CU-H5R or CU-N1F/CU-N1R (Barbazan et al., 2008) were

271

used, respectively. The PCR-program was as follow: 95°C 2 min, 35 × (95°C, 30

272

sec; 55°C, 45 sec; 72°C, 1min) 72°C 10 min. PCR products were analysed by 1%

273

(w/v) agarose gel electrophoresis.

Ac ce pt e

d

M

an

us

cr

ip t

256

12

Page 12 of 43

274

3. Results 3.1 Identification of potential H5-specific antigenic sites

276

Out of 20 predicted potential epitopes four regions, designated P1, P2, P3 and P4

277

(Table 3 and Figure 1) were eventually chosen that comprises epitopes with

278

conserved sequences within subtype H5 but sequence differences to other subtypes.

279

Additionally, these epitopes have not been described for Influenza H5 in literature.

280

Polypeptide P1 comprises aa 43-82 and corresponds in its location to the neutralising

281

epitope site E of H3 subtype (Skehel et al., 1984; Wiley et al., 1981). Glycosylated

282

polypeptide P2 (aa 165-261) encompasses the receptor binding site and parts of

283

identified site B epitope of H3, also described for H5 (site 2, Kaverin et al., 2002). P3

284

comprises the C-terminus of chain HA1 (aa 292-330), which is unique for H5 protein

285

of highly pathogenic H5N1 viruses, due to multiple basic amino acid residues at the

286

cleavage site. Polypeptide P4 is located within HA2 moiety (aa 38-132), which is

287

highly conserved among different influenza A virus subtypes.

288

3.2 Production of mAbs

289

Secretory expression of polypeptides P1-P4 in Pichia pastoris was verified by

290

Western blot using (His)6-tag-antibody (Figure 2). Purified peptides were used for

291

immunization of Balb/c mice. Polypeptides P1, P2 and P4 but not P3 induced

292

seroconversion. Serum antibodies against P1, P2 and P4 reacted with homologous

293

antigens (polypeptide used for immunisation) and with whole H5N1 virus antigen in

294

ELISA, although with 50-100-fold lower endpoint titers. No cross reactivity was

295

observed with viruses of subtype H3 and H1 using ELISA (data not shown). Sera

296

obtained from P1, P2, and P4 vaccinated mice reacted only with H5 viruses based on

Ac ce pt e

d

M

an

us

cr

ip t

275

13

Page 13 of 43

IFA performed on Vero cells infected with different influenza virus subtypes (Figure

298

3). No neutralisation or inhibition of haemagglutination was observed for any of the

299

sera (data not shown).

300

After somatic cell hybridisation of mouse spleen cells with myeloma cells several

301

hybridoma cell lines were selected by HAT supplemented medium. Screening by

302

ELISA on homologous antigens and whole virus H5N1 revealed two monoclonal cell

303

lines, designated HE4 and FD6 that produced mAbs with reactivity and specificity to

304

H5N1 virus.

305

3.3 Characterisation of mAb FD6 and HE4

306

In Western blot using recombinant polypeptides or whole virus as antigens, mAb FD6

307

(isotype IgM) reacted specifically with polypeptides comprising region P1 and viruses

308

of subtype H5 (Figure 4A). Similar reactions were detected when mAb FD6 was used

309

as primary antibody for ELISA, although it failed to react with H5N6-virus coated with

310

a concentration of 1 HAU/well (Figure 4C). The mAb HE4 (isotype IgG1) reacted

311

specifically with recombinant peptides comprising region P2 (i.e. P2, rHA1) and

312

viruses of subtype H5 using Western blot (Figure 4B) and ELISA (Figure 4C). Both

313

mAbs did not react with influenza viruses of other subtypes i.e. H3 and H1. IFA was

314

performed on Vero cells infected with H5N1 and influenza virus of other subtypes to

315

determine the ability of mAbs to recognise conformational haemagglutinin. MAb HE4

316

and mAb FD6 showed a specific staining of H5N1 virus infected cells with low

317

background (Figure 4D). As observed with ELISA, mAb FD6 did not recognise H5N6

318

infected cells, although it reacted with denatured H5N6 virus in Western blot.

319

Both mAbs failed to neutralise or inhibit H5N1 virus (data not shown).

Ac ce pt e

d

M

an

us

cr

ip t

297

14

Page 14 of 43

Immunoblotting using the membrane with spotted peptides covering P1-sequence

321

indicated that the sequence VKPLILRDCSV is the linear epitope of mAb FD6. Using

322

the membrane with spotted peptides covering HA1-sequence, minimal amino acid

323

sequence required for mAb FD6 binding was determined as VKPLI (data not shown).

324

For mAb HE4, the sequence QEDLLVLWGIH was identified as linear epitope using

325

the membrane with spotted peptides covering sequence of P2 and rHA1. Amino acid

326

residues at the N and C terminus, i.e. QE as well as IH, are the essential

327

components of the epitope binding site. No antibody binding was observed with

328

peptides missing residues QE or IH.

329

3.4 Homology and cross-reactivity analysis

330

Using „Identify Short Peptides in Proteins“, isolates of H1, H3, H7, H9 and H5

331

subtypes were analysed for similarities with determined epitope sequence. Of the

332

1537 analysed H5N1 isolates 94% exhibited the epitope sequence of mAb FD6,

333

while 49% of 461 analysed H5 isolates (other than H5N1) comprised the sequence.

334

But it was not found within the H1, H3, H7 or H9 sequence (Table 4). Analysis of 11

335

amino acid long epitope of HE4 revealed no exact matches in H1 or H3 protein

336

sequence, but 29.6% of H3Nx isolates and 90% of H1Nx isolates held a sequence

337

similarity higher than 50%. But all the detected similarities were located between the

338

identified anchor amino acids QE and IH (Figure 5). Looking for identities in anchor

339

amino acids only, no consensus was found in H3 and H1 isolates. Of the 574

340

analysed H7Nx isolates only 1.9% and 17.7% of the 937 H9Nx isolates exhibit a

341

sequence similarity of 55% to the epitope of mAb HE4. But similarities are mainly

342

found in the C-terminal epitope region, including anchor amino acids IH. But as N-

343

terminal anchor amino acids QE are not present a binding of the mAb to H7 or H9 is

Ac ce pt e

d

M

an

us

cr

ip t

320

15

Page 15 of 43

not expected. However, 92% of the 1537 analysed H5N1 isolates and 39.9% of 461

345

H5 isolates other than H5N1 contain the anchor amino acids (Table 4).

346

3.5 Antigen Capture ELISA (AC-ELISA)

347

The optimal antibody concentrations was determined as 100 ng/well for FD6 (capture

348

antibody) and 100 ng/well mAb HE4 (detector antibody) in conjunction with HRP-

349

conjugated Fcɣ1-specific antibody. Signal-to-noise ratios indicated that H5N1

350

samples are distinguishable from samples containing H3N8, H1N1 and H5N6 (5 HAU

351

each). Interestingly, the AC-ELISA did not work when mAb HE4 was used as capture

352

and FD6 as detector in conjunction with HRP-conjugated µ-chain specific antibody.

353

This might indicate a degradation of the IgM antibody within its Fc-part.

354

Optimized conditions using FD6 as capture antibody were used to analyse analytical

355

specificity of the test. It turned out that HPAIV H5N1 from Thailand and Egypt are

356

detected specifically while other HA-subtypes like H1 and H3 were not detected

357

(Figure 6A) even at high concentrations of 1500 HAU (data not shown). Additionally,

358

other H5-subtypes like H5N3, H5N2, low pathogenic H5N1 and outgrouped high

359

pathogenic H5N1 from 1959 are not detected, even at high concentrations. Subtype

360

H5N6 on the other hand is detected in a concentration dependent manner (Figure

361

6B). Compared to H5N1 50 HAU of H5N6 are needed to obtain values above OD450

362

0.2. These concentrations are usually not obtained in specimens, thus AC-ELISA is

363

specific for H5N1, which can be detected even at low concentrations of 2 HAU.

364

To verify applicability of AC-ELISA, thirty specimens derived from chickens showing

365

clinical manifestations of H5N1 infection were tested by AC-ELISA and RT-PCR in

366

parallel. By AC-ELISA 13 out of 30 specimens were tested positive, whereas 16

Ac ce pt e

d

M

an

us

cr

ip t

344

16

Page 16 of 43

samples were tested RT-PCR positive. In comparison to RT-PCR the AC-ELISA

368

exhibits 81% relative sensitivity and 100% relative specificity (Table 5).

Ac ce pt e

d

M

an

us

cr

ip t

367

17

Page 17 of 43

369

4. Discussion In the present work two mAbs were generated that are specific for H5 Influenza virus

371

antigen and allow its specific detection in an antigen-capture ELISA. As the

372

emergence of new pandemic influenza viruses of different HA subtypes is a

373

continuous threat, easy-to-handle rapid antigen detection tests for point of care

374

testing are required that allow viral subtyping. Therefore, HA subtype specific

375

monoclonal antibodies are advantageous. The HA, a type I membrane protein,

376

comprises the major antigen of influenza A virus but its immunodominant epitopes

377

underlie continuous mutations resulting in viral escape from host immune detection

378

(Laursen and Wilson, 2013). Due to this immunodominant regions should not be

379

targets of diagnostic antibodies, as they might loose affinity. Accordingly, in this study

380

two mAbs were generated which target immune-subdominant epitopes. These are

381

not overlapping with known neutralising epitopes and were identified by analysis of

382

the H5 protein with algorithms for prediction of linear B-cell epitopes. Sequence

383

conservation within subtype H5 and differences to other subtypes were also

384

important epitope characteristics. The pre-identificaiton of potential H5-specific

385

epitopes allows the recombinant expression of defined HA protein regions and the

386

directed production of antibodies towards these epitopes, which reduces extensive

387

selection procedures in the aftermath necessary when whole virus is used as

388

immunisation antigen (Linke et al., 2011). MAb FD6 (IgM) and mAb HE4 (IgG1) were

389

obtained by immunisation of mice with recombinant peptides P1 and P2, followed by

390

somatic cell hybridisation and selection, respectively. Both antibodies bind to

391

denatured whole virus of subtype H5 as verified by Western Blot but not to other

392

influenza virus subtypes. While mAb HE4 binds also to native forms of the H5 protein

393

of all H5 isolates, the specificity of FD6 is restricted to recent high pathogenic H5N1

Ac ce pt e

d

M

an

us

cr

ip t

370

18

Page 18 of 43

strains. It reacted not with low pathogenic H5N1, the outgrouped H5N1 or other H5

395

subtypes. The difference in recognition by mAb FD6 might be explained by the

396

recognised epitope and the HA protein sequences. By epitope mapping the linear

397

epitope (DLDG)VKPLILRDCSVAGW (residues 43-60 in H5 protein) with a sequence

398

minimum of VKPLI was identified for mAb FD6. Sequence analysis of HA protein

399

from H5N1 and H5N6 revealed amino acid substitutions D43S and D45N in H5N6,

400

which might influence conformation of the protein and therefore epitope accessibility.

401

Thus mAb FD6 seems to distinguish between H5N1 and other H5 subtypes.

402

Sequence analysis indicates that 98.3% of H5Nx isolates (x = subtype 2-10, 461

403

isolates) misses DLDGVKPLIL, with mutations predominantly within D43, L44 and

404

D45. On the other hand, the vast majority of analysed H5N1 isolates (n=1537) carry

405

the identical sequence with maximum one mutation at residue 43 or 45 (91.5%).

406

Experiments using such strains like A/Hong Kong/485/97 are necessary to confirm

407

antibody binding to native HA in spite of one mutated residue.

408

Epitope of mAb HE4 has been identified as the N-terminal residues 5-15

409

QEDLLVLWGIH of peptide P2, which corresponds to residues 165-179 in H5 protein

410

and thus lies between the two sites comprising the discontinuous epitope site 2

411

(Kaverin et al., 2002). Epitope analysis of mAb HE4 revealed terminal residues QE

412

and IH as important anchor sites for antibody binding, whereas amino acid residues

413

in between are not important. This finding was supported by ELISA, Western Blot and

414

IFA where H1N1 and H3N2 strains were not detected by mAb HE4 although

415

homologous epitope region of H3 protein KFDKLYIWGVH has a similarity of 55% to

416

epitope of mAb HE4. Similar results were found for homologous region of H1 protein

417

(EKEVLVLWGVH). As only a minority of the avian influenza H7 and H9 proteins

418

comprise the C-terminal anchor amino acids, but all isolates miss the N-terminal

Ac ce pt e

d

M

an

us

cr

ip t

394

19

Page 19 of 43

anchor amino acids a binding of mAb HE4 is not expected. As IgM antibody FD6 has

420

a lower avidity to its antigen (data not shown) but in a competitive ELISA it is not

421

competed by HE4, which suggests that binding of one antibody is not blocked by the

422

other (data not shown) and they can be used as capture and detector antibody in an

423

AC-ELISA. Additionally, due to the selective epitope binding of mAB FD6 to

424

conformational haemagglutinin the AC-ELISA allows the discrimination of recent

425

HPAIV H5N1 to low pathogenic H5N1 and other H5Nx subtypes. With a reproducible

426

detection limit of 2 HAU this mAb combination is not more sensitive than other

427

available test (general sensitivity of >1 HAU). In contrast Linke and co-workers

428

generated a test with a detection limit of 0.04-0.05 HAU. Besides the combination of

429

two mAbs for detection and or capture, the use of Poly-HRP40 conjugate improved

430

the detection limit significantly (Linke et al., 2011). The described two mAbs were

431

used singular as detector or capture antibody and they were not HRP conjugated.

432

Instead bound capture antibody was verified by heavy chain specific HRP-conjugated

433

antibody. This resulted in relatively high background, which did not allow detection of

434

lower antigen amounts i.e. 1 HAU. By conjugation the high background might be

435

overcome and detection limit improved.

436

To verify practicability of the AC-ELISA with field samples, 30 tracheal swabs from

437

commercial layer chickens were comparatively analysed by AC-ELISA and RT-PCR.

438

The AC-ELISA had a specificity of 100% but a somewhat reduced sensitivity,

439

particularly with samples of low viral load. Conjugation of detector antibody HE4 to

440

HRP or biotin might improve the performance.

441

Although no monoclonal antibodies were generated, immunisation of Balb/c mice

442

with P4 resulted in H5-subtype specific serum antibodies, which did not cross-react

443

with HA subtypes H3 and H1. This renders the approach promising for generation of

Ac ce pt e

d

M

an

us

cr

ip t

419

20

Page 20 of 43

subtype specific antibodies binding within HA2 moiety, which contributes less to the

445

antigenicity of HA (Vareckova et al., 2003) and therefore should acquire fewer

446

mutations that interfere antibody binding.

447

Somewhat unexpected was the low immunogenicity in inbred Balb/c mice of P3 as

448

the conformation of peptides derived from N- or C-termini of proteins exhibit more

449

likely a conformation similar to this within the native protein and therefore should

450

induce suitable antibodies (Hanly et al., 1995).

451

In conclusion, this paper describes the generation of H5-specific monoclonal

452

antibodies with high specificity and their use in an AC-ELISA. Their use in

453

combination with other described H5 specific antibodies or with commercial available

454

test for nucleoprotein detection may help to create a highly H5-specific and sensitive

455

test system for influenza virus diagnostic without sophisticated technologies.

Ac ce pt e

d

M

an

us

cr

ip t

444

21

Page 21 of 43

456 457

5. References Bao, Y., Bolotov, P., Dernovoy, D., Kiryutin, B., Zaslavsky, L., Tatusova, T., Ostell, J., Lipman D., 2008. The Influenza Virus Resource at the National Center for

459

Biotechnology Information. J Virol 82 (2), 596–601.

460

ip t

458

Barbazan, P., Thitithanyanont, A., Missé, D., Dubot, A., Bosc, P., Luangsri, N.,

Gonzalez, J.P., Kittayapong, P., 2008. Detection of H5N1 avian influenza virus

462

from mosquitoes collected in an infected poultry farm in Thailand. Vector Borne

463

Zoonotic Dis. 8 (1), 105–109.

us

Caton, A.J., Brownlee, G.G., Yewdell, J.W., Gerhard, W., 1982. The antigenic

an

464

cr

461

structure of the influenza virus A/PR/8/34 hemagglutinin (H1 subtype). Cell 31 (2

466

Pt 1), 417–427.

467

M

465

Claas, E.C., Osterhaus, A.D., van Beek, R., De Jong JC, Rimmelzwaan, G.F., Senne, D.A., Krauss, S., Shortridge, K.F., Webster, R.G., 1998. Human influenza

469

A H5N1 virus related to a highly pathogenic avian influenza virus. Lancet 351

470

(9101), 472–477.

Ac ce pt e

471

d

468

Cui, S., Tong, G., 2008. A chromatographic strip test for rapid detection of one

472

lineage of the H5 subtype of highly pathogenic avian influenza. J Vet Diagn Invest

473

20 (5), 567–571.

474

Emini, E.A., Hughes, J.V., Perlow, D.S., Boger, J., 1985. Induction of hepatitis A

475

virus-neutralizing antibody by a virus-specific synthetic peptide. J Virol. 55, 836–

476

839.

477 478

Fouchier, R.A., Munster, V., Wallensten, A., Bestebroer, T.M., Herfst, S., Smith, D., Rimmelzwaan, G.F., Olsen, B., Osterhaus, A.D., 2005. Characterization of a novel

22

Page 22 of 43

479

influenza A virus hemagglutinin subtype (H16) obtained from black-headed gulls.

480

J Virol 79 (5), 2814–2822.

481

Frey, A., Di Canzio, J., Zurakowski, D., 1998. A statistically defined endpoint titer determination method for immunoassays. J Immunol Methods 221 (1-2), 35–41.

483

Gao, R., Cao, B., Hu, Y., Feng, Z., Wang, D., Hu, W., Chen, J., Jie, Z., Qiu, H., Xu,

ip t

482

K., Xu, X., Lu, H., Zhu, W., Gao, Z., Xiang, N., Shen, Y., He, Z., Gu, Y., Zhang, Z.,

485

Yang, Y., Zhao, X., Zhou, L., Li, X., Zou, S., Zhang, Y., Li, X., Yang, L., Guo, J.,

486

Dong, J., Li, Q., Dong, L., Zhu, Y., Bai, T., Wang, S., Hao, P., Yang, W., Zhang,

487

Y., Han, J., Yu, H., Li, D., Gao, G.F., Wu, G., Wang, Y., Yuan, Z., Shu, Y., 2013.

488

Human Infection with a Novel Avian-Origin Influenza A (H7N9) Virus. N Engl J

489

Med 368 (20), 1888–1897.

an

us

cr

484

Gasteiger, E., Gattiker, A., Hoogland, C., Ivanyi, I., Appel, R., Bairoch, A., 2003.

491

ExPASy: the proteomics server for in-depth protein knowledge and analysis.

492

Nucleic Acids Res 31, 3784–3788.

d

M

490

Hanly, W.C., Artwohl, J.E., Bennett, B.T., 1995. Review of polyclonal antibody

494

production procedures in mammals and poultry. ILAR J 37 (3), 93–118.

495

Ac ce pt e

493

He, F., Kwang, J., 2013. Monoclonal Antibody Targeting Neutralizing Epitope on

496

H5N1 Influenza Virus of Clade 1 and 0 for Specific H5 Quantification. Influenza

497

Research and Treatment 2013 (4), 1–6.

498

He, Q., Velumani, S., Du, Q., Lim, C.W., Ng, F.K., Donis, R., Kwang, J., 2007.

499

Detection of H5 avian influenza viruses by antigen-capture enzyme-linked

500

immunosorbent assay using H5-specific monoclonal antibody. Clin Vaccine

501

Immunol 14 (5), 617–623.

502

Herfst, S., Schrauwen, E., Linster, M., Chutinimitkul, S., Wit, E. de, Munster, V.J.,

503

Sorrell, E.M., Bestebroer, T.M., Burke, D.F., Smith, D.J., Rimmelzwaan, G.F., 23

Page 23 of 43

504

Osterhaus, A., Fouchier, R.A.M., 2012. Airborne transmission of influenzaA/H5N1

505

virus between ferrets. Science 336 (6088), 1534–1541.

506

Imai, M., Watanabe, T., Hatta, M., Das, S., Ozawa, M., Shinya, K., Zhong, G., Hanson, A., Katsura, H., Watanabe, S., Li, C., Kawakami, E., Yamada, S., Kiso,

508

M., Suzuki, Y., Maher, E., Neumann, G., Kawaoka, Y., 2012. Experimental

509

adaptation of an influenza H5 HA confers respiratory droplet transmission to a

510

reassortant H5 HA/H1N1 virus in ferrets. Nature 486, 420–428.

cr

Janin, J., Wodak, S., Levitt, M., Maigret, B., 1978. Conformation of amino acid side-

us

511

ip t

507

chains in proteins. Journal of Molecular Biology 125 (3), 357–386.

513

Karplus, P., Schulz, G., 1985. Prediction of Chain Flexibility in Proteins.

515

Naturwissenschaften 72, 212–213.

Kaverin, N.V., Rudneva, I.A., Govorkova, E.A., Timofeeva, T.A., Shilov, A.A.,

M

514

an

512

Kochergin-Nikitsky, K.S., Krylov, P.S., Webster, R.G., 2007. Epitope mapping of

517

the hemagglutinin molecule of a highly pathogenic H5N1 influenza virus by using

518

monoclonal antibodies. J Virol 81 (23), 12911–12917.

Ac ce pt e

519

d

516

Kaverin, N.V., Rudneva, I.A., Ilyushina, N.A., Varich, N.L., Lipatov, A.S., Smirnov,

520

Y.A., Govorkova, E.A., Gitelman, A.K., Lvov, D.K., Webster, R.G., 2002. Structure

521

of antigenic sites on the haemagglutinin molecule of H5 avian influenza virus and

522

phenotypic variation of escape mutants. J Gen Virol 83 (Pt 10), 2497–2505.

523

Kolaskar, A.S., Tongaonkar, P.C., 1990. A semi-empirical method for prediction of

524 525

antigenic determinants on protein antigens. FEBS Lett 276, 172–174. Koopmans, M., Wilbrink, B., Conyn, M., Natrop, G., van der Nat, H., Vennema, H.,

526

Meijer, A., van Steenbergen, J., Fouchier, R., Osterhaus, A., Bosman, A., 2004.

527

Transmission of H7N7 avian influenza A virus to human beings during a large

24

Page 24 of 43

528

outbreak in commercial poultry farms in the Netherlands. Lancet 363 (9409), 587–

529

593.

530 531

Laursen, N.S., Wilson, I.A., 2013. Broadly neutralizing antibodies against influenza viruses. Antiviral Research 98 (3), 476–483. Linke, S., Neubauer, K., Dorner, M.B., Dorner, B.G., Pauli, G., Schweiger, B., 2011.

533

Generation and characterisation of monoclonal antibodies against influenza virus

534

A, subtype H5N1. J Virol Methods 175, 85–94.

cr

Parker, J., Guo, D., Hodges, R., 1986. New hydrophilicity scale derived from high-

us

535

ip t

532

performance liquid chromatography peptide retention data: correlation of

537

predicted surface residues with antigenicity and X-ray-derived accessible sites.

538

Biochemistry 25, 5425–5432.

Peiris, M., Yuen, K.Y., Leung, C.W., Chan, K.H., Ip, P.L., Lai, R.W., Orr, W.K.,

M

539

an

536

Shortridge, K.F., 1999. Human infection with influenza H9N2. Lancet 354 (9182),

541

916–917.

Pellequer, J.-L., Westhof, E., van Regenmortel, M., 1993. Correlation between the

Ac ce pt e

542

d

540

543

location of antigenic sites and the prediction of turns in proteins. Immunol Lett 36,

544

83–99.

545

Ponnuswamy, P.K., Prabhakaran, M., Manavalan, P., 1980. Hydrophobic packing

546

and spatial arrangement of amino acid residues in globular proteins. Biochim

547

Biophys Acta 623, 301–316.

548

Puthavathana, P., Auewarakul, P., Charoenying, P.C., Sangsiriwut, K., Pooruk, P.,

549

Boonnak, K., Khanyok, R., Thawachsupa, P., Kijphati, R., Sawanpanyalert, P.,

550

2005. Molecular characterization of the complete genome of human influenza

551

H5N1 virus isolates from Thailand. J Gen Virol 86 (Pt 2), 423–433.

25

Page 25 of 43

Russell, C., Fonville, J.M., Brown, A.E.X., Burke, D.F., Smith, D.L., James, S.L.,

553

Herfst, S., van Boheemen, S., Linster, M., Schrauwen, E.J., Katzelnick, L.,

554

Mosterín, A., Kuiken, T., Maher, E., Neumann, G., Osterhaus, A., Kawaoka, Y.,

555

Fouchier, R., J, S.D., 2012. The potential for respiratory droplet–transmissible

556

A/H5N1 influenza virus to evolve in a mammalian host. Science 336 (6088),

557

1541–1547.

Saha, S., Raghava, G. BcePred: Prediction of Continuous B-Cell Epitopes in

cr

558

ip t

552

Antigenic Sequences Using Physico-chemical Properties. In G.Nicosia, V.Cutello,

560

P.J. Bentley and J.Timis (Eds.) ICARIS 2004, LNCS 3239, 197-204, Springer,

561

2004.

564

an

563

Shaw, M.L., Stone, K.L., Colangelo, C.M., Gulcicek, E.E., Palese, P., 2008. Cellular proteins in influenza virus particles. PLoS Pathog 4 (6), e1000085.

M

562

us

559

Shehata, A., Fiebig, P., Sultan, H., Hafez, M., Liebert, U.G., 2012. Development of a recombinant ELISA using yeast (Pichia pastoris)-expressed polypeptides for

566

detection of antibodies against avian influenza A subtype H5. J Virol Methods 180

567

(1-2), 18–25.

Ac ce pt e

d

565

568

Skehel, J.J., Stevens, D.J., Daniels, R.S., Douglas, A.R., Knossow, M., Wilson, I.A.,

569

Wiley, D.C., 1984. A carbohydrate side chain on hemagglutinins of Hong Kong

570

influenza viruses inhibits recognition by a monoclonal antibody. Proc Natl Acad

571

Sci USA 81 (6), 1779–1783.

572

Squires, B., Macken, C., Garcia-Sastre, A., Godbole, S., Noronha, J., Hunt, V.,

573

Chang, R., Larsen, C.N., Klem, E., Biersack, K., Scheuermann, R.H., 2007.

574

BioHealthBase: informatics support in the elucidation of influenza virus host

575

pathogen interactions and virulence. Nucleic Acids Research 36 (Database),

576

D497. 26

Page 26 of 43

577 578 579

Sullivan, S.J., Jacobson, R.M., Dowdle, W.R., Poland, G.A., 2010. 2009 H1N1 influenza. Mayo Clin Proc 85 (1), 64–76. Tong, S., Li, Y., Rivailler, P., Conrardy, C., Castillo, D.A.A., Chen, L.-M., Recuenco, S., Ellison, J.A., Davis, C.T., York, I.A., Turmelle, A.S., Moran, D., Rogers, S., Shi,

581

M., Tao, Y., Weil, M.R., Tang, K., Rowe, L.A., Sammons, S., Xu, X., Frace, M.,

582

Lindblade, K.A., Cox, N.J., Anderson, L.J., Rupprecht, C.E., Donis, R.O., 2012. A

583

distinct lineage of influenza A virus from bats. Proceedings of the National

584

Academy of Sciences.

cr us

585

ip t

580

Tong, S., Zhu, X., Li, Y., Shi, M., Zhang, J., Bourgeois, M., Yang, H., Chen, X., Recuenco, S., Gomez, J., Chen, L.-M., Johnson, A., Tao, Y., Dreyfus, C., Yu, W.,

587

McBride, R., Carney, P.J., Gilbert, A.T., Chang, J., Guo, Z., Davis, C.T., Paulson,

588

J.C., Stevens, J., Rupprecht, C.E., Holmes, E.C., Wilson, I.A., Donis, R.O.,

589

Subbarao, K., 2013. New World Bats Harbor Diverse Influenza A Viruses. PLoS

590

Pathog 9 (10), e1003657.

M

d

Ungchusak, K., Auewarakul, P., Dowell, S.F., Kitphati, R., Auwanit, W.,

Ac ce pt e

591

an

586

592

Puthavathana, P., Uiprasertkul, M., Boonnak, K., Pittayawonganon, C., Cox, N.J.,

593

Zaki, S.R., Thawatsupha, P., Chittaganpitch, M., Khontong, R., Simmerman, J.M.,

594

Chunsutthiwat, S., 2005. Probable person-to-person transmission of avian

595

influenza A (H5N1). N Engl J Med 352 (4), 333–340.

596

Vareckova, E., Mucha, V., Wharton, S.A., Kostolansky, F., 2003. Inhibition of fusion

597

activity of influenza A haemagglutinin mediated by HA2-specific monoclonal

598

antibodies. Arch Virol 148 (3), 469–486.

599

Wang, H., Feng, Z., Shu, Y., Yu, H., Zhou, L., Zu, R., Huai, Y., Dong, J., Bao, C.,

600

Wen, L., Wang, H., Yang, P., Zhao, W., Dong, L., Zhou, M., Liao, Q., Yang, H.,

601

Wang, M., Lu, X., Shi, Z., Wang, W., Gu, L., Zhu, F., Li, Q., Yin, W., Yang, W., Li, 27

Page 27 of 43

602

D., Uyeki, T.M., Wang, Y., 2008. Probable limited person-to-person transmission

603

of highly pathogenic avian influenza A (H5N1) virus in China. The Lancet 371

604

(9622), 1427–1434. Webster, R.G., Bean, W.J., Gorman, O.T., Chambers, T.M., Kawaoka, Y., 1992.

606

Evolution and ecology of influenza A viruses. Microbiol Rev 56 (1), 152–179.

608

Webster, R.G., Monto, A.S., Braciale, T.J., Lamb, R.A., 2013. Textbook of Influenza. John Wiley & Sons, Ltd, Oxford, UK.

cr

607

ip t

605

Wei, S.-H., Yang, J.-R., Wu, H.-S., Chang, M.-C., Lin, J.-S., Lin, C.-Y., Liu, Y.-L., Lo,

610

Y.-C., Yang, C.-H., Chuang, J.-H., Lin, M.-C., Chung, W.-C., Liao, C.-H., Lee, M.-

611

S., Huang, W.-T., Chen, P.-J., Liu, M.-T., Chang, F.-Y., 2013. Human infection

612

with avian influenza A H6N1 virus: an epidemiological analysis. The Lancet

613

Respiratory Medicine 1 (10), 771–778.

an

M

614

us

609

WHO, 2002. WHO manual on animal influenza diagnosis and surveillance. (document WHO/CDS/CSR/NCS/2002.5. Rev.1). http/ / www.who.int/ csr/

616

resources/ publications/ influenza/ whocdscsrncs20025rev.pdf. Accessed 10

617

December 2013.

Ac ce pt e

618

d

615

WHO, 2007. Recommendations and laboratory procedures for detection of avian

619

influenza A(H5N1) virus in specimens from suspected human cases.

620

http://www.who.int/influenza/resources/documents/h5n1_laboratory_procedures/e

621

n/. Accessed 4 November 2013.

622 623

WHO, 2008. Update on Avian Influenza A (H5N1) Virus Infection in Humans. N Engl J Med 358 (3), 261–273.

624

Wiley, D.C., Wilson, I.A., Skehel, J.J., 1981. Structural identification of the antibody-

625

binding sites of Hong Kong influenza haemagglutinin and their involvement in

626

antigenic variation. Nature 289 (5796), 373–378. 28

Page 28 of 43

627

World Organisation for Animal Health (OIE), 2004. Manual of Diagnostic Tests and

628

Vaccines for Terrestrial Animals, Chapter 2.3.4. Avian Influenza, version adopted

629

may 2012. www.oie.int/international-standard-setting/terrestrial-manual/access-

630

online/. Accessed 4 November 2013. Yang, M., Clavijo, A., Graham, J., Salo, T., Hole, K., Berhane, Y., 2009. Production

ip t

631

and diagnostic application of monoclonal antibodies against influenza virus H5. J

633

Virol Methods 162 (1-2), 194–202.

Ac ce pt e

d

M

an

us

cr

632

29

Page 29 of 43

Legends to Figures

635

Figure 1: Diagram of influenza A virus HA protein. Truncated sequences (P1, P2,

636

P3, and P4 as well as recombinant rHA1) and their position within the linear protein

637

sequence are shown. Arrow indicates cleavage site between HA1 and HA2, which

638

remain connected via a disulfide bond.

ip t

634

639

Figure 2: Western blot of purified polypeptides using (His)6-tag-antibody. The

641

theoretical molecular weights of P1, P2, P3 and P4 are 7.59 kDa, 16.06 kDa, 7.67

642

kDa and 14.26 kDa, respectively.

us

cr

640

an

643

Figure 3: Immunofluorescence assay using murine immune sera (dilution 1:50)

645

on Vero cells infected with influenza virus. As control monoclonal Influenza-A

646

nucleoprotein-specific antibody (anti-NP, kindly provided by Prof. Jassoy, Institute of

647

Virology, Leipzig University) was used. No reactivity was seen with P3 immune

648

serum.

d

Ac ce pt e

649

M

644

650

Figure 4: Reactivity of mAb FD6 and HE4. Using Western blot mAb FD6 (A) and

651

HE4 (B) reacted only with H5 viruses. Indirect ELISA (C) and IFA (D) revealed

652

reactivity of both mAbs with H5N1, mAb HE4 reacted additionally with H5N6.

653 654

Figure 5: Sequence alignment of identified linear epitopes (shaded in grey) with

655

adjoining residues and the homologous regions of other influenza virus

656

subtypes. Identical residues are shown by dots. Sequence conservation is detected

657

within H5 subtype for both epitopes, while sequence differences are found in H1, H2, 30

Page 30 of 43

658

H3, H7 proteins. Binding of FD6 is influenced by adjacent residues DLD, while for

659

HE4 residues QE and IH are important for binding.

660

Figure 6: Antigen-Capture ELISA using FD6 as capture antibody. Recent isolates

662

from HPAIV (H5N1 Th, H5N1 Eg) were well detected, while historic isolates (H5N1

663

Sc and H5N1 Ge) were not (A). The detection of H5N6 by AC-ELISA is concentration

664

dependent (B).

cr

ip t

661

Ac ce pt e

d

M

an

us

665

31

Page 31 of 43

665

Tables

666

Table 1:

667

Influenza viruses used in this study with GenBank protein accession numbers

668

Accession number

human

H5N1

A/Thailand/1(Kan-1)/04

AAS65615

H1N1

A/Fukushima/141/06

ACM17297

A/Brisbane/59/07

ACA28844

A/New Caledonia/20/99 H3N2

A/California/7/04

ip t

Virus strain

cr

Virus subtype

ABH01021

an

A/Wisconsin/67/05

M

A/Brisbane/10/07

ACF54576 ABW23353

A/chicken/Scotland/59

ACZ48553

H5N1

A/teal/Germany/WV632/05

CAL59779

H5N1

A/chicken/Egypt/5610NAMRU-

d

H5N1

Ac ce pt e

Avian

AAP34324

us

Origin

DQ837587

F3/2006

Swine

H5N2

A/duck/Potsdam/1402-6/86

ABI84497

H5N3

A/teal/England/7894/06

Not available

H5N3

A/turkey/Germany/R1612/08

Not available

H5N6

A/duck/Potsdam/2216-4/84

ABB88348

H3N8

A/duck/Ukraine/1/63

ABB88369

H1N1

VL2008/32261-3/1

Not available

H3N2

VL2008/27783-2/4

Not available

669

32

Page 32 of 43

669

Table 2:

670

Sequence of primers used for amplification of truncated HA sequences Primer sequence (5’- 3’)b

P1

P1s

GTA CTC GAG AAG AGA GAG GCT GAA GCA GAT CTA GAT GGA GTG AAG CC

P1as

CAT GCG GCC GCC TTC TCC ACT ATG TAG GAC C

P2s

GTA CTC GAG AAG AGA GAG GCT GAA GCA AAT AAT ACC AAC CAA GAA GAT C

P2as

CAT GCG GCC GCG TCC CCT TTC TTG ACA ATT TTG

P3s

GAA CTC GAG AAG AGA GAG GCT GAA GCA CAC AAT ATA CAC CCT CTC ACC

P3as

CAT GCG GCC GCT CTC TTT TTT CTT CTT CTC TCT C

P4s

GCA CTC GAG AAG AGA GAG GCT GAA GCA AAA GAA TCC ACT CAA AAG GC

P4as

CTT GCG GCC GCT TCC TTT GCA TTA TCC CTA AGC

rHA1s

GTA CTC GAG AAG AGA GAG GCT GAA GCA GAT CAG ATT TGC ATT GGT TAC C

rHA1

cr

us

an

M

P4

d

P3

Ac ce pt e

P2

ip t

PolyPrimer a peptide

rHA1as

GAT GCG GCC GCT CTT TGA GGG CTA TTT CTG AGC C

671

a

672

virus subtype H5N1 (A/Thailand/1(Kan-1)/ 2004).

673

b

674

NotI (antisense primer) used for cloning are written in bold.

P1, P2, P3, P4 and rHA1 DNA fragments coding for HA sequences of influenza A

Gene specific sequences are underlined, restriction sites of XhoI (sense primer) and

675

33

Page 33 of 43

675

Table 3:

676

Designation,

677

haemagglutinin polypeptides P1-P4.

Poly-

location

Location

within

H5

protein

and

sequence

Sequenceb

four

DLDGVKPLIL RDCSVAGWLL GNPMC

us

HA1 165-261

7.59

cr

HA1 43-82

ip t

[kDa]

DEFIN VPEWSYIVEK P2

the

mol wtc

peptidea P1

of

NNTNQEDLLV LWGIHHPNDA AEQTK

16.06

an

LYQNP TTYISVGTST LNQRLVPRIA

TRSKVNGQSG RMEFFWTILK PNDAI

M

NFESN GNFIAPEYAY KIVKKGD HA1 292-330

HNIHPLTIGE CPKYVKSNRL VLATG

d

P3

7.67

P4

Ac ce pt e

LRNSP QRERRRKKR

HA2 38-132

KESTQKAIDG VTNKVNSIID KMNTQ

14.26

FEAVG REFNNLERR IENLNKKMED GFLDVWTYNA ELLVLMENER TLDFH DSNVK NLYDKVRLQL RDNAKE

678

a

679

(A/Thailand/1(Kan-1)/ 2004) haemagglutinin.

680

b

681

using method of Kolaskar and Tongaonkar (1990) are written bold.

P1, P2, P3 and P4 truncated sequences of influenza A subtype H5N1

Epitopes predicted by three of seven algorithms are underlined, epitopes predicted

34

Page 34 of 43

682

c

683

glycosylation site, molecular weight of N-linked oligosaccharide is calculated for the

684

dominant oligosaccharide structure found in Pichia pastoris, i.e. Man9GlcNAc2 with 2

685

kDa (Montesino et al., 1998).

Calculated molecular weight of his-tagged polypeptides. P2 contains a potential N-

ip t

686 687

cr

688

us

689

Table 4:

691

Relative conservation of epitopes of mAbs FD6 and HE4 identified using

692

epitope mapping.

an

690

mAb

Epitopea

94

VKPLI

Ac ce pt e

HE4

H5Nx

H7Nx

H9Nx

H3Nx

H1Nx

49

-

-

-

-

39,9

-

-

-

-

d

H5N1 FD6

M

Relative conservationb [%]

QEDLLVLWGIH

92

693

a

Amino acids important for antibody binding and used for analysis are written bold.

694

b

Relative epitope conservation in isolates from 1990-2013 of subtypes H5N1 (1537

695

isolates), H5Nx (461 isolates, excluding N1-subtype), H7Nx (574 isolates), H9Nx

696

(937 isolates), H3Nx (3673 isolates) including 360 Canadian/American H3N2 variants

697

from swine and H1Nx (5991 isolates) was determined using „Identify Short Peptides

698

in

699

http://www.fludb.org, 16.05.2014, Squires et al., 2007).

Proteins“

tool

provided

by

Influenza

Research

Database

(IRD,

700

35

Page 35 of 43

700

Table 5:

701

Relative sensitivity and specificity between AC-ELISA and RT-PCR

704

Positive

13

0

Negative

3

14

Total

16

14

Relative sensitivity

81%

Relative specificity

100%

cr

13

ip t

Negative

17

M

an

30

d

703

Positive

Ac ce pt e

702

Total

RT-PCR

us

AC-ELISA

36

Page 36 of 43

704

Highlights

705 706

-

Algorithms were used to indentify B-cell epitopes within influenza H5.

707

-

Conserved

708

-

Expressed antigens were used for generation of monoclonal antibodies.

709

-

Two

710

-

Antigen-Capture ELISA for specific detection of H5N1 virus was developed.

expressed

antibodies

specific

for

in

Pichia

H5

pastoris.

were

established.

cr

monoclonal

were

ip t

epitopes

us

711 712

Ac ce pt e

d

M

an

713

37

Page 37 of 43

Ac

ce

pt

ed

M

an

us

cr

i

Figure(s)

Page 38 of 43

Ac

ce

pt

ed

M

an

us

cr

i

Figure(s)

Page 39 of 43

Ac

ce

pt

ed

M

an

us

cr

i

Figure(s)

Page 40 of 43

Ac ce p

te

d

M

an

us

cr

ip t

Figure(s)

Page 41 of 43

Ac

ce

pt

ed

M

an

us

cr

i

Figure(s)

Page 42 of 43

Ac ce p

te

d

M

an

us

cr

ip t

Figure(s)

Page 43 of 43

Generation of monoclonal antibodies reactive against subtype specific conserved B-cell epitopes on haemagglutinin protein of influenza virus H5N1.

H5-specific monoclonal antibodies may serve as valuable tools for rapid diagnosis of H5N1 avian influenza virus. Therefore, conserved H5-specific sequ...
347KB Sizes 0 Downloads 11 Views