Vaccine Design against Coronavirus Spike (S) Glycoprotein in Chicken: Immunoinformatic and Computational Approaches

DOI: https://doi.org/10.21203/rs.3.rs-25375/v2

Abstract

Background: Infectious bronchitis (IB) is a highly contagious respiratory disease in chickens and produces economic loss within the poultry industry. This disease is caused by a single stranded RNA virus belonging to Cronaviridae family. This study aimed to design a potential multi-epitopes vaccine against Infectious bronchitis virus spike protein (S). Protein characterization was also performed for IBV spike protein.

Methods: The present study used various tools in Immune Epitope Database (IEDB) to predict conserved B and T cell epitopes against IBV spike (S) protein that may perform a significant role in provoking the resistance response to IBV infection.

Results: In B cell prediction methods, three epitopes (1139KKSSYY1144, 1140KSSYYT1145, 1141SSYYT1145) were selected as surface, linear and antigenic epitopes. 

Many MHCI and MHCII epitopes were predicted for IBV S protein. Among them 982YYITARDMY990 and 983YITARDMYM991 epitopes displayed high antigenicity, no allergenicity and no toxicity as well as great linkage with MHCI and MHCII alleles. Moreover, docking analysis of MHCI epitope produced strong binding affinity with BF2 alleles.  

Conclusion: Five conserved epitopes were expected from spike glycoprotein of IBV as the best B and T cell epitopes due to high antigenicity, no allergenicity and no toxicity. In addition, MHC epitopes showed great linkage with MHC alleles as well as strong interaction with BF2 alleles. These epitopes should be designed and incorporated and then tested as multi-epitope vaccine against IBV.

Introduction

Infectious bronchitis virus (IBV) is a single Positive stranded RNA that belonging to coronavirus of the chicken (Gallus gallus). It's a highly contagious respiratory disease in chickens that's mainly severe for very young chicks. The signs of illness include tracheal rales, coughing, sneezing, nasal discharge and some strains may cause kidney damage [1, 2]. The disease can be transmitted in respiratory discharges and feces by infected chickens, and it is spread by aerosol, ingestion of contaminated feed and water, and contact with contaminated equipment or clothing. The virus is not transmitted via eggs [3]. The disease causes economic loss within the poultry industry, affecting the performance of meat-type and egg-laying birds. The disease can affect all ages, but the clinical disease is more severe in young chicks. Chicks become more resistant to IBV-induced mortality with the increasing age [4].  

There are four structural proteins related to the envelope, the spike (S), membrane (M), envelope (E), and nucleocapsid (N) protein [5]. The spike 'S' glycoprotein which located at the surface of the virion. The membrane 'M' glycoprotein is partially exposed at the surface of the virion and also the nucleocapsid 'N' protein that located internally. The spike glycoprotein of IBV induces virus neutralizing (VN) and HI antibodies and has been considered as the most likely inducer of protection [2, 4].  The S protein is either a dimer or trimer. It has two recognized functions; binding the virus to receptor molecules on host cells, and activating fusion of the virion membrane with host cell membranes, releasing the viral genome into the cell [2]. The spike gene in particular the S1 part, is highly variable, due to insertions, deletions, substitutions and recombination events [6].  Application of vaccine is the most effective way to protect against pathogenic diseases, particularly when these pathogens have a high mortality rate such as IBV and viruses in general. On the other hand, the large number of serotypes and strains (genotype) of IBV specifically complicate control method. IBV has shift and drift property [7].

Inactivated and live-attenuated vaccines are employed to control the disease. However, inactivated vaccines often fail to induce strong cellular immunity, while live-attenuated vaccines can contribute to development of antigenic variant viruses [5].  The increasing number of new IBV serotypes, caused by frequent gene mutation and recombination, poses a major challenge for the prevention and control of infectious bronchitis disease [8].

RNA viruses such as IBV have high mutational rates. Thus, the most important step in the design of cross-protective peptide vaccine against IBV is to target the conserved epitopes of different IBV serotypes [5].

Presentation by MHC molecules is important for developing vaccinal immunity. MHC class I and class II molecules are typically highly polymorphic and polygenic [9].  Avian MHC class I and class II genes are localized into two regions (MHC-B and MHC-Y) on the chromosome 16. The MHC-B and MHC-Y haplotypes assort independently as the result of an intervening region that supports highly frequent recombination [9, 10]. Chicken MHC B–F molecules have been structurally and functionally related to mammalian MHC class I molecules and have been involved in the presentation of antigen to CD8+T lymphocytes, which is important for antiviral immune response [11].  Recently, the design of epitope-based vaccines has been expanded by developments in genomics, proteomics and the understanding of pathogens. Epitope is the negligible immunogenic region of a sequence of proteins that specifically produces accurate immune responses [12]. The identification of specific B and T cell epitopes produced more desirable manipulation of immune response [13]. It is known that designing of multi-epitopes vaccine using bioinformatics tools can significantly reduce the time and cost of production and produce satisfactory results [14, 15].

The production of safer and more reliable vaccines for controlling IBV is important. Therefore, the aim of this study is to analyze strains of spike (S) glycoprotein of infectious bronchitis virus reported in NCBI database using immunoinformatics and computational approaches to select all possible epitopes that can be used as multi-epitopes vaccine. Protein characterization was also achieved for IBV spike protein.

Material And Method

2.1. Protein Sequence Retrieval

Spike (S) protein sequences of different infectious bronchitis virus (IBV) strains were retrieved from the GeneBank of National Central Biotechnology Information (NCBI) (http://www.ncbi.nlm.nih.gov/protein/) database in March 2019. The sequences were saved in FASTA format (Table 1).

2.2 Structural analysis

Reference sequence of spike S protein (NP_040831.1) was analyzed to identify chemicals and physical properties including GRAVY (Grand average of hydropathicity), half-life, molecular weight, stability index and amino acids atomic composition using an online tool Protparam  [16].

Secondary structure of spike S protein of IBV was analyzed through PSIPRED [17]. The secondary structure of protein including helix, sheet, turn, and coil parameters was predicted using GOR IV server at https://npsa-prabi.ibcp.fr/cgi-bin/secpred_sopma.pl. TMHMM an online tool (http://www.cbs.dtu.dk/servi ces/TMHMM/), used to examine the trans-membrane topology of S protein. Presence of disulphide-bonds were predicted through an online tool DIANNA v1.1. It makes prediction based on trained neural system [18]. CDD-BLAST (http://www.ncbi.nlm.nih.gov/BLAST/) [19-21] and PFAM (https://pfam.xfam.org/) [22] were used to search the defined conserved domains in the targeted protein sequences. Blastp in NCBI database (https://blast.ncbi.nlm.nih.gov/Blast.cgi) using reference sequence (refseq-protein) database was used to compare spike reference sequences of different coronaviruses in human and animals against IBV spike protein sequence. Phylogenetic tree was also constructed based on COBALT multiple alignment (https://www.ncbi.nlm.nih.gov/blast/treeview/treeView.cgi) [19, 20].

Table 1. Accession numbers, date and area of collection of the retrieved sequences of Spike protein sequences of IBV.

No

Accession No

Country

Year

No

Accession No

Country

Year

1

NP_040831.1*

UK

2018

47

AAV98206.1

USA

2002

2

AHX25911.1

China

2016

48

AVX27612.1

India

2004

3

AHX25902.1

China

2016

49

ALE71331.1

India

2018

4

AHX25893.1

China

2016

50

AJP16712.1

China

2015

5

AMK51938.1

China

2016

51

AJP16739.1

China

2015

6

AEP84746.1

China

2016

52

AFP50306.1

Korea

2015

7

AEP84736.1

China

2016

53

AFP50302.1

Korea

2012

8

ACX71849.1

China

2011

54

AFP50294.1

Korea

2012

9

ACX71844.1

China

2011

55

AFP50274.1

Korea

2012

10

ACX71842.1

China

2011

56

AEL12221.1

China

2012

11

AAU09490.1

China

2011

57

ADY62552.1

China

2012

12

AAY24433.1
Singapore

2005

58

ADV71785.1
Netherlands

2010

13

AAY24423.1

 

Singapore

 

2005

59

ACQ55230.1

 

Netherlands

 

2011

14

AAY21248.1
Singapore

 

2005

60

ARE67884.1
 
Pakistan

 

2017

15

AAY21247.1
 
Singapore

 

2005

61

ARB66180.1

 

China

2017

16

AAY21246.1

 

Singapore

 

2005

62

AQY55821.1
 

China

2017

17

AAY21245.1

 

Singapore

 

2005

63

AHX26172.1
 

China

2016

18

AAY21244.1

 

Singapore

 

2005

64

AHX26163.1
 

China

2016

19

AAY21243.1

 

Singapore

 

2005

65

AHX26154.1
 

China

2016

20

AAY21242.1

Singapore

2005

66

AHX26145.1
 

China

2016

21

AGW24533.1

India

2015

67

AHX26136.1

China

2016

22

AAW33786.1

USA

2006

68

AHX26127.1

China

2016

23

AER08740.1

Sweden

2012

69

AHX26118.1

China

2016

24

AER08739.1

 

Sweden

2012

70

AHX26109.1
 

China

2016

25

AER08729.1

Sweden

2012

71

AHX26073.1
 

China

2016

26

AER08728.1

Sweden

2012

72

AHX26064.1
 

China

2016

27

AER08727.1

Sweden

2012

73

AHX26055.1

China

2016

28

AER08726.1

Sweden

2012

74

AHX26046.1

 

China

2016

29

AER08725.1

Sweden

2012

75

AHX26037.1

 

China

2016

30

AER08724.1

Sweden

2012

76

AHX26028.1

 

China

2016

31

AER08723.1

Sweden

2012

77

AHX26019.1

 

China

2016

32

AER08722.1

Sweden

2012

78

AHX26010.1

 

China

2016

33

AER08721.1

Sweden

2012

79

AHX26001.1

China

2016

34

ADA83557.1

 

USA

2011

80

AHX25992.1
 

China

2016

35

ADA83467.1

USA

2011

81

AHX25983.1
 

China

2016

36

ABH01142.1

USA

2007

82

AHX25974.1
 
China

2016

37

ABH01141.1

USA

2007

83

AHX25965.1
 
China

2016

38

ABI26423.1

USA

2006

84

AHX25956.1
 
China

2016

39

AAK27168.1

China

2005

85

AHX25947.1
 
China

2016

40

ACH72794.1

China

2009

86

AHX25938.1
 
China

2016

41

AAW83034.1

China

2006

87

AHX25929.1
 
China

2016

42

ARS23139.1

 
Egypt

2014

88

ACJ50199.1

 
Singapore
 

2005

43

AHX25920.1

China

2016

89

ACO37566.1

 
Singapore
 

2005

44

ADP06504.1

 
USA

2012

90

AYG86360.1

 
SouthKorea

2018

45

AAA66578.1

UK

1995

91

AYG86347.1

 

SouthKorea

2018

46

AAA70235.1

USA

2002

92

AAV28722.1

 

China

2006

*Refseq


2.3 Multiple sequence alignment and Epitope Conservancy Assessment

The retrieved sequences of IBV S protein were aligned using Clustal program and consensus sequence was generated using the multiple sequence alignment (MSA) tool, Jalview version 2.10.5.  (http://www.jalview.org/about/jalview-scientific-advisory-committee) [23]. Epitope conservancy analysis in Immune Epitope Database (IEDB) was used to detect potential epitope conservancy (http://tools.iedb.org/conservancy/) [24]. For calculating the conservancy score, the sequence identity threshold was kept at 80%.

2.4 Phylogeny analysis

Phylogenetic tree of the retrieved sequences of spike (S) protein was performed using MEGA7.0.26 (7170509) software using maximum likelihood parameter [25]. 

2.5 B cell prediction

The Immune Epitope Database (IEDB) (http://tools.iedb.org/mhci/) was used to predict B and T cell epitopes of IBV reference sequence of S proteins (NP_040831.1) [26].   Linear B-cell epitopes were predicted using BepiPred from IEDB [27]. Emini surface accessibility prediction tool was used to predict surface located epitopes [28]. Whereas, the antigenic epitopes were investigated using kolaskar and Tongaonkar antigenicity method [29].

Discontinuous epitopes were predicted using DiscoTope server [30]. The parameter was set at ≥ 0.5 which indicated 90% specificity and 23% sensitivity. This method is based on surface accessibility and amino acid statistics in a collected form dataset of discontinuous epitopes found out by X-ray crystallography of antigen/antibody protein buildings. Chimera software was used to display the position of predicted epitopes clusters on 3D structure of S protein [31].

2.6 T-cell epitope prediction

The T cell epitopes were predicted in human among different alleles of major histocompatibility complex class I (MHCI) and class II (MHCII).

MHC-I binding epitopes were predicted by the IEDB MHC I prediction tool at http://tools.iedb.org/mhci.  The binding affinity of peptides to MHC I molecules was measured using artificial neural networks (ANN) method [32, 33]. Prior to prediction, peptide lengths were set as 9 mers. The half maximum inhibitory concentration (IC50) values needed for the binding of peptide to MHC-I molecules were set less than or equal to 300 nM.

The IEDB MHCII prediction tool was used for MHC class II molecules at (http://tools.iedb.org/mhcii/) [26]. Human MHC class II alleles (HLA DR, HLADP and HLADQ) were used for MHCII binding predication. The NN-align method was used with IC50 less or equal to 1000 nM [34].

2.7 Antigenicity, allergenicity and toxicity of epitopes

VaxiJen v2.0 server was used to predict the antigenicity of the conserved regions (http://www.jenner.ac.uk/VaxiJen) [35]. The default prediction parameters and a threshold value of 0.4 were used. The in silico allergenicity prediction of epitopes was investigated using AllerTop v .2.0 (http://www.ddg-pharmfac.net/AllerTOP)[36]. While ToxiPred server was used to evaluate the toxicity of predicted epitopes. (http://crdd.osdd.net/raghava/toxinpred/) [37].

2.8 Homology Modeling:

IBV reference sequences and the protein sequences of BF alleles (BF2 *2101 and BF2*0401) were submitted to Raptor X server (http://raptorx.uchicago.edu/) to design their three D structures [38-40]. PEPFOLD3 server was used for the homology modelling of MHCI epitopes (http://bioserv.rpbs.univ-paris-diderot.fr/services/PEP-FOLD3/) from amino acid sequences [41-43]. Chimera software 1.8 was used to visualize 3D structures of IBV spike S protein reference sequence and BF alleles [31].

2.9 Molecular Docking                                                                                            

To perform molecular docking, 3D structures of MHCI epitopes and BF alleles were submitted simultaneously to the PatchDock online autodock tools; an automatic server for molecular docking (https://bioinfo3d.cs.tau.ac.il/PatchDock/) [44]. The five top models were selected using firedock [44]. The results were visualized using the UCSF-Chimera software 1.8 [31]. 

Results

3.1 Structural analysis

The physiochemical properties of the spike S protein, measured through protparam, showed that it contained 1162 amino acids (aa) with a molecular weight of 128046.70 kDa. The spike protein showed an antigenic nature when subjected to vaxijen v2.0.

Theoretical isoelectric point (PI) of spike protein was 7.71, indicating its positive in nature. An isoelectric point above 7 indicates the protein is charged positively. Near to 81 aa charges were found negative, whereas 84 aa found positive.

Protparam computed instability-index (II) 35.53, this categorize the protein as stable. Aliphatic-index 86.05, which devotes a thought to the proportional volume holding by aliphatic side chain and GRAVY value of the protein sequence is 0.012. Half-life of protein shown as the total time taken for its vanishing after it has been synthesized in cell, computed as 30 h for mammalian-reticulocytes, > 20 h for yeast, > 10 h for Escherichia coli. The total numbers of Carbon (C), Oxygen (O), Nitrogen (N), Hydrogen (H) and Sulfur (S) were entitled by the formula: C5737H8847N1495O1718S56

The secondary structure of IBV spike S protein was analyzed through PSIPRED and GOR IV server. The components of secondary structure prediction by GOR IV server are alpha helix (29.43%), extended strand (27.37%), beta turn (5.25%), and random coil (37.95%) (Fig.1).

DiANNA1.1 tool calculated 19 disulphides bond (S–S) positions and assign them a score and makes prediction based on trained neural system. The trans-membrane protein topology was investigated via online tool TMHMM. Residues from 1 to 1093 were found to be exposed to the surface, residue from 1094 to 1116 were found inside trans-membrane-region and residues from 1117 to 1162 were buried within the core-region of the S protein (Fig.1).

In refseq of IBV spike protein two conserved domains (Corona-S2, Corona-S2) were identified. The conserved domains were sequenced by Conserved Domain (CDD) BLAST search. The results revealed that corona-S1 (pfam01600) is the only member of the superfamily cl03276 and corona-S2 domain (pfam01601) is the only member of the superfamily cl20218. The top associated sequences in both domains were Feline infectious peritonitis virus (strain 79-1146), Avian infectious bronchitis virus (strain Beaudette), and Human coronavirus 229E while Severe acute respiratory syndrome-related coronavirus sequences were associated only with corona-S2 domain. The closest homologue obtained from BLASTP (refseq-protein) results was the Turkey coronavirus S protein with E value 0.00 followed by Murine hepatitis virus strain JHM with E value 9e-109 when comparing various coronaviruses in human and animals with IBV spike protein sequence (Table 2).   Phylogenetic tree of IBV against other coronaviruses in human and animals was created based on COBALT multiple alignment see Fig. 2.

Table 2: Blastp similarity search of IBV against other coronaviruses in human and animals.

NCBI Protein ID

Protein Name

E- value

identity

YP_001941166.1

Turkey coronavirus

0.0

38.59%

YP_009194639.1

Camel alphacoronavirus

8e-126

34.05%

YP_009199242.1

Swine enteric coronavirus

4e-124

31.47%

YP_003767.1

Human coronavirus NL63

2e-121

34.01%

NP_598310.1

Porcine epidemic diarrhea virus

8e-120

31.36%

YP_009273005.1

Rousettus bat coronavirus

1e-115

32.86%

NP_058424.1

Transmissible gastroenteritis virus

2e-109

32.03%

YP_209233.1

Murine hepatitis virus strain JHM

9e-109

37.20%

YP_004070194.1

Feline infectious peritonitis virus

1e-108

31.70%

YP_003858584.1

Bat coronavirus BM48-31/BGR/2008

4e-107

35.69%

NP_828851.1

E2 glycoprotein precursor [Severe acute respiratory syndrome-related coronavirus]

5e-107

36.28%

YP_009724390.1

Severe acute respiratory syndrome coronavirus 2

1e-106

36.31%

YP_009555241.1

Human coronavirus OC43

3e-105

31.42%

YP_009047204.1

Middle East respiratory syndrome-related coronavirus

2e-104

34.71%


3.2 Multiple sequence alignment

Jalview was used to visualize the multiple sequence alignment of the retrieved sequences. Several areas in alignment were shown to have mutation see Fig. 3.

3.3 Phylogeny

Phylogenetic trees for IBV spike S proteins were constructed using MEGA7.0.26 (7170509) software using maximum likelihood parameter see Fig. 4.

3.4 B-cell Epitopes

Several epitopes were predicted in B cell prediction methods using the Bepipred Linear Epitope Prediction tool. The conservancy percentages of these epitopes are presented in table 3.   After shortening of predicted epitopes, twenty one linear conserved epitopes were recognized. Of these, seven epitopes with different lengths were identified as linear, surface and antigenic epitopes between the positions 1139 – 1146 (see table 4).  These epitopes were 1139KKSSYY1144, 1140KSSYYT1145, 1141SSYYTT1146, 1141SSYYT1145, 1142SYYTT1146, 1142SYYT1145, and 1143YYTT1146. Based on the length and antigenicity score, three epitopes (1139KKSSYY1144, 1140KSSYYT1145, 1141SSYYT1145) were selected as top B cell epitopes.

Discotope 2.0 server was used to predict the discontinuous epitopes from the 3D structure of S protein (PDB ID: 6CV0), 90% specificity, − 3.700 threshold and 22.000 Angstroms propensity score radius[45]. Total 30 discontinuous epitopes were recognized at different exposed surface areas (Table 5). The position of each predicted epitope on the surface of 3D structure of S protein is shown in Fig.5 using Chimera visualization tool [31].

 Table 3.   Conservancy assessment of B cell linear epitopes 

Epitope no 

Epitope sequence

Start

End

Epitope length

Percent of protein sequence matches at identity <= 100%

1

MTAPSSGMAW

83

92

10

89.13% (82/92)

2

GGPI

193

196

4

90.22% (83/92)

3

TGNFSD

235

240

6

97.83% (90/92)

4

GPLQGGCK

352

359

8

94.57% (87/92)

5

DSAV

450

453

4

91.30% (84/92)

6

VNPCEDV

488

494

7

96.74% (89/92)

7

RNETGSQ

512

518

7

94.57% (87/92)

8

VGQKE

642

646

5

81.52% (75/92)

9

STKPAGFNTP

656

665

10

81.52% (75/92)

10

PQNAPN

926

931

6

98.91% (91/92)

11

ANASQY

959

964

6

98.91% (91/92)

12

IVPA

966

969

4

86.96% (80/92)

13

DFDFN

1026

1030

5

84.78% (78/92)

14

SKWWNDTKHELP

1034

1045

12

94.57% (87/92)

15

GKKSSYYTT

1138

1146

9

97.83% (90/92)

 

 

Table 4:  List of shortened B cell epitopes predicted by different B cell scale 

No.

Peptide

Start

End

Length

Emini

koleskar

1

MTAP

83

86

4

0.949

0.966

2

GSRIQT

406

411

6

1.273

0.973

3

SRIQT

407

411

5

1.583

0.992

4

SRIQ

407

410

4

1.355

1.013

5

STKP

656

659

4

2.543

0.979

6

VGLP

704

707

4

0.398

1.143

7

VGLPT

704

708

5

0.465

1.096

8

NASQY

960

964

5

2.034

1.006

9

SKWW

1034

1037

4

1.26

0.932

10

KKSSYYTT

1139

1146

8

6.723

1.003

11

KSSYYTT

1140

1146

7

4.166

1.013

12

SSYYTT*

1141

1146

6

2.568

1.027

13

SYYTT*

1142

1146

5

2.359

1.03

14

YYTT*

1143

1146

4

1.26

1.035

15

KKSSYYT

1139

1145

7

5.773

1.016

16

KKSSYY*

1139

1144

6

4.931

1.034

17

KSSYYT*

1140

1145

6

3.559

1.031

18

KKSSY

1139

1143

5

3.875

1.009

19

KKSS

1139

1142

4

3.054

0.971

20

SSYYT*

1141

1145

5

2.191

1.051

21

SYYT*

1142

1145

4

2.019

1.061

            *Shortened peptide that has high score in both Emini and kolaskar 


Table 5.  Discontinuous epitopes predicted through DISCOTOPE 2.0 Server 

Residue ID

Residue Name

Contact Number

Propensity Score

Discotope Score

262

SER

2

-3.91

-3.69

263

VAL

4

-2.626

-2.784

264

ASN

0

-0.238

-0.211

265

THR

19

-1.418

-3.44

266

THR

5

0.483

-0.148

267

PHE

25

-0.627

-3.43

268

THR

7

-0.463

-1.215

387

GLY

1

-3.678

-3.37

414

GLU

7

-0.476

-1.226

415

PRO

8

0.187

-0.754

417

VAL

5

-0.324

-0.862

419

THR

6

1.351

0.506

420

ARG

0

1.529

1.353

421

HIS

11

0.482

-0.838

422

ASN

12

-2.504

-3.596

515

THR

4

-3.071

-3.178

531

GLY

5

-1.472

-1.877

532

THR

5

1.933

1.136

533

ARG

0

1.404

1.243

534

ARG

0

-0.425

-0.376

648

MET

5

-1.103

-1.551

649

GLU

16

-1.752

-3.39

650

LEU

26

-4.013

-6.541

651

LEU

10

-2.379

-3.256

652

ASN

12

-1.72

-2.902

655

SER

7

-2.994

-3.454

685

SER

0

-3.842

-3.4

741

ILE

15

-1.303

-2.878

893

GLN

7

-2.411

-2.939

896

GLU

9

-2.901

-3.602

 

3.5 Prediction of MHC class I epitopes

In this study, the Human MHC class-I HLA alleles were used to explore the interaction of epitopes with MHCI alleles as chicken MHC alleles don't exists in IEDB database. MHC-1 binding prediction tool using IEDB database expected thirteen conserved epitopes of Spike protein (S) which were interacted with many cytotoxic T cell alleles.  These epitopes were 1115FFMTGCCGC1123, 590FNLTVTDEY598, 734GLLVLPPII742, 1105IIFILILGW1113, 1139KKSSYYTTF1147, 1087KTYIKWPWY1095, 166SVYLNGDLV174, 985TARDMYMPR993, 1145TTFDNDVVT1153, 983YITARDMYM991, 1144YTTFDNDVV1152, 982YYITARDMY990, 1143YYTTFDNDV1151.

3.6 Prediction of MHC class II epitopes

MHC-II binding prediction tool based on NN-align with half-maximal inhibitory concentration (IC50) ≤ 1000 was used. Thirty conserved core sequences were predicted to interact with MHCII alleles. These cores were 694EDLLFTSVE702, 1147FDNDVVTEQ1155, 1115FFMTGCCGC1123, 1116FMTGCCGCC1124, 590FNLTVTDEY598, 734GLLVLPPII742, 1105IIFILILGW1113, 902INECVKSQS910, 984ITARDMYMP992, 901KINECVKSQ909, 1139KKSSYYTTF1147, 1140KSSYYTTFD1148, 1187KTYIKWPWY1195, 735LLVLPPIIT743, 592LTVTDEYIQ600, 1014NKTVITTFV1022, 893QQRELATQK901, 894QRELATQKI902, 895RELATQKIN903, 589SFNLTVTDE597, 1141SSYYTTFDN1149, 166SVYLNGDLV174, 1142SYYTTFDND1150, 985TARDMYMPR993, 1146TFDNDVVTE1154, 593TVTDEYIQT601, 1013VNKTVITTF1021, 983YITARDMYM991, 1144YTTFDNDVV1152, 982YYITARDMY990 and 1143YYTTFDNDV1151

3.7 Antigenicity, allergenicity and toxicity of MHCI and MHCII epitopes:

The predicted epitopes of MHCI and MHCII were subjected to VaxiJen v2.0 server, AllerJen v2.0. and ToxiPred to estimate the potential antigenicity, allergenicity and toxicity of epitopes. Five MHCI epitopes were identified as antigenic, non-allergic and non-toxic, but only three epitopes (985TARDMYMPR993, 983YITARDMYM991 and 982YYITARDMY990) showed a high linkage with MHCI alleles (Table 6). Furthermore, six MHCII epitopes were predicted to be antigenic, non-allergic and non-toxic epitopes (Table 7). However, 983YITARDMYM991 and 982YYITARDMY990 epitopes which were also presented in MHCII prediction methods, showed high antigenicity, no allergenicity and no toxicity. These epitopes were interacted with 52 and 38 alleles in MHCII see Fig. 6.

Table 6: Antigenic, non-allergic and non-toxic MHCI epitopes

Peptide

Start

End

Antigenicity

Allele

ic50

YYITARDMY*

982

990

0.8845

HLA-A*29:02

14.52

HLA-A*30:02

160.94

HLA-C*14:02

27.32

YITARDMYM*

983

991

0.7901

HLA-A*02:01

233.08

HLA-A*02:06

212.86

HLA-C*03:03

29

HLA-C*06:02

200.39

HLA-C*07:01

267.22

HLA-C*14:02

49.52

HLA-C*15:02

77.63

TARDMYMPR*

985

993

0.6914

 

HLA-A*30:01

56.23

HLA-A*31:01

14.3

HLA-A*68:01

28.24

IIFILILGW

1105

1113

0.6749

 

HLA-B*57:01

78.45

HLA-B*58:01

64.27

KKSSYYTTF

1139

1147

1.1865

HLA-A*32:01

182.52

 

 

Table 7: Antigenic, non-allergic and non-toxic MHCII epitopes

Core Sequence

Antigenicity 

Peptide Sequence

Start

End

Allele

IC50

IIFILILGW

0.6914

IAFATIIFILILGWV

1100

1114

HLA-DRB1*15:01

454.6

KKSSYYTTF

0.6749

MSKCGKKSSYYTTFD

1134

1148

HLA-DPA1*01:03/DPB1*02:01

872.7

SKCGKKSSYYTTFD

1135

1149

HLA-DPA1*01/DPB1*04:01

408.1

HLA-DPA1*01:03/DPB1*02:01

301.5

HLA-DPA1*02:01/DPB1*05:01

953.4

KCGKKSSYYTTFDND

1136

1150

HLA-DPA1*01/DPB1*04:01

276.8

HLA-DPA1*02:01/DPB1*05:01

853.9

CGKKSSYYTTFDNDV

1137

1151

HLA-DPA1*02:01/DPB1*05:01

958.9

MSKCGKKSSYYTTFD

1134

1148

HLA-DPA1*01:03/DPB1*02:01

872.7

KSSYYTTFD

 

0.6466

MSKCGKKSSYYTTFD

1134

1148

HLA-DRB1*04:05

155

SKCGKKSSYYTTFDN

1135

1149

HLA-DRB1*04:05

125.6

KCGKKSSYYTTFDND

1136

1150

HLA-DRB1*04:05

92.2

CGKKSSYYTTFDNDV

1137

1151

HLA-DRB1*04:05

51.9

GKKSSYYTTFDNDVV

1138

1152

HLA-DRB1*04:05

46.9

KKSSYYTTFDNDVVT

1139

1153

HLA-DRB1*04:05

45.3

TARDMYMPR

 

0.7901

SYYITARDMYMPRAI

981

995

HLA-DRB1*03:01

269.3

YYITARDMYMPRAIT

982

996

HLA-DRB1*03:01

281.9

YITARDMYMPRAITA

983

997

HLA-DRB1*03:01

618.8

YITARDMYM

1.1865

QVNGSYYITARDMYM

 

977

 

991

 

HLA-DRB1*01:01

22

HLA-DRB1*04:01

145

HLA-DRB1*04:04

331.2

HLA-DRB1*07:01

20.3

HLA-DRB3*01:01

550.7

HLA-DRB5*01:01

227.8

VNGSYYITARDMYMP

 

978

 

992

 

HLA-DQA1*01:02/DQB1*06:02

338.6

HLA-DRB1*01:01

25.8

HLA-DRB1*03:01

447.6

HLA-DRB1*04:01

105.8

HLA-DRB1*04:04

248.3

HLA-DRB1*07:01

27.8

HLA-DRB1*15:01

380.6

HLA-DRB3*01:01

577.8

HLA-DRB5*01:01

198.6

NGSYYITARDMYMPR

979

993

HLA-DQA1*01:02/DQB1*06:02

393.3

HLA-DQA1*05:01/DQB1*03:01

817.3

HLA-DRB1*01:01

19.8

HLA-DRB1*03:01

176.5

HLA-DRB1*04:01

65.2

HLA-DRB1*04:04

225

HLA-DRB1*07:01

40.2

HLA-DRB1*15:01

291.2

HLA-DRB3*01:01

635

HLA-DRB5*01:01

93.5

GSYYITARDMYMPRA

980

994

HLA-DQA1*01:02/DQB1*06:02

218

HLA-DRB1*01:01

14

HLA-DRB1*03:01

197.3

HLA-DRB1*04:01

47.8

HLA-DRB1*04:04

242.4

HLA-DRB1*07:01

57.3

HLA-DRB1*15:01

288.6

HLA-DRB3*01:01

780.4

HLA-DRB5*01:01

61.4

SYYITARDMYMPRAI

981

995

HLA-DRB1*01:01

23.1

HLA-DRB1*04:01

65.3

HLA-DRB1*04:04

249.2

HLA-DRB1*04:05

356.4

HLA-DRB1*07:01

72.2

HLA-DRB1*15:01

284.7

HLA-DRB5*01:01

87.4

YYITARDMYMPRAIT

982

996

HLA-DRB1*01:01

40.8

HLA-DRB1*04:01

108.8

HLA-DRB1*04:04

269.1

HLA-DRB1*04:05

706.3

HLA-DRB1*07:01

160.6

HLA-DRB5*01:01

121.3

YITARDMYMPRAITA

983

997

HLA-DRB1*04:01

145.4

HLA-DRB1*04:04

652.4

HLA-DRB1*07:01

355.4

HLA-DRB1*08:02

955

HLA-DRB5*01:01

206.9

YYITARDMY

 

0.8845

IQVNGSYYITARDMY

976

990

HLA-DQA1*05:01/DQB1*02:01

491.6

HLA-DRB1*04:01

723.4

HLA-DRB1*04:04

819.7

HLA-DRB1*11:01

72

HLA-DRB1*11:01

72

QVNGSYYITARDMYM

977

991

HLA-DPA1*01/DPB1*04:01

710.8

HLA-DPA1*01:03/DPB1*02:01

875.8

HLA-DQA1*05:01/DQB1*02:01

292.7

HLA-DRB1*03:01

588

HLA-DRB1*11:01

32.4

HLA-DRB1*11:01

32.4

HLA-DPA1*01/DPB1*04:01

557.6

HLA-DPA1*01:03/DPB1*02:01

860.8

HLA-DQA1*05:01/DQB1*02:01

311.9

HLA-DRB1*11:01

17.9

HLA-DRB1*11:01

17.9

NGSYYITARDMYMPR

979

993

HLA-DPA1*01/DPB1*04:01

503

HLA-DPA1*01:03/DPB1*02:01

763.2

HLA-DQA1*05:01/DQB1*02:01

387.6

HLA-DRB1*09:01

858.7

HLA-DRB1*09:01

858.7

HLA-DRB1*11:01

11

HLA-DRB1*11:01

11

GSYYITARDMYMPRA

980

994

HLA-DPA1*01/DPB1*04:01

504.5

HLA-DPA1*01:03/DPB1*02:01

790.4

HLA-DQA1*05:01/DQB1*02:01

482.9

HLA-DRB1*11:01

15.2

HLA-DRB1*11:01

15.2

SYYITARDMYMPRAI

981

995

HLA-DPA1*01/DPB1*04:01

480

HLA-DPA1*01:03/DPB1*02:01

733.8

HLA-DQA1*05:01/DQB1*02:01

526.3

HLA-DRB1*11:01

26.8

HLA-DRB1*11:01

26.8

YYITARDMYMPRAIT

982

996

HLA-DPA1*01/DPB1*04:01

705.6

HLA-DPA1*01:03/DPB1*02:01

931.5

HLA-DQA1*05:01/DQB1*02:01

678.7

HLA-DRB1*11:01

51.8

HLA-DRB1*11:01

51.8

3.8 Molecular Docking  

The molecular docking was achieved by docking MHCI epitopes with chicken BF alleles (BF2 * 2101 & BF2 * 0401) using peptide-binding groove affinity. The chicken alleles were used as receptors, and the top MHCI epitopes 982YYITARDMY990, 983YITARDMYM991 and 985TARDMYMPR993 were used as ligands.  Docking of 983YITARDMYM991 epitope with BF2*2101 and BF2*0401 alleles showed – 72.11 and – 37.39 global energy respectively, indicating a strong binding affinity between the ligands and the receptors compared to other epitopes (Fig. 7, 8 and 9). In general, the global binding affinity of ligands with the receptor BF2*2101 alleles was found to be lower compared to BF2*0401, suggesting strong receptor-ligand interaction.  

Discussion

Epitopes capable of inducing immunity in both types (B-cell and T-cell) are considered to be strong candidates for the vaccine [46]. There are several potential benefits offered by peptide vaccine over traditional vaccines against organisms. Most importantly, it allows the immune response to focus only on relevant epitopes and avoid those leading to non-protective responses, immune evasion, or unwanted side effects, such as autoimmunity [47].  

IBV vaccination studies have always focused on humoral immune responses regarding protection. Acquired immunity results in the activation of antigen-specific effector mechanisms including B-cells (humoral), T-cells (cellular) and macrophages, and memory cells production [4]. Chickens develop a good humoral response to IBV infections, which measured by ELISA, virus neutralizing (VN) and haemagglutination-inhibition HI antibodies tests [48].

IBV glycoprotein S1 is known to be responsible for virus neutralization (VN) and haemagglutination-inhibition HI antibodies and has been considered the most likely protective inducer [4]. Multi-peptide vaccines using immunoinformatics tools have recently been conducted in Sudan for several viral diseases in chicken such as ILTV, fowlpox, Newcastle and marek's disease virus [15, 49-51].

In the present study, IBV spike protein was analyzed using various prediction servers. Protein characterization of IBV spike S protein using protparam confirmed its positive in nature and stable. The protein also exhibited good antigenic properties using Vaxijen 2.0v server.

Corona-S1 and Corona S2 have been identified as major conserved domains in the IBV spike glycoprotein refseq. Conserved Domain (CDD) BLAST search revealed that corona-S1 (pfam01600) is the only member of the superfamily cl03276 and corona-S2 domain (pfam01601) is the only member of the superfamily cl20218. The main related sequences in both domains were Feline infectious peritonitis virus (strain 79-1146), Avian infectious bronchitis virus (strain Beaudette), and Human coronavirus 229E. However, Severe acute respiratory syndrome-related  coronavirus sequences was only associated with corona-S2 domain [52].

Prediction of B-cell epitopes is essential for the design vaccine components and immuno-diagnostic reagents. B-cell antigenic epitopes are either continuous or discontinuous in nature.

Most epitope prediction methods are based on continuous epitopes [53]. It has been reported that linear B cell epitopes play a role in virus neutralization [11]. IEDB prediction tool was used to predict linear, surface and antigenic epitopes based on the properties of amino acids such as hydrophilicity, surface accessibility, flexibility, and antigenicity [15]. 

In this study, seven shortened conserved epitopes (1139KKSSYY1144, 1140KSSYYT1145, 1141SSYYTT1146, 1141SSYYT1145, 1142SYYTT1146, 1142SYYT1145, and 1143YYTT1146) were predicted from B cell prediction methods as surface, linear and antigenic epitopes. The predicted epitopes were adjacent to each other from the position 1139 – 1146.   In a similar study, only one epitope (YTSNETTDVTS175-185) was predicted within the S1 glycoprotein of M41 IBV strains using BepiPred epitope prediction server version 1, and three such epitopes (VSNASPNSGGVD279-290, HPKCNFRPENI328-338, NETNNAGSVSDCTAGT54-69 ) were predicted in CR88 IBV strains [11].

The majority of B-cell epitopes are conformational (around a 90%) and only a minority of native antigens have linear B-cell epitopes [54]. Discotope server has been used for predicting discontinuous

Epitopes from the 3D structure of the spike IBV reference sequence. Around 30 discontinuous epitopes with a specificity of 90 % were recognized at different exposed surface areas. These epitopes have a significant advantage in identifying the native well-structured protein Ag [55].

Cytotoxic T lymphocytes (CTL) provide a critical arm of the immune system in eliminating autologous cells expressing foreign antigen. Unlike humoral immunity, the specificity of CTL activation depends on membrane receptors rather than secreted molecules, and antigen receptors of CTL interact with peptide determinants only in association with matched major histocompatibility complex (MHC) molecules. Virus-specific CTL have been shown to be important, if not critical, for resolution of infection and elimination of viral shedding [1].

It is stated that, the major histocompatibility complex MHC restricted CTL response can be associated with decreases in viral load, and CD8+ lymphocytes were mostly responsible for the observed protection [1, 56]. Responses to infectious bronchitis virus (IBV) with cytotoxic T-lymphocyte (CTL) were calculated at regular intervals between 3 and 30 days post infection [1].

However, MHCI prediction methods showed three conserved CTL epitopes 985TARDMYMPR993, 983YITARDMYM991 and 982YYITARDMY990 as they linked to 7 and 3 human MHCI alleles respectively and showed high antigenicity, no allergenicity and no toxicity. Recent studies showed that vigorous cytotoxic T lymphocyte (CTL) responses that correlate with initial decrease in infection and illness can be detected after IBV infection. It has been identified that the CD8+ T cells were exhausted without CD4+ helper T cells.  CD4+ T cells do not seem important in the initial resolution of IBV infection in chickens [56].

In MHCII prediction method, several core peptides were predicted to interact with MHCII alleles, but surprisingly the top core peptides were also 983YITARDMYM991 and 982YYITARDMY990 which were presented in MHCI prediction methods. They linked with 52 and 38 human alleles respectively. These epitopes showed high antigenicity, no allergincity and no toxicity.

Molecular docking was performed to display the interaction between BF alleles (BF2*2101 & BF2*0401) and MHCI epitopes (982YYITARDMY990, 983YITARDMYM991 and 985TARDMYMPR993). The 3D structures of MHC I epitopes were designed using PEPFOLD and docked with BF alleles via Patchdock server. Docking the epitope 983YITARDMYM991 with both BF2 alleles produced strong binding affinity (- 72.11 and -37.97 global energy respectively) followed by 982YYITARDMY990 (- 64.68 and – 37.57 global energy respectively). This indicates the strong interaction between the ligand and the receptor compared to other epitopes (see Fig. 5, 6 and 7).

Ligands' interaction with the receptor BF2*2101 alleles was found to be better compared with BF2 * 0401. However for both BF alleles, the docked molecules showed different groove binding site.  Future studies should test the predicted epitopes for therapeutic potency to prove their safety and effectiveness.

Conclusion

In this study, five epitopes were predicted from spike glycoprotein of IBV as the best B cell (1139KKSSYY1144, 1140KSSYYT1145 and 1141SSYYT1145) and T cell epitopes (982YYITARDMY990 and 983YITARDMYM991). They showed high antigenicity, no allergenicity and no toxicity as well as great linkage of MHC epitopes with their alleles. The suggested epitopes should be designed, incorporated and tested as multi-epitopes vaccine against IBV. This vaccine may serve as a possible peptide vaccine to control IBV infection in chicken by inducing humoral and cellular responses. 

Peptide vaccination against IBV spike protein (S) can strongly replace traditional vaccines as it is designed to cover all strains in different serotypes, which can reduce recurring outbreaks and their associated massive economic losses.

Abbreviations

IB: Infectious bronchitis;  IBV: Infectious Bronchitis Virus; IEDB: Immune Epitope Database; S: spike; MHC: major histocompatibility complex; BF: The genetic polymorphism of properdin factor B; refseq: reference sequence; NCBI: National Central Biotechnology Information; MSA: multiple sequence alignment; GRAVY; Grand average of hydropathicity; CDD: Conserved Domain Database; IC50: The half maximal inhibitory concentration; ANN: artificial neural networks; NN-align: artificial neural network-based alignment; HLA: The human leukocyte antigen; CTL: Cytotoxic T lymphocytes.   

Declarations

Acknowledgements

Not applicable.

Authors’ contributions

Eman, A. Awadelkareem and Sumaia A. Ali designed this study, accomplished the experiments and analyze the results. Sumaia A. Ali interpreted the data and wrote the manuscript. The authors read and approved the final manuscript. 

Funding

Not applicable. 

Availability of data and materials

All the data supporting the findings are contained within the manuscript 

Ethics approval and consent to participate

Not applicable. 

Consent for publication

Not applicable. 

Competing interests

The authors declare that they have no competing interests.

All manuscripts must contain the following sections under the heading 'Declarations':

Authors' details

1Faculty of Veterinary Medicine, University of Khartoum, Khartoum, Sudan. 2Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, Sudan University of Science and Technology, Khartoum, Sudan.

References

  1. Seo SH, Collisson EW. Specific cytotoxic T lymphocytes are involved in in vivo clearance of infectious bronchitis virus. Journal of Virology. 1997;71(7):5173-7.
  2. Cavanagh D. Coronavirus avian infectious bronchitis virus. Veterinary Research. 2007;38(2):281-97.
  3. Ignjatovic J, Sapats S. Avian infectious bronchitis virus. Revue Scientifique et Technique-Office International des Epizooties. 2000;19(2):493-501.
  4. Raj GD, Jones R. Infectious bronchitis virus: immunopathogenesis of infection in the chicken. Avian Pathology. 1997;26(4):677-706.
  5. Yang T, Wang H-N, Wang X, Tang J-N, Lu D, Zhang Y-F, et al. The protective immune response against infectious bronchitis virus induced by multi-epitope based peptide vaccines. Bioscience, Biotechnology, and Biochemistry. 2009: 0864-1–5,
  6. Abro SH, Ullman K, Belák S, Baule C. Bioinformatics and evolutionary insight on the spike glycoprotein gene of QX-like and Massachusetts strains of infectious bronchitis virus. Virology Journal. 2012;9(1):211.
  7. Abdel-Moneim A, Madbouly H, Gelb J, Ladman B. Isolation and identification of Egypt/Beni-Seuf/01 a novel genotype of infectious bronchitis virus. Veterinary Medical Journal-GIZA. 2002;50(4):1065-78.
  8. Mo M-L, Hong S-M, Kwon H-J, Kim I-H, Song C-S, Kim J-H. Genetic diversity of spike, 3a, 3b and e genes of infectious bronchitis viruses and emergence of new recombinants in Korea. Viruses. 2013;5(2):550-567.
  9. Miller MM, Taylor Jr RL. Brief review of the chicken major histocompatibility complex: the genes, their distribution on chromosome 16, and their contributions to disease resistance. Poultry Science. 2016; 1;95(2):375-92.
  10. Shina T, Hosomichi K, Hanzawa K. Comparative genomics of the poultry major histocompatibility complex. Animal Science Journal. 2006 Apr;77(2):151-62.
  11. Bande F, Arshad SS, Hair Bejo M, Kadkhodaei S, Omar AR. Prediction and in silico identification of novel B-cells and T-cells epitopes in the S1-spike glycoprotein of M41 and CR88 (793/B) infectious bronchitis virus serotypes for application in peptide vaccines. Advances in bioinformatics. 2016;2016.
  12. Dash R, Das R, Junaid M, Akash MF, Islam A, Hosen SZ. In silico-based vaccine design against Ebola virus glycoprotein. Advances and Applications in Bioinformatics and Chemistry: AABC. 2017;10:11.
  13. Gaafar B, Ali SA, Abd-elrahman KA, Almofti YA. Immunoinformatics Approach for Multiepitope Vaccine Prediction from H, M, F, and N Proteins of Peste des Petits Ruminants Virus. Journal of Immunology Research. 2019;2019.
  14. Zheng J, Lin X, Wang X, Zheng L, Lan S, Jin S, Ou Z, Wu J. In silico analysis of epitope-based vaccine candidates against hepatitis B virus polymerase protein. Viruses. 2017;9(5):112.
  15. Ali SA, Almofti YA, Abd-elrahman KA. Immunoinformatics approach for multiepitopes vaccine prediction against glycoprotein B of avian infectious laryngotracheitis virus. Advances in Bioinformatics. 2019;2019.
  16. Gasteiger E GA, Hoogland C, Ivanyi I, Appel RD, Bairoch A. ExPASy: the proteomics server for in-depth protein knowledge and analysis. Nucleic Acids Research 2003;31:3784–8.
  17. Buchan DW MF, Nugent TC, Bryson K, Jones DT. . Scalable web services for the PSIPRED Protein Analysis Workbench. Nucleic Acids Research. 2013;41:W349–57.
  18. Ferre and P. Clote. Disulfide connectivity prediction using secondary structure information and diresidue frequencies. Bioinformatics. 2005;;21(10):2336-46.
  19. Altschul SF, Madden TL, Schäffer AA, Zhang J, Zhang Z, Miller W, et al. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Research. 1997;25(17):3389-402.
  20. Marchler-Bauer A, Anderson JB, Derbyshire MK, DeWeese-Scott C, Gonzales NR, Gwadz M, Hao L, He S, Hurwitz DI, Jackson JD, Ke Z. CDD: a conserved domain database for interactive domain family analysis. Nucleic Acids Research. 2007;35:D237-40.
  21. Eddy, S.R., Profile hidden Markov models. Bioinformatics (Oxford, England), 1998. 14(9):755-763.
  22. El-Gebali, S., et al., The Pfam protein families database in 2019. Nucleic Acids Research, 2019. 47(D1):D427-D432.
  23. Waterhouse AM, Procter JB, Martin DM, Clamp M, Barton GJ. Jalview Version 2—a multiple sequence alignment editor and analysis workbench. Bioinformatics. 2009;25(9):1189-91.
  24. Bui H-H, Sidney J, Li W, Fusseder N, Sette A. Development of an epitope conservancy analysis tool to facilitate the design of epitope-based diagnostics and vaccines. BMC Bioinformatics. 2007;8(1):361.
  25. Kumar S, Stecher G, Tamura K. MEGA7: Molecular evolutionary genetics analysis version 7.0 for bigger datasets. Molecular Biology and Evolution. 2016;33(7):1870-4.
  26. Vita R, Overton JA, Greenbaum JA, Ponomarenko J, Clark JD, Cantrell JR, et al. The immune epitope database (IEDB) 3.0. Nucleic Acids Research. 2014;43(D1):D405-D12.
  27. Larsen JE, Lund O, Nielsen M. Improved method for predicting linear B-cell epitopes. Immunome Research. 2006;2(1):2.
  28. Emini EA, Hughes JV, Perlow D, Boger J. Induction of hepatitis A virus-neutralizing antibody by a virus-specific synthetic peptide. Journal of Virology. 1985;55(3):836-9.
  29. Kolaskar A, Tongaonkar PC. A semi-empirical method for prediction of antigenic determinants on protein antigens. FEBS Letters. 1990;276(1-2):172-4.
  30. Sun P, Ju H, Liu Z, Ning Q, Zhang J, Zhao X, et al. Bioinformatics resources and tools for conformational B-cell epitope prediction. Computational and Mathematical Methods in Medicine. 2013;2013.
  31. Chan WM, Rogers SE, Nash SM, Buning PG, Meakin R. User’s manual for Chimera grid tools, version 1.8. NASA Ames Research Center, URL: http://people nas nasa gov/~ rogers/cgt/doc/man html. 2003.
  32. Patronov A, Doytchinova I. T-cell epitope vaccine design by immunoinformatics. Open Biology. 2013;3(1):120139.
  33. Abdelbagi M, Hassan T, Shihabeldin M, Bashir S, Ahmed E. Immunoinformatics Prediction of Peptide-Based Vaccine Against African Horse Sickness Virus. Immunome Research. 2017;13(135):2.
  34. Nielsen M, Lund O. NN-align. An artificial neural network-based alignment algorithm for MHC class II peptide binding prediction. BMC bioinformatics. 2009;10(1):296.
  35. Doytchinova IA, Flower DR. VaxiJen: a server for prediction of protective antigens, tumour antigens and subunit vaccines. BMC Bioinformatics. 2007;8(1): 1-7.
  36. Dimitrov I, Bangov I, Flower DR, Doytchinova I. AllerTOP v. 2—a server for in silico prediction of allergens. Journal of Molecular Modeling. 2014;20(6):2278. 1-6.
  37. Gupta S, Kapoor P, Chaudhary K, Gautam A, Kumar R, Raghava GP, et al. In silico approach for predicting toxicity of peptides and proteins. PloS One. 2013;8(9). 1-10.
  38. Källberg M, Wang H, Wang S, Peng J, Wang Z, Lu H, et al. Template-based protein structure modeling using the RaptorX web server. Nature Protocols. 2012;7(8):1511-1522.
  39. Peng J, Xu J. RaptorX: exploiting structure information for protein alignment by statistical inference. Proteins: Structure, Function, and Bioinformatics. 2011;79(S10):161-71.
  40. Peng J, Xu J. A multiple‐template approach to protein threading. Proteins: Structure, Function, and Bioinformatics. 2011;79(6):1930-9.
  41. Maupetit J, Derreumaux P, Tufféry P. A fast method for large‐scale De Novo peptide and miniprotein structure prediction. Journal of Computational Chemistry. 2010;31(4):726-738.
  42. Beaufays J, Lins L, Thomas A, Brasseur R. In silico predictions of 3D structures of linear and cyclic peptides with natural and non‐proteinogenic residues. Journal of Peptide Science. 2012;18(1):17-24.
  43. Shen Y, Maupetit J, Derreumaux P, Tufféry P. Improved PEP-FOLD approach for peptide and miniprotein structure prediction. Journal of Chemical Theory and Computation. 2014;10(10):4745-58.
  44. Andrusier N, Nussinov R, Wolfson HJ. FireDock: fast interaction refinement in molecular docking. Proteins: Structure, Function, and Bioinformatics. 2007;69(1):139-59.
  45. Shang J, Zheng Y, Yang Y, Liu C, Geng Q, Luo C, et al. Cryo-EM structure of infectious bronchitis coronavirus spike protein reveals structural and functional evolution of coronavirus spike proteins. PLoS Pathogens. 2018;14(4):1-9.
  46. Verma SK, Yadav S, Kumar A. In silico prediction of B-and T-cell epitope on Lassa virus proteins for peptide based subunit vaccine design. Advanced Biomedical Research. 2015; 4. 201.
  47. Reche PA, Fernandez-Caldas E, Flower DR, Fridkis-Hareli M, Hoshino Y. Peptide-based immunotherapeutics and vaccines. Journal of Immunology Research. 2014; 2014.
  48. Sylvester SA, Dhama K, Kataria J, Rahul S, Mahendran M. Avian infectious bronchitis: A review. Indian Journal of Comparative Microbiology Immunology and Infectious Diseases. 2005;26:1-14.
  49. Idris S, Salih S, Basheir M, Elhadi A, Kamel S, Abd-elrahman K, et al. In silico Prediction of Peptide based Vaccine against Fowlpox Virus (FPV). Immunome Research. 2018;14(2):1-11.
  50. Badawi MM, Fadl Alla A, Alam SS, Mohamed WA, Osman D, Alrazig Ali S, et al. Immunoinformatics predication and in silico modeling of epitope-based peptide vaccine against virulent Newcastle disease viruses. American Journal of Infectious Diseases and Microbiology. 2016;4(3):61-71.
  51. Bashir S, Abd-elrahman KA, Hassan MA, Almofti YA. Multi Epitope Based Peptide Vaccine against Marek’s Disease Virus Serotype 1 Glycoprotein H and B. American Journal of Microbiological Research. 2018;6(4):124-39.
  52. J. Cloning and sequencing of the gene encoding the spike protein of the coronavirus IBV. Journal of General Virology. 1985; 66:719-26.
  53. Yao B, Zheng D, Liang S, Zhang C. Conformational B-cell epitope prediction on antigen protein structures: a review of current algorithms and comparison with common binding site prediction methods. PloS One. 2013;8(4). 1-4
  54. Sanchez-Trincado JL, Gomez-Perosanz M, Reche PA. Fundamentals and methods for T-and B-cell epitope prediction. Journal of Immunology Research. 2017;2017. 1-14.
  55. Demolombe V, de Brevern AG, Felicori L, NGuyen C, de Avila RAM, Valera L, et al. PEPOP 2.0: new approaches to mimic non-continuous epitopes. BMC Bioinformatics 20. 2019:387. 1-14.
  56. Pei J, Briles WE, Collisson EW. Memory T cells protect chicks from acute infectious bronchitis virus infection. Virology. 2003;306 (2):376-84.