160
Expression and Characterization of Infectious Bursal Disease Virus Protein for Poultry Vaccine Development and Application in Nanotechnology Von der Fakultät für Mathematik, Informatik und Naturwissenschaften der RWTH Aachen University zur Erlangung des akademischen Grades eines Doktors der Naturwissenschaften genehmigte Dissertation vorgelegt von Omid Taghavian, M.Sc. aus Teheran, Iran Berichter: Universitätsprofessor Dr. Rainer Fischer Honorarprofessor Dr. Stefan Schillberg Tag der mündlichen Prüfung: 11. November 2013 Diese Dissertation ist auf den Internetseiten der Hochschulbibliothek online verfügbar.

Expression and Characterization of Infectious Bursal

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Expression and Characterization of Infectious Bursal

Expression and Characterization of Infectious Bursal Disease Virus Protein for Poultry Vaccine Development

and Application in Nanotechnology

Von der Fakultät für Mathematik, Informatik und Naturwissenschaften der RWTH Aachen University zur Erlangung des akademischen Grades eines Doktors der Naturwissenschaften

genehmigte Dissertation

vorgelegt von

Omid Taghavian, M.Sc.

aus Teheran, Iran

Berichter: Universitätsprofessor Dr. Rainer Fischer Honorarprofessor Dr. Stefan Schillberg

Tag der mündlichen Prüfung: 11. November 2013

Diese Dissertation ist auf den Internetseiten der Hochschulbibliothek online verfügbar.

Page 2: Expression and Characterization of Infectious Bursal

To my adorable parents and my wonderful wife for their endless love, support and

encouragement

Page 3: Expression and Characterization of Infectious Bursal

Table of content

I Introduction ............................................................................................................................................................ 1

I.1 Recombinant vaccines _________________________________________________________________ 1 DNA-based vaccine ................................................................................................................................... 1 I.1.1 Antigen-based vaccine (subunit vaccine) .................................................................................................. 2 I.1.2

I.2 Vaccine administration ________________________________________________________________ 4 Parenteral immunization ............................................................................................................................ 4 I.2.1 Mucosal immunization .............................................................................................................................. 4 I.2.2

Challenges in mucosal immunization .............................................................................................. 7 I.2.2.1 Oral tolerance ................................................................................................................................... 9 I.2.2.2

I.3 The chicken immune system ____________________________________________________________ 9 Chicken diseases and vaccination ............................................................................................................ 10 I.3.1

I.4 Infectious bursal disease virus (IBDV) ___________________________________________________ 11 Virus classification .................................................................................................................................. 11 I.4.1 Route of infection and pathology............................................................................................................. 11 I.4.2 Virus detection, classification and diagnostic tests .................................................................................. 12 I.4.3 Treatment and prevention ........................................................................................................................ 12 I.4.4 IBDV structure ........................................................................................................................................ 12 I.4.5 Structure and characteristics of the viral antigen IBD-VP2 ..................................................................... 14 I.4.6

I.5 Vaccine development against IBDV _____________________________________________________ 17

I.6 Application of VLPs and SVPs in nanotechnology _________________________________________ 20

I.7 Aim of this thesis ____________________________________________________________________ 21

II Materials and Methods ........................................................................................................................................ 23

II.1 Materials ___________________________________________________________________________ 23 Chemicals and consumables .................................................................................................................... 23 II.1.1 Buffers, media and solutions ................................................................................................................... 23 II.1.2 Matrices and membranes ......................................................................................................................... 23 II.1.3 Enzymes and reaction kits ....................................................................................................................... 23 II.1.4 Antibodies and substrates ........................................................................................................................ 24 II.1.5 Special chemicals .................................................................................................................................... 24 II.1.6 Oligonucleotides ...................................................................................................................................... 24 II.1.7 Vectors ..................................................................................................................................................... 26 II.1.8 Bacterial, yeast and virus strains ............................................................................................................. 26 II.1.9 Plants and animals ................................................................................................................................... 27 II.1.10 Equipment and applications ..................................................................................................................... 27 II.1.11

II.2 Methods ___________________________________________________________________________ 28 Recombinant DNA technologies ............................................................................................................. 28 II.2.1

Preparation of heat-shock competent E. coli .................................................................................. 28 II.2.1.1 Transformation of E. coli by heat-shock ........................................................................................ 29 II.2.1.2 Preparation of electrocompetent E. coli ......................................................................................... 29 II.2.1.3 Transformation of E. coli by electroporation ................................................................................. 29 II.2.1.4 Preparation of electrocompetent A. tumefaciens ............................................................................ 29 II.2.1.5 Transformation of A. tumefaciens by electroporation .................................................................... 30 II.2.1.6 Determination of the transformation efficiency of competent bacteria .......................................... 30 II.2.1.7 Transformation of P. pastoris by electroporation .......................................................................... 30 II.2.1.8 Culturing of E. coli and glycerol stock preparation ....................................................................... 31 II.2.1.9

Culturing of recombinant A. tumefaciens and glycerol stock preparation ...................................... 32 II.2.1.10 Culturing of recombinant P. pastoris and glycerol stock preparation ............................................ 32 II.2.1.11 Isolation of plasmid from recombinant E. coli ............................................................................... 32 II.2.1.12 Analytical agarose gel electrophoresis of DNA ............................................................................. 32 II.2.1.13 Preparative agarose gel electrophoresis of DNA............................................................................ 33 II.2.1.14 Restriction digest of DNA and preparative agarose gel electrophoresis ........................................ 33 II.2.1.15 Quantification of DNA ................................................................................................................... 33 II.2.1.16 Ligation of the DNA fragments ..................................................................................................... 33 II.2.1.17 PCR amplification .......................................................................................................................... 33 II.2.1.18 DNA sequencing ............................................................................................................................ 35 II.2.1.19

Page 4: Expression and Characterization of Infectious Bursal

Table of content

Expression and purification of recombinant protein ................................................................................35 II.2.2 Growth and maintenance of tobacco plants....................................................................................35 II.2.2.1 Preparation of recombinant A. tumefaciens ....................................................................................35 II.2.2.2 Vacuum infiltration and/or injection of tobacco leaves with recombinant agrobacteria ................36 II.2.2.3 Recombinant Agrobacterium-mediated stable transformation of Nicotiana tabacum ...................37 II.2.2.4 Expression and purification of IBD-VP2 from tobacco leaves ......................................................38 II.2.2.5 Expression and purification of IBD-VP2 from P. pastoris ............................................................39 II.2.2.6

Protein analysis ........................................................................................................................................40 II.2.3 Quantification of proteins...............................................................................................................40 II.2.3.1 SDS-PAA gel electrophoresis and Coomassie brilliant blue staining ............................................41 II.2.3.2 Immunoblot analysis ......................................................................................................................41 II.2.3.3 Dot blot analysis .............................................................................................................................42 II.2.3.4 Size exclusion chromatography (SEC) ...........................................................................................42 II.2.3.5 Immunogold labelling and transmission electron microscopy (TEM) ...........................................43 II.2.3.6

Glycan analysis ........................................................................................................................................43 II.2.4 Immunoblot ....................................................................................................................................43 II.2.4.1 Enzymatic deglycosylation ............................................................................................................43 II.2.4.2 Glyco-proteomics analysis .............................................................................................................43 II.2.4.3

Immunization of mice ..............................................................................................................................44 II.2.5 Determination of antibody titer ......................................................................................................44 II.2.5.1

Immunization of chickens ........................................................................................................................44 II.2.6 Vaccine component preparation .....................................................................................................44 II.2.6.1 Immunization plan .........................................................................................................................45 II.2.6.2 Health status monitoring ................................................................................................................45 II.2.6.3 Antibody titer determination ..........................................................................................................46 II.2.6.4 IBDV antibody test kit ...................................................................................................................47 II.2.6.5 Detection of IBDV in bursa tissue .................................................................................................47 II.2.6.6 Histological bursa lesion ................................................................................................................47 II.2.6.7 Statistical analysis ..........................................................................................................................48 II.2.6.8

Bioconjugation analysis ...........................................................................................................................48 II.2.7 Stability analysis of IBD-sub viral particles (IBD-SVPs) ..............................................................48 II.2.7.1 Native agarose gel electrophoresis (NAGE) ..................................................................................48 II.2.7.2 Spectroscopy ..................................................................................................................................48 II.2.7.3 Dynamic light scattering (DLS) .....................................................................................................48 II.2.7.4 Chemical bioconjugation................................................................................................................49 II.2.7.5

III Results ...................................................................................................................................................................50

III.1 Expression and characterization of IBD-VP2 in N. tabacum _________________________________ 50 Cloning of IBD-VP2 gene sequence into pTRAkc vector for generation of tobacco expression vectors 50 III.1.1 Transient expression of IBD-VP2 in tobacco leaves ...............................................................................52 III.1.2

Figure III.3 .....................................................................................................................................................................54 Generation and screening of transgenic tobacco plants expressing IBD-VP2 .........................................54 III.1.3 Purification of IBD-VP2 from tobacco leaves .........................................................................................54 III.1.4 Glycosylation analysis of IBD-VP2apo ...................................................................................................56 III.1.5

Immunoblot analysis ......................................................................................................................56 III.1.5.1 Enzymatic deglycosylation of VP2apo ..........................................................................................57 III.1.5.2 Glyco-proteomics analysis .............................................................................................................58 III.1.5.1

Analysis of plant-produced VP2 assembly ..............................................................................................60 III.1.6 Immunogenicity of tobacco-produced VP2 in mice ................................................................................63 III.1.7

III.2 Expression and characterization of IBD-VP2 in P. pastoris __________________________________ 64 Cloning of IBD-VP2 gene sequence into pPICZ_B vector for generation of Pichia expression vectors 64 III.2.1 Pichia transformation, screening and expression .....................................................................................65 III.2.2 Expression and purification of IBD-VP2 and analysis of SVP assembly ................................................68 III.2.3 Glycosylation analysis of yVP2sec ..........................................................................................................72 III.2.4

Lectin blot ......................................................................................................................................73 III.2.4.1 Enzymatic deglycosylation of yVP2sec-m .....................................................................................73 III.2.4.2 Glyco-proteomic analysis of yVP2sec ...........................................................................................74 III.2.4.3

III.3 Chicken immunization with yVP2cyto __________________________________________________ 76 Health monitoring, clinical signs .............................................................................................................78 III.3.1

Page 5: Expression and Characterization of Infectious Bursal

Table of content

Immune response and protection analysis ............................................................................................... 80 III.3.2

III.4 IBD-SVPcyto as a nanoplatform _______________________________________________________ 87 Solvent stability of IBD-SVPcyto............................................................................................................ 87 III.4.1 Temperature stability of IBD-SVPcyto ................................................................................................... 88 III.4.2 Stability of IBD-SVPcyto in different pH environment .......................................................................... 89 III.4.3 Addressability of IBD-SVPcyto .............................................................................................................. 90 III.4.4

Addressability of amine groups...................................................................................................... 91 III.4.4.1 Addressability of carboxyl groups ................................................................................................. 92 III.4.4.2

IV Discussion ............................................................................................................................................................. 93

IV.1 Characterization of tobacco-produced IBD-VP2 __________________________________________ 94

IV.2 Characterization of Pichia-produced IBD-VP2 ___________________________________________ 97

IV.3 Vaccine preparation _________________________________________________________________ 98

IV.4 Induction of immune response against IBDV by applying Pichia-produced IBD-SVPcyto administered either orally or parenterally _____________________________________________________ 100

IV.5 IBD-SVPcyto as a potential platform for nanobiotechnology _______________________________ 105

IV.6 Conclusion and future prospects ______________________________________________________ 107

V Summary ............................................................................................................................................................ 109

VI Zusammenfassung ............................................................................................................................................. 111

VII References ........................................................................................................................................................... 113

VIII Appendices .......................................................................................................................................................... 135

VIII.1 Abbreviations ______________________________________________________________________ 135

VIII.2 Schematic presentation of PCR & cloning procedure for generation of tobacco expression vectors 138

VIII.3 Schematic presentation of PCR and cloning procedure for generation of Pichia expression vectors 141

VIII.4 Protein sequence alignment of IBD-VP2 ________________________________________________ 143

VIII.5 Monitoring of IBDV clinical signs among the vaccinated chickens after viral challenge._________ 144

VIII.6 Serological VP2 antibody titers _______________________________________________________ 146

VIII.7 Figure legends _____________________________________________________________________ 148

VIII.8 Tables ____________________________________________________________________________ 150

Page 6: Expression and Characterization of Infectious Bursal
Page 7: Expression and Characterization of Infectious Bursal

Chapter I Introduction

1

I Introduction I.1 Recombinant vaccines

Since Edward Jenner’s time (1798), comprehensive vaccination programs have prodigiously influenced human and animal health, saved millions of lives and billions of dollars worldwide. Immunization is the most effective, convenient and economical method for prevention of many diseases especially contagious infections. Conventional vaccines consist of attenuated or killed viruses, bacteria or inactivated bacterial toxins (toxoid), which are mostly administered parenterally. Although these types of vaccines are very effective, their production and administration are costly. Additionally, application may accompanied by side effects such as reversion of virulence due to imperfect attenuation or inactivation or even recombination or mutagenesis that could lead to generation of pathogens with higher contagious strength during production. Using attenuated or killed pathogens in vaccines eliminates the need to identify distinct protective antigens. Still safety and efficacy raise concerns. This could be alleviated by identification of protective antigens. By using the protective antigens, the safety of a vaccine combines with the advantages of focusing the immune response on an optimal target. Development in purification techniques refined previous production methods and provided large-scale manufacturing of extremely pure antigens. This led to the development of “purified subunit vaccines”. In this vaccination method, only the antigenic part of the pathogen is being extracted and, with or without modification, applied as vaccine. Hereby many previous hazardous contaminants are prevented (Ulmer et al., 2006). The emerging recombinant technologies provided a very safe and economical way for large-scale production of subunit vaccines (Farchaus et al., 1998; Ribot et al., 2006). The technology facilitates identification, extraction, amplification and modification of DNA/RNA fragments encoding the antigenic part of a pathogen for recombinant expression. To conquer a disease with a subunit vaccine, the first step is the identification of a suitable antigen with the ability to elicit a protective, long lasting immune response. The new generation of vaccines exploiting recombinant DNA technology is being called recombinant vaccines. They can be divided into two major groups: DNA and antigen-based vaccines.

DNA-based vaccine I.1.1The use of DNA as vaccine is a new and promising approach in vaccination since it is safe (no infectious antigen is involved), specific, easy and economical. The term refers to induction of immune response against a protein(s) expressed in vivo (vaccine recipient) subsequent to the injection of plasmid DNA, therefore mimicking the live pathogen in immune system induction. The plasmid DNA is carrying an antigen-coding sequence under a host-specific promoter. This method is attractive due to its simplicity. It only requires plasmid construction, proliferation and extraction (Dertzbaugh, 1998). DNA vaccines are able to induce both humoral and cellular immunity (Chang et al., 2001). Cytotoxic T lymphocyte (CTL) response is usually efficiently induced when the antigen is

Page 8: Expression and Characterization of Infectious Bursal

Chapter I Introduction

2

expressed by the host cells (Ulmer et al., 2006). Especially when the antigen is expressed in antigen-presenting cells (APCs), it most likely leads to major histocompatibility complex (MHC) class II as well as class I presentation (Condon et al., 1996; Casares et al., 1997). It was shown that DNA vaccines also have the potential to induce a potent systemic immune response (Donnelly et al., 1997; Davis and McCluskie, 1999). DNA vaccines are mostly administered parenterally by intramuscular or interadermal injection (named “naked DNA”). So far, they show moderate immune response stimulation in human but two veterinary vaccines, one for horse and one for farm-raised salmon, have already been licensed (Brun et al., 2011). Therefore, an optimal delivery strategy should be found to increase DNA vaccine potency (Ulmer et al., 2006). To induce mucosal immunity mucosal administration of DNA vaccines has also been accomplished (McCluskie and Davis, 1999). The main drawbacks of DNA vaccines are the need for large amounts of DNA to achieve protective immune response in large animals and human and the slow development of a strong immunity (which limits the application in disease outbreaks where quick control in required). Last but not least, DNA vaccines can trigger disease exacerbation instead of protection if a non-proper antigen and administration pathway is chosen (Brun et al., 2011). Improvement in immune response induction has been tried by realizing enhanced plasmid uptake by exploring various administration routes such as electroporation or gene-gun, delivery of vaccines in association with cytokines (Littel-van den Hurk et al., 2004) and by co-delivery of DNA vaccines, adjuvant and cytokines to exploit different local subsets of antigen presenting cells (APCs) (McKay et al., 2004). Although the risk of DNA integration into the host genome is one of the main concerns, it was shown that the frequency is less than the frequency of spontaneous mutation (Gravier et al., 2007; Donnelly et al., 2005). Alternatively, live viral vectors have been introduced instead of plasmid DNA. These vectors have been derived from adenovirus, poxviruses or alpha viruses, which have been investigated so far for carrying the gene of interest for vaccine application. Although manufacturing viral vector-based vaccines are more complicated, they are more immunogenic than naked DNA vaccines (McConkey et al., 2003). Vector design, genetic stability, producer cell lines, virus propagation and purifications and product characterization are the main challenges to ensure the safety and quality of vector-based vaccines.

Antigen-based vaccine (subunit vaccine) I.1.2Known protective antigens are expressed recombinantly in a heterologous expression system and formulated for further vaccine application. Using a specific protective antigen guaranties the induction of specific and focused immune response. Such antigen-based or subunit vaccines raise less concern than conventional vaccination methods since administration of live or killed organisms to the recipient is not required. Also, recipients are not transiently transformed with foreign genetic materials as is essential in DNA vaccination. Moreover, the chemical composition and the processes used for production of subunit vaccines should be thoroughly defined. Using well-characterized organisms as expression hosts may reduce the risk of contamination of the target antigen with harmful agents. However, most of the recombinant systems have certain limitations in synthesizing the antigen in proper immunogenic form. With respect to post-translational modifications, lack of glycosylation in prokaryotic organisms and hyperglacosylation

Page 9: Expression and Characterization of Infectious Bursal

Chapter I Introduction

3

in yeasts are the major concerns. Other issues in developing subunit vaccines are related to the high cost for production and downstream processing based on the need for expensive fermentation systems, manufacturing, purification and the procedure required to adjust the dosage. Thus, finding new expression systems with high expression level, easy purification procedure is crucial in reducing the costs. In contrast to live attenuated pathogens which attack and replicate in the host body in natural manner and normally elicit strong immune responses, recombinant subunit vaccines lack this characteristic and are inherently less immunogenic. To enhance immunogenicity to a protective level it is necessary to adapt the vaccine formulation and vaccination regime by using a strong adjuvant and several boosts, respectively (O’Hagan, 1998; Brun et al., 2011). Recently, virus-like particles (VLPs) and subviral particles (SVPs) have received great attention due to their potential application in vaccine development as well as in drug targeting and gene therapy (Zhao et al., 2011). VLPs are composed of one or more recombinantly expressed viral proteins, which spontaneously assemble into supermolecular structure. VLPs have the same size and morphology as the parent virus, whereas SVPs are smaller in size or possessing a lesser degree of organization than a comparable intact viral particle because the parent virus comprises several different coat protein subunits while SVPs are homomeric (Tsoka et al., 2000; Brumfield et al., 2004; Allnutt et al., 2007; Brown et al., 2009; Manzenrieder et al., 2011). VLPs and SVPs, like the viral particles of their origin, are robust, symmetrical uniform nano-scale structures, but in contrast to them, are devoid of the genome. Therefore, they mimic the structure of the native virus but provide a non-replicative but immunogenic form, which provides a safe tool for a vaccine approach (Roy and Noad, 2008). Moreover, lacking the genome prevents any risk associated with virus replication, mutation, reversion and reassortment (Jennings and Bachmann, 2008). Both VLPs and SVPs have shown strong immunogenicity due to their strong stability and multivalency of antigen presentation. Their strong immunogenicity in stimulation of humoral and cellular responses may compensate for the need for an adjuvant to induce a strong immune response in vaccination (Grgacic and Anderson, 2006). In addition, antigenic particles are efficiently taken up by M cells and dendritic cells (DCs) in the small intestine when orally administered and consequently lead to an improved stimulation of immune cells. So far, only two kinds of VLPs have been licensed as commercial vaccines for application in humans, which are only administered through injection: the human papillomavirus and Hepatitis B vaccines are the first VLP-based vaccines approved by the Food and Drug Administration (FDA), but many others are still in preclinical trials (Brun et al., 2011). Also, in veterinary vaccination some VLP and SVP-based vaccines have been generated such as Porcine parvovirus, equine Rhinitia A virus, Bluetongue virus, Chicken anemia virus and Infectious bursal disease virus (IBDV) (Crisci et al., 2012). But among them, only the Porcine parvovirus type2 (PCV2) VLP-based vaccine found the way to the market under two brands name Ingelvac CircoFLEX (Boehringer Ingelheim, Germany) and Porcilis PCV (Intervet International, he Netherlands). In the last decades, plants have been investigated as alternative systems for production of subunit vaccines (Tiwari et al., 2009). Even though sometimes low expression levels due to poor protein folding and low stability in plants create a bottleneck (Fischer et al., 2004), the excellent scalability (Kusnadi et al., 1998; Yonekura-Sakakibara, 2006) and cost efficiency (Hood et al., 2002) as well

Page 10: Expression and Characterization of Infectious Bursal

Chapter I Introduction

4

as their potential use in direct oral delivery of vaccines make them very attractive compared to conventional microbial and mammalian production systems. In addition, plants are free of human and animal pathogens (safety). Finally, a major advantage of using plants for animal vaccines is that the plant constituents such as pigments, alkaloids and polyphenols as well as plant sugar structures are able to stimulate both innate and acquired immunity (Bae et al., 2003; Paul and Ma, 2010; Bosch et al., 2010; Malabadi et al., 2011). However, the vast majority of recombinant proteins, especially therapeutics including vaccines are still successfully produced in microbial systems (Allnut et al., 2007; Lin et al., 2012) or mammalian cell cultures. For example, Pichia pastoris has many of the advantages of higher eukaryotic expression systems such as protein processing, protein folding, and posttranslational modification, while being as easy to manipulate as Escherichia coli or Saccharomyces cerevisiae. It is faster, easier, and less expensive to use than other eukaryotic expression systems and generally gives higher expression levels. It shares the advantages of molecular and genetic manipulations with Saccharomyces, and it has the extra advantage of 10- to 100-fold higher heterologous protein expression levels. Both S. cerevisiae and P. pastoris have a majority of N-linked glycosylation of the high-mannose type; however, the length of the oligosaccharide chains added post-translationally to proteins in Pichia (average 8-14 mannose residues per side chain) is much shorter than those in S. cerevisiae (50-150 mannose residues) (Grinna and Tschopp, 1989; Tschopp et al., 1987b). Very little O-linked glycosylation has been observed in Pichia. These features make Pichia very useful as a protein expression system especially for vaccine production. Presently, P. pastoris is one of the most heterologous systems being extensively used for commercial production of high valuable proteins such as pharmaceuticals (Li et al., 2012), enzymes (Wongwisansri et al., 2012; Soares-Costa et al., 2012) as well as vaccines (Jha et al., 2007). Pichia system like plant cells is free of pathogenic and toxic hazards.

I.2 Vaccine administration

Parenteral immunization I.2.1Parenteral immunization (injection or shot-based vaccination) is the most common method of vaccine administration. There are various ways of administration (e.g. subcutaneous, intravenous, interadermal, intramascular). By these methods, the quantity of vaccine can be easily controlled resulting in a measurable antibody and lymphoid response in the blood. This procedure quite efficaciously elicits protective immunity. But there are also some objections, for example the need to cold chain transportation and trained medical personnel, needles and syringes requirement, which increases the cost of administration and restricts the accessibility to mass endangered populations. Moreover, using needle and syringe imposes fear and stress, which has a negative effect on vaccination efficiency and increases the risk of infectious transmission. Due to the mentioned concerns, immunization without needles and syringes has been received great interest (Aziz et al., 2007).

Mucosal immunization I.2.2In contrast to parenteral immunization, mucosal immunization is inexpensive, more safe and

Page 11: Expression and Characterization of Infectious Bursal

Chapter I Introduction

5

acceptable. It provides rapid immunization of a population with less concern. Moreover, mucosal immunization stimulates the mucosal immune system as a first frontier line in preventing many pathogens to enter the body, the event which is absent in parenteral immunization. Many pathogens enter the body via mucous membranes either orally, nasally or genitally. In conjunction with skin, these membranes make a comprehensive anatomical barrier that constitutes the first line of defense against pathogens. These mucosal membranes are defended by an underlying well-organized mucosal lymphoid tissue (MALT). The MALT can be divided into tissues such as GALT (gut-associated lymphoid tissue), BALT or NALT (bronchus/nasal- associated lymphoid tissue), RALT (rectal associated lymphoid tissue), genitourinary tract, etc. Secretory immunoglobulin A (SIgA or S-IgA) is the main part of the mucosal immune system. Moreover, mucosal immunization also leads to the generation of systemic immunity by producing serum immunoglobulin G (IgG). Thus, SIgA is of primary importance for the host defense not only against mucosal pathogens but also pathogens causing systemic diseases. Therefore, SIgA provide two layers of defense against mucosal pathogens (Salyaev et al., 2010). To mimic the effect of the pathogen on the immune system and to induce immunity in the same manner by vaccination, choosing the mucosal route for delivery of the vaccine is crucial. The gut-associated lymphoid tissues (GALT) are the primary targets for the oral delivery of vaccination. In the oral immune system, antigens are delivered via the gastrointestinal tract to inductive sites of the mucosal system named Payer’s patches, a large cluster of lymphoid follicles. The surface of epithelia in Payer’s patches regions constitutes of a single layer of cells known as follicle-associated epithelium (FAE). These regions contain dispersed microfold cells (M cells) in the structure underlying the FAE. M cells provide transportation of intact macromolecules and microorganisms across the epithelial barrier to sub-epithelial dendritic cells (DCs), which are subsequently being presented in the adjacent mucosal T-cell area (Neutra and Kozlowski, 2006). The abundance of M cells is quite low but it was shown that the FAE can alternate its epithelial and lymphoid cell populations in response to antigen stimulation (Kerneis et al., 1997). The epithelial layer has a crucial role in the immune system by constant delivery of antigens from the intestinal lumen to underlying GALT via M cells. Beneath the M cells, highly compartmentalized follicles are settled. This region is rich in B cells as well as T cells, macrophages and DCs (figure I.1). M cells are physiologically and morphologically different from epithelial cells and are specialized to uptake particulate antigens. They lack brush microvilli and have a thin layer of glycocalyx. Besides, they have relatively smaller vesicles and less lysosome, which contribute to the relative intact delivery of antigens to APCs (Aziz et al., 2007). This characteristic opens a gateway for some pathogens such as Salmonella and Vibrio cholera to enter the body via M cells and cause disease. A positive aspect is that M cells facilitate an entranceway for micro/nano particle and mucosal vaccines to stimulate mucosal immune response (Neutra et al., 1996). M cells also have deep invagination in the plasma membrane, which facilitates a strong contact with lymphocytes. When antigens are taken up and transcytosed through M cells, they are mainly digested and presented by DCs (Kelsall et al., 1996). The antigen-loaded DCs migrate to local lymph nodes where they present the antigen to naïve lymphocytes, which originate from the blood. This interaction leads to lymphocyte maturation. Thus, they were subjected to clonal selection, class switching and expansion under the effect of presented antigen by activated APCs and

Page 12: Expression and Characterization of Infectious Bursal

Chapter I Introduction

6

cytokines. These antigen-specific B cells along with T cells including helper T cells (Th1/Th2) and cytotoxic T lymphocytes (CTLs) migrate to mesenteric lymph nodes (MLNs) and travel to mucosal effector sites such as intestinal mucosal respiratory and genital tract. MLNs serve as a connector between peripheral lymph nodes and mucosal recirculating pathway.

Figure I.1 The schematic feature of mucosal immune system in the gut. Inductive sites of mucosal immunity are constituted by MALT (mucosa-associated lymphoid tissue) with their B-cell follicles and M cells that pass the antigen from the lumen to antigen presenting cells (APCs) including follicular dendritic cells (FDCs), macrophages and B cells. In addition, dendritic cells (DCs) may receive the antigen at the effector sites. APCs move to local lymph nodes, become active and stimulate naïve T and B cells. APCs also move to mesenteric lymph nodes via lymphatic vessels and stimulate Naïve T and B cells reside there. Stimulated lymphocytes proliferate and migrate from lymph nodes, distribute to peripheral blood and finally localize at mucosal effector sites. Differentiations of SIgA B cells take place during their dispersion to the effector sites. At effector site, e.g. gut lamina propria, many T helper cells (CD4+) and B cells reside. B cells start to secrete J-chain expressing IgA (dimmers/polymers) and IgM (pentamers). The generated secretory IgA (SIgA) and secretory IgM (SIgM) interact with pIgR (polymeric Ig receptor or secretory component (SC)-mediated epithelial transport) on the surface of epithelial cells and transferred by transcytosis to the other side of the cell and into the lumen. The secretory component (SC), which is part of pIgR, is added to the antibody polymer to protect it from enzymatic degradation. Finally, the expression of memory/effector T cells can take place within the surface epithelium. The small picture in the left down corner shows an M cell and its “pocket” containing different cell types. Figure reference is Brandtzaeg et al. (2008). Activated lymphocytes enter to the blood stream and leave it when they reach to the effector sites such as lamina proporia. In lamina proporia, IgA-committed B cells interact with T cells moderated by cytokines, TGF-β (transforming growth factor-β) and IL-10 (Interleukin-10), which are produced by T cells, subsequently leading to the production of SIgA (Guy-Grand et al., 1974; Brandtzaeg et al., 2008). A large number of activated B cells (IgA plasma cells) reside in these sites and start to secrete IgA. Polymeric forms of the IgA produced by mucosal plasma cells bind to a glycoprotein named polymeric Ig receptor (pIgR) on the surface of epithelial cells. This receptor mediates external transport of dimeric and polymeric forms of IgA and is cleaved from it on the luminal epithelial face. Thus, the polymeric form of SIgA secretes to the lumen while

Page 13: Expression and Characterization of Infectious Bursal

Chapter I Introduction

7

carrying a part of pIgR, which is known as secretory component (SC). In this manner the local mucosal immunity is induced and by producing memory/effector B and T cells and secretion of IgA, inhibits the pathogen to interact with receptors on the mucosal cell surface and consequently stops the pathogen in the preliminary stage of entrance. Thus, the long-term mucosal as well as systemic immunity is generated as a result of cooperation between epithelial cells, B and T cells and stimuli elicited by Th1, Th2 and cytokines. This explains why mucosal vaccination is essential to protect against mucosal pathogens. Interestingly, stimulated lymphocytes in the GALT mainly tend to return and reside in the mucosa of their origin (gut: their inductive site) and differentiate into effector/memory cells. This process that directs and recruits certain lymphocytes to become activated in order to remove mucosal antigens in certain parts and as a result limits their presence in specific organ, is called “homing”. This compartmentalization within the mucosal immune system has a great impact on choosing the proper vaccination route to induce effective immune responses in the target sites (Holmgren and Czerkinsky, 2005). Accordingly, oral immunization induces antibody response in small intestine, ascending colon, mammary and salivary glands. Nasal vaccination in human elicits immunity in upper airway mucosa, regional secretions (saliva and nasal secretions) and vaginal organs. Conversely, rectal and vaginal vaccinations lead to just local immune response (Holmgren and Czerkinsky, 2005).

Challenges in mucosal immunization I.2.2.1Exploiting attenuated bacterial or viruses either for immune-stimulation against their own antigens or recombinant antigens, which are carried and presented by them has been accomplished as vaccine application (Yin et al., 2010; Brennan et al., 2001; Hwang, 2010; Schoen et al., 2004; Vajdy et al., 2004). Despite the advantages of mucosal vaccines, just few vaccines of this type are approved for human use. Examples are Poliovirus, rotavirus, Vibrio cholera, Salmonella typhi Ty21 (against Typhoid), Vibrio cholera, Rotavirus and attenuated and nasal influenza vaccines, which are licensed for human use (McCluskie and Davis, 1999; Holmgren & Czerkinsky, 2005; Salyaev et al., 2010). Oral delivery of subunit vaccines is an attractive option due to its safety, cost-effectiveness and easy handling properties. The main concerns of mucosal vaccine delivery are: (1) the vaccine should be protected from low pH and enzymatic digestion, (2) proper targeting to mucosal inductive sites or M cells (3) proper stimulation of the innate immune response to generate effective adaptive immunity. During the relatively long journey of the oral vaccine from the mouth to the inductive site (small intestine) through the gastrointestinal tract, it encounters to obstacles such as acidic pH and proteolytic environment. These obstacles may partly degrade the vaccine before reaching the inductive sites and consequently decrease the efficacy of the vaccination. Therefore, in contrast to parenteral immunization, it is difficult to measure the actual dose of mucosally delivered vaccine reaching the inductive sites resulting in the stimulation of the immune system. To achieve immunogenicity, either a large amount of vaccine should be administered or the antigen should be protected from acidic pH and proteolysis through encapsulation. In addition, the majority of vaccine is not immunogenic when delivered mucosally and need to be co-administered with strong oral adjuvants (Salyaev et al., 2010) and/or efficiently being targeted to the M cells.

Page 14: Expression and Characterization of Infectious Bursal

Chapter I Introduction

8

Using lipid-based structures with entrapped antigen (Copland et al., 2005), various types of biodegradable complexes (Rieger et al., 2009; Mishra et al., 2010; Garg et al., 2010; Demento et al., 2010), different mucosa-binding proteins including either classical plant lectin or bacterial proteins such as the binding subunit protein of cholera toxin or E. coli heat labile enterotoxin, which are linked either chemically or as gene fusion protein to antigens, have been addressed (Holmgren and Czerkinsky, 2005). In addition, synthetic oligodeoxynucleotides (ODNs) such as CpG motifs (CpG ODN), which resembles genomic DNA extracted from E. coli (poephogus grunniens strain) (McCluskie et al., 2000; Dar et al., 2009) and co-administration of cytokines and chemokines (Staats et al., 2001; Eo et al., 2001; Lillard et al., 2001) have been introduced as promising methods for increasing the vaccine efficiency. In the late 1990’s, a novel approach known as “edible plant vaccines” received great interest (Mason et al., 1996; Yu et al., 2001). Transgenic plants that produce the target antigen are promising tools for mucosal delivery of large amounts of protective antigens in a plant cell-encapsulated form. Plant cell walls protect the antigen from harsh environmental condition and enable it to reach the inductive sites (Streatfield, 2006). Transgenic plants such as tobacco, potato, lettuce, spinach, tomato, carrot, pea, alfalfa and soybean have been used for plant-produced vaccines (Fischer and Schillberg, 2004). Plant-made mucosal vaccine have several advantageous including, low cost of production due to elimination of downstream processing of traditional vaccines such as purification, sterilization and cold chain transport. Plant systems are free of human and animal pathogens. In addition, plant expression system facilitates post-translational modifications, which may be essential for antigen folding into the immunogenic form. Plants are the majority part of human and animal’s diet therefore can carry a large amount of vaccine with less concern about inducing allergic and autoimmune diseases. Mucosal delivery of plant-produced antigen has been accomplished in several cases and led to the generation of antigen-specific antibodies in mice and humans such as in vaccination against cholera (Davoodi-Semiromi et al., 2009), rabies (Yusibov et al., 2008), Norwalk virus (Zhang et al., 2006), diarrhea (Tacket, 2007) and hepatitis B (Thanavala et al., 2005). Some of the plant-produced antigens are currently in preclinical and clinical trials (Davoodi-Semiromi et al., 2009). Despite the great appeal of plant-derived vaccines, up to now only one animal vaccine against Newcastle disease in chicken has achieved approval by the USDA, but due to a strategic decision of the responsible company, Dow AgroSciences (Indianapolis, USA), the vaccine has not been introduced into the market although the plant-produced product was compatible to others produced in alternative production hosts. A major challenge in oral vaccine delivery is that a 1000-fold dose needs to be administered orally in comparison with parenteral delivery. However, the normal yield range in plant expression is 0.01-0.4% of total soluble protein (TSP) (Salyaev et al., 2010) which is often too low to provide sufficient amounts for immunization. The yeast expression system P. pastoris has also been widely used for the expression of viral and bacterial antigens (Lin et al., 2012). Pichia is also not pathogenic for human and most of the animals. Expression of recombinant proteins in Pichia is faster, more simple and less expensive compare to other eukaryotic expression systems (Ruth et al., 2010).

Page 15: Expression and Characterization of Infectious Bursal

Chapter I Introduction

9

Until 2009, about 20% of the produced recombinant biopharmaceuticals on the market were produced in yeast systems, nearly all in S. cerevisiae. In 2009 the first biopharmaceutical protein, Kallikrein inhibitor (Kalbitor by Dynax Inc.), produced in P. pastoris was approved by the FDA (Mattanovich et al., 2012). Shanvac-B, antigen of hepatitis B virus (HBsAg) produced in Pichia was also approved in Indian market (Chakma et al., 2011).

Oral tolerance I.2.2.2The mucosal immune system should protect the individual from attacking pathogens while being tolerant against innocuous substances found in food. Accordingly, oral immunization has the potential to induce either protection or oral tolerance corresponding to the administered antigen. As a consequence the immune system is paralyzed in response to the relevant pathogen. It was shown that the induction of oral tolerance is related to the route of antigen internalization. Passing through intestinal epithelial cells instead of lamina proporia and transport through blood vessels to the liver induce oral tolerance (Mayer and Shao, 2004). In other words, inefficient transport of the antigen from the gut lumen to the GALT may induce system immune tolerance. Orally delivered subunit vaccines that are composed of soluble, nonparticulate antigens are inherently less immunogenic and can induce oral tolerance. The effectiveness of mucosal vaccines dramatically increases when the antigens are multimeric and/or in particulate form. From this aspect, VLPs and SVPs are promising mucosal vaccines due to their size and subsequent uptake by M cells and DCs (Huang et al., 2005; Neutra and Kozlowski, 2006). Alternatively, administration of a single high dose of the antigen or repeated low doses induce tolerance. These types of tolerance are called high and low-dose tolerance, respectively, and they are mediated through different mechanisms. T cells have a crucial role in the induction of tolerance. The mechanism of oral tolerance is not well understood. As a result of oral tolerance, when DCs encounter the antigen, they behave differently and secrete IL-10 and TGF-β. This leads to the deletion of antigen specific T cells as well as the generation of regulatory T cells (Tr). Tr suppress the systemic immune response (Aziz et al., 2007).

I.3 The chicken immune system Bird’s immune system is very similar to vertebrates. Whether or not a bird develops a disease depends on bird’s condition, level of immunity, number of invading pathogens and their virulence strength. Innate immunity is affected by genetic factors (ability to develop required receptor against pathogens), body temperature (higher body temperature prevents some bacteria to survive) as well as the health of the anatomical barriers like skin, the mucous and the respiratory tract cilia. Other factors like nutrition, age, stress, inflammatory processes and metabolic factors have great impact on the effectiveness of the immune response. A specific immune response in birds could be generated via two ways. First, passive immunity is achieved when antibodies are transferred to the individual either maternal or orally/through injection. Second, active immunity is the production of antibodies by individuals against a pathogen as a result of vaccine application or recovery from disease, which eventually leads to the generation of memory cells. Birds lack lymph nodes, but they have an avian-specific primary lymphoid organ, the bursa of Fabricius (BF), which is the organ for development of their B-cell repertoire (Kaiser, 2007). When a pathogen enters the body, it is engulfed by APCs and exposed through T cells to B cells. B cells are produced in the liver of

Page 16: Expression and Characterization of Infectious Bursal

Chapter I Introduction

10

the chicken embryo, the yolk sac and the bone marrow. They move to BF for maturation when an antigen is encountered, the BF programs these cells to attack the antigen. The programmed cells then move to the blood, spleen, cecal tonsil, bone marrow, Harderian gland and thymus. It takes five days for B cells to be programmed and to go through clonal expansion resulting in the production of specific antibodies. Antibodies attach to antigens and neutralize them. On the other hand, the antibody-antigen immune complex triggers macrophages to remove the antigens. There are three classes of antibodies found in chickens when they are exposed to antigens: IgM, IgY (or chicken IgG) and IgA. IgM appears after 4-5 days following exposure to an antigen and then disappears by day 10-12. IgY is detectable after 5 days following exposure, peaks at 3 to 3.5 weeks, and then slowly decreases. IgY instead of IgG in mammals is the most important protective antibody in chickens and can be measured by most serological test systems. IgA appears after 5 days following exposure. This antibody isotype is found primarily in the mucus system of the eyes, gut and respiratory tract and provides local mucosal protection to these tissues (Tizard, 2002). T cells in chickens like mammals are programmed and maturated in the thymus. Like mammals, some T cells producing cytokines; some T cells are facilitated to directly destroy the contaminated cells (CTLs); T helpers enhance the activity of B cell, macrophages and other T cells.

Chicken diseases and vaccination I.3.1Innate immune system seems to be fully functional in the newly hatched chickens whereas the optimal adaptive immune responses only develop following the first weeks after hatch. Consequently, they are not able to develop a fast immune response, which makes them susceptible to various diseases in early days. To compensate this deficiency, they receive maternal antibodies via the egg (Davison et al., 2008). These antibodies are specific against all pathogens that the mother has encountered or has been vaccinated against. This antibody reservoir lifetime is long enough to donate immunity until the chicken’s immune system becomes active. Knowing the maternal antibody level is critical for managing vaccination schedules. If chickens receive a vaccine at the time when the maternal antibody level is still high, the vaccine may be inactivated violently by maternal antibodies, which leads to a weak immune response. In contrast, if vaccination takes place long after the decline of maternal antibody levels, chickens would be susceptible to diseases prior to be immunized (Hair-Bejo et al., 2004). Damages to the BF due to late immunization of chickens at early susceptible days of age by IBDV or Marek’s disease inhibits proper B cell maturation and programming. This leads to immune susceptibility so that affected chickens will neither be able to raise immune response against attacking pathogens nor respond to immunization programs, adequately. As a consequence, the right time for vaccination and boosting the immune system is critical for the poultry industry. Newcastle, Influenza and infectious bronchitis as well as infectious bursal disease (IBD) are the most important poultry disease worldwide which need to be controlled by proper vaccination programs (Tao et al., 2009). The majority of currently used vaccines is against systemic pathogens or their toxins and administered parenterally (Zhang et al., 2008). Although parenteral route of vaccination is very effective in stimulation of the systemic immune response, this response does not extend to mucosal interfaces. To stimulate mucosal immunity, the pathogen or vaccine should be delivered via the

Page 17: Expression and Characterization of Infectious Bursal

Chapter I Introduction

11

mucosal route. By this way, mucosal as well as systemic immunity are being generated (Boyaka et al., 2003).

I.4 Infectious bursal disease virus (IBDV) Infectious bursal disease virus (IBDV) is an acute, highly contagious pathogen, which causes intensive irreparable immunosuppression or often death in young chickens, often at an age of 3-6 weeks when the B lymphocytes are developing, and as a result imposes severe losses to the poultry industry annually worldwide. Younger chickens are passively protected by maternal antibodies transmitted via egg yolk and older ones are able to produce antibodies against the virus, therefore rarely develop clinical signs of the disease (Macreadie et al., 1990). Since the first outbreaks occurred in Gumboro, Delaware, “Gumboro disease” became a synonym for IBD (Cosgrove, 1962).

Virus classification I.4.1IBDV belongs to the genus Avibirnavirus of the family Birnaviridae. The virion is composed of a non-enveloped icosahedral capsid with a bi-segmented double strand RNA genome. It has two distinct serotypes (I and II) but only serotype I is virulent for chickens. An intrinsic characteristic of RNA viruses is the high variability of their genome due to the low proofreading activity of their viral replicases. Accordingly, many strains have been generated. Based on the pathotype detectable IBDV in the field can be divided to classical, antigenic variant and very virulent strains. Classical strains cause inflammation and severe lymphoid necrosis, immunosuppression and up to 30% mortality. Variant strains cause bursal atrophy and immunosuppression, with very low mortality whereas very virulent IBDV (vvIBDV) may cause acute clinical disease with 60-100% mortality (Snyder et al., 1988; Jackwood and Jackwood., 1994; Brown and Skinner, 1996). vvIBDV strains have emerged in Europe in 1987 and spread to Latin America, South-East Asia, Africa and the Middle East since the last three decades. One reason for the worldwide distribution of the virus is that it is extremely resistant in the environment. The Office of International des Epizooties (OIE) estimates that IBD is present in more than 95% of the 117 member countries. The acute clinical form of IBD caused by vvIBDV isolates has been observed in over 80% of these countries. It has been reported in Europe, Asia, Africa, South America and Central America.

Route of infection and pathology I.4.2The route of IBDV infection is usually oral (from contaminated food, water or excrements), but may be via the eye or respiratory tract, with an incubation period of 2-3 days. Following oral infection, the virus replicates in gut-associated macrophages as well as lymphoid cells (Mahgoub, 2012). The virus attacks the immature IgM-bearing B-lymphocytes in the BF and destructs the tissue resulting in the immunodeficiency of its host (Shaarma et al., 2000). Depression, anorexia, ruffled feathers, unsteady gait, trembling, huddling under equipment, watery diarrhea and the most prominent lesion, hemorrhage and necrosis of bursa of Fabricius, followed by bursal atrophy are some clinical symptoms of this disease (Chang et al., 2001). If chickens are infected within 1-3 weeks of hatch, they show immunodeficiency without symptoms whereas infection within 3-6 weeks of hatch accompanying symptoms leading to death; but no influence on older chickens has

Page 18: Expression and Characterization of Infectious Bursal

Chapter I Introduction

12

been observed. Immunodeficient chickens are susceptible to other disease and show no adequate response to vaccination.

Virus detection, classification and diagnostic tests I.4.3To distinguish between classical, antigenic variant, very virulent and non-virulent strains, it is important to identify the IBDV serotype in order to be able to choose the best strategy for vaccination. Since the distinction between different strains is not possible through serological techniques like agar-gel precipitation test (AGPT) or enzyme linked immunosorbent assay (ELISA), reverse transcription-polymerase chain reaction (RT-PCR) has been used as rapid, sensitive and specific method for detection of IBDV in clinical samples and for phylogenetic studies (Lee et al., 1992; Li et al., 2009). Some laboratories utilized joined RT-PCR/RFLP (Juneja et al., 2008; Bahmaninejad et al., 2008), RT-PCR and nucleotide sequencing (Stoute et al., 2009; Li et al., 2009) and SYBR green I based one-step real-time RT-PCR (Kong et al., 2009) for diagnostic and classifications. Recently, the more sensitive method called reverse-transcription loop-mediated isothermal amplification (RT-LAMP) has been established for rapid diagnostics (Xu et al., 2009).

Treatment and prevention I.4.4No specific treatment is available for IBD. The use of vitamin E and arginine supplements (Ruiz-Feria and Abdukalykova, 2009) and facilitating access to water may help to increase the resistance of chickens against the disease. Antibiotic medication may be recommended if secondary bacterial infection occurs. Therefore, prevention is the best method of choice. Passive immunity protects against IBDV, as does previous infection with avirulent strains. In broiler farms, breeder flocks were immunized against IBD and maternal immunity via protective antibodies passed to their offspring. In each farm region, vaccination should be done according to prevalent strains. Accordingly, determination of virus serotype is very important in choosing the kind of vaccination. Previously, it has been shown that attenuated vaccines were efficient to cope with IBDV (Wyeth and Cullen, 1979). By outbreak of variant strains in the US and vvIBDV strains in Europe, traditional attenuated strain vaccines are not totally effective against new strains. Therefore new vaccines based on vvIBDV strains are being developed. Despite of vaccination, outbreaks of IBD are still reported for many countries (Li et al., 2009).

IBDV structure I.4.5The IBDV genome has two segments of double-stranded RNA, named A and B (Azad et al., 1985; Muller et al., 1979), which are packed in a non-enveloped icosahedral (T=13) shell with 60 nm in diameter (Böttcher et al., 1997). The smaller segment B (~2.8 kb) encodes the protein VP1 (97 kDa), which is an RNA-dependant polymerase. The segment A (~3.3 kb) contains two partially overlapping open reading frames. The smaller ORF encodes the non-structural protein VP5, which is not essential for virus replication neither in vitro nor in vivo (Mundt et al., 1997) but has a role in virus release (Gariiga et al., 2006; Wu et al., 2009). The larger ORF encodes a precursor 110 kDa polyprotein (VP2-VP4-VP3) that is post-translationally cleaved into three distinct proteins: pre-VP2 (54 kDa), VP4 (25 kDa) and VP3 (28 kDa). Processing of the precursor protein is done by the

Page 19: Expression and Characterization of Infectious Bursal

Chapter I Introduction

13

auto-catalytic activity of VP4 which is a viral protease (Azad et al., 1987; Jagadish et al., 1988). Pre-VP2 (pVP2 or VPX) is a precursor and truncates at its C-terminal in four steps into VP2 (441 amino acid and 48 kDa), through the proteolytic activity of VP4 in three Ala-Ala bond positions (487, 494 and 501) and one unknown cleavage between residues 441- 442 (Luque et al., 2007). The whole cleavage sites have been determined through deletion mutants (Caston et al., 2001; Da Costa et al., 2002). The pVP2 processing to VP2 is essential for correct assembly of the viral capsid. VP2 (major protein with ~51% frequency in a virus) assembles into trimers, the structural unit of the virus capsid and forms the external surface of the virion while VP3 trimers form the interior face. VP3 is a multifunctional protein, which reacts with itself (Casanas et al., 2008), VP2 (Saugar et al., 2005), VP1 (Maraver et al., 2003; Garriga et al., 2007) and the viral genome (Hjalmarsson et al., 1999). Briefly, VP3 has a key role in interacting with pVP2 for correct assembly into the capsid format (Chevalear et al., 2002; Chevalier et al., 2004; Ona et al., 2004; Saugar et al., 2005; Luque et al., 2007). It interacts with VP1 by its carboxyl terminal ten amino acids. It also binds to both segments of the dsRNA by positively charged domain immediately upstream of the VP1 binding domain (Tacken et al., 2002; Kochan et al., 2003; Luque et al., 2007). As a result, VP3 is essential for proper assembly and virus morphogenesis (Martinez-Torrecuadrada et al., 2000). In addition, VP3 seems to be involved in initial virus attachment (Reddy et al., 1992). After expressions and maturations of all viral proteins, the full-size capsid including the genome and viral proteins including VP1, VP2, VP3, VP4 and VP5 proteins is formed (figure I.2).

Figure I.2 Schematic configuration of IBDV and its genome. Segment A of the viral genome encodes two overlapping ORFs: ORF1 encodes VP5 with a role in virus release (Gariiga et al., 2006; Wu et al., 2009) and ORF2 encodes a polyprotein. The polyprotein is cleaved by the proteolytic activity of VP4 into three separate proteins. VPX or pVP2 is truncated to VP2 via proteolytic cleavage. VP2 is the capsid protein of virus, which is the main antigenic protein containing major epitopes responsible for eliciting immunogenic responses (Becht et al., 1988; Azad et al., 1991; Heine et al., 1991): Picture taken from the viralZone website (www.http://viralzone.expasy.org/viralzone/all_by_species/572.html).

Page 20: Expression and Characterization of Infectious Bursal

Chapter I Introduction

14

Native IBDV has an icosahedral symmetry with triangulation number (T) of 13 and 60 nm diameter made of 12 pentamers and 120 hexamers according to trimers decoration (Luque et al., 2007). Structural studies indicated that the virus capsid is composed of 780 VP2 molecules in form of 260 structural trimer subunits (figure I.3), which stand on the outer face of the particle (Böttcher et al., 1997). On the inner side, 600 (or ~ 450 (Luque et al., 2007)) copies of VP3 in Y-shaped trimers make a scaffold responsible for the stability of the particle. VP4 is located on 60 positions next to 5-fold axes of icosahedrons (Böttcher et al., 1997; Hu et al., 1999). The RNA genome is in contact with positively charged C-terminus of VP3 inside the particles (Böttcher et al., 1997; Hudson et al., 1986).

Figure I.3 Three VP2 proteins assemble together and form a trimer, the structural unit of the virus. Virus particles obtained by assembly of trimers (VP2 trimers outside and VP3 trimers inside of the particles) and engulfment of VP1, VP4, VP5 and bi-segmental genome. (A) The conformation of the trimer (VP2 proteins were shown in orange, red and blue) structural unit in spike-like form on the surface of the viral particle. (B) Electron micrograph of IBDV particles with about 60 nm in diameter. Figure A and B references are http://www.igs.cnrs-mrs.fr/elnemo/NORMA/ibdv_vp2_fit.12modes.gif and http://www.uni-leipzig.de/forsch98/vetmed/virolo.htm, respectively.

Structure and characteristics of the viral antigen IBD-VP2 I.4.6Sequence comparison between corresponding regions of genomes in different IBDV strains revealed that generally all viruses are very closely related but they show a hyper variable (hv) region (amino acid 206-350) in VP2, which is responsible for the antigenic variation observed in different viruses (Bayliss et al., 1990; Heine et al., 1991). In other word, although there are more than 93% similarity between sequences of proteins of known IBD-VP2 described all over the world (this number is more than 98% between Asian and European vvIBDV strains), this virus has been classified into different strains with different pathogenesis according to their diversity in the hypervariable region of VP2 (Liu et al., 2002; Kasanga et al., 2007). The hyper variable region is the most interested region in most epidemiology and phylogenetic studies. In spite of high frequency of mutation in this region, this part of the genome also contains relatively conserved sequence regions unique for vvIBDV strains (Jackwood et al., 2005; Parede et al., 2003; Hoque et al., 2001). On the other hand, this variable region with frequent mutations provides greater discrimination between closely related genomes and consequently is more important in evolutionary tracking and categorization than the constant regions (Levin et al., 1999; Li et al., 2009)

Page 21: Expression and Characterization of Infectious Bursal

Chapter I Introduction

15

VP2, which makes 51% of the total IBDV protein content (Böttcher et al., 1997), is the main antigenic protein containing major epitopes responsible for eliciting immunity (Becht et al., 1988; Azad et al., 1991; Heine et al., 1991). The hv region involves two major hydrophilic regions (figure I.4) (Kasanga et al., 2007). Nevertheless, the whole VP2 is characterized as a hydrophobic protein (Wu et al., 2004; figure 1.4). Each major hydrophilic region (amino acid 210-225 and 312-324) carries one part of the neutralizing epitope and altogether provides the conformational epitope of the virion (Yamaguchi et al., 1996; Cui et al., 2003).

Figure I.4 Hydrophobicity plot of IBD-VP2 provided by CLC Main workbench 6.5 using Kyte-Doolittle hydrophobicity scale. Two major hydrophilic regions (amino acids 210-225 and 312-324), which are also carrying the virus neutralizing epitopic regions have been circled in orange. As illustrated in this graph, VP2 is a hydrophobic protein. Since disulfide bond formation, glycosylation or phosphorylation has not been reported in VP2, oligomerization is known as the main factor providing conformational epitopes (Yamaguchi et al., 1996). The atomic structure of intact virions was solved by X-ray crystallography (Coulibaly et al., 2005). Further studies were performed with higher resolution (2.6 Aº) on the VP2 structure (Garriga et al., 2006; Lee et al., 2006a). These studies revealed that the VP2 subunit is folded into three domains termed the projectin (P), shell (S) and base (B) domains (figure 1.5). The P and S domains are β-barrels, which are in radial and tangential format to the particle surface, respectively. The P domain carries the neutralizing conformational epitope region. This region involves 210-222 amino acids and 312-324 amino acid sequences, matching with two major hydrophobic regions. The N and C terminal VP2 regions are involved in the formation of the α-sheet structure (figure I.5) (Letzel et al., 2007; Luque et al., 2007).

Page 22: Expression and Characterization of Infectious Bursal

Chapter I Introduction

16

Figure I.5 (A) Protein structure of VP2; which is composed of three domains named P (brown), S (blue) and B (green). Domain P carries the virus neutralizing epitope region (comprising amino acids 210-222, shown in red and amino acids 312-324, shown in yellow). The figure reference is Saugar et al. (2010). (B) The trimeric VP2 structure from the top view. The conformational epitope is shown in red (comprising amino acids 210-222) and yellow (comprising amino acids 312-324). The figure was rendered using the program PyMOL using the file with PDB code 2DF7 (Lee et al., 2006a). As described above, VP2 is the most frequent protein and the major host protective immunogen of the virion, which contains the antigenic regions of the virion (conformational epitope) responsible for inducing neutralizing antibodies against the parent virus in the host (Azad et al., 1987; Becht et al., 1988). It has been proven that VP2 alone is able to stimulate an immune response and eventually triggers antibody generation in the same way as the intact virus (Fahey et al., 1989). Immunodominant epitopes in VP2 region converts it to a target for designing subunit vaccines and diagnostic tests (Sedighzadeh et al., 2012). On the other hand, it was shown that recombinant expression of capsid proteins of non-enveloped viruses led to different grades of assembly with variable efficiencies (Chevalier et al., 2004). Recombinant expression of VP2 in combination with other IBD viral proteins and alone with different length, has led to various forms of assembly. Different types of assembly are also seen in IBDV infected cells in nature; In addition to IBD virions, tubular forms with a diameter of 55 nm (type I), and 24-26 nm (type II) are also formed during infection. Tubes type I is made only by pVP2 while type II contains VP4 (Chevalier et al., 2004). The expression of VP2 in companion with VP3 and VP4 in the whole polyprotein A (VP2-VP4-VP3) in different heterologous system resulted in the formation of VLPs with icosahedral symmetry based on T= 13 lattice and 60 nm in diameter. Production of IBD-VLPs has been observed in E. coli (Rogel et al., 2003), yeasts such as S. cerevisiae (Macreadie et al., 1990; Jagadish et al., 1990), baculovirus and insect cell lines e.g. Sf9 and Hi-5 (Vakharia et al., 1993; Kibenge et al., 1999; Hu et al., 1999; Lee et al., 2006b) as well as mammalian cell lines like the African green monkey kidney epithelial BSC-1 (Fernandez-Arias et al., 1998). As previously mentioned, an interesting trait of VP2 is its ability of self-assembly. In fact, different sizes of VP2 ranging from 512 amino acids (pVP2) to 441 amino acids (mature VP2) have been recombinantly expressed and led to different conformations (Saugar et al, 2005). While expression of mature VP2 resulted in the formation of dodecahedral T= 1 subviral particles (SVP), pVP2 expression led to the assembly of tubular structures with hexagonal lattice (Luque et al., 2007). T= 1 IBD-SVPs are composed of 20 VP2 trimers as subunits (Caston et al., 2001) and made of only the first 441 amino

Page 23: Expression and Characterization of Infectious Bursal

Chapter I Introduction

17

acids of VP2 (Coulibaly et al., 2005). Production of IBD-SVPs has been reported in different hosts like E. coli (Chen et al., 2005), S. cerevisiae (Garriga et al., 2006; Dey et al., 2009), SF9 and H5 insect cell lines (Dybing and Jackwood, 1997; Wang et al., 2000; Caston et al., 2001; Lin et al., 2007; Luque et al., 2007). The ability of self-assembly along with carrying the virus neutralizing epitope, make VP2 a promising subunit vaccine candidate.

I.5 Vaccine development against IBDV

Control of IBD has been initially achieved by maternal-derived antibodies (MDA) induced by live or attenuated vaccines applied to breeder progenies and by active immunization of chickens with inactivated viruses. During outbreaks of very virulent strains, traditional vaccines lost their effectiveness since they were not able to stimulate a strong immune response and are risky to apply due to their potential exacerbation of the disease. To cope with vvIBDV strains, new vaccines are being developed but they evoke only a moderate immune response and therefore are not able to fully prevent virus pathogenesis. Generation of genetically engineered IBDV using reverse genetics was afforded based on the introduction of mutation in hv region of VP2 to create attenuated IBDV with the potential application as vaccine. IBDV immune complex vaccines, which consist of a mixture of certain amount of VP2-specific antibodies extracted from the sera of hyper-immunized chickens and infectious virus was also introduced with in ovo application (Müller et al., 2012). Extraction of virus from infected BF of the chickens is one of the major sources for generation of most effective vaccines but to obtain large quantities of this antigen a large number of contaminated chickens has to be sacrificed (Pitcovski et al., 2003). Virus proliferation in cell culture and subsequent extraction, purification and attenuation or inactivation is a common but expensive and laborious method in vaccine production. It was shown that transferring anti-VP2 antibodies to susceptible chickens can passively protect them from IBDV (Fahey et al., 1989; Macreadie et al., 1990; Vakharia et al., 1993). It was also shown that VP2 is essential and adequate for stimulation of the immune system in chickens and as a result could be a powerful candidate for antigen-based vaccines either in oral or parenteral vaccination strategies (Wu et al., 2007, Rong et al., 2007). By development of recombinant DNA technology, many recombinant vaccines including DNA-based and antigen-based have been revealed. In previous years, several IBD vaccines have been introduced but still need time to be improved. There is a need to improve safety, tolerability and potency. Besides, decreasing the cost and speeding up the rate and scale of vaccine production to meet daily increasing demands are global aims. Recombinant expression of IBDV polyproteins in several heterologous systems has been extensively reported (Chevalier et al., 2002). Among those, some have shown the formation of virus-like particles (VLPs) (Fernandez-Arias et al., 1998; Kibenge et al., 1999). In this type of vaccine, VP2 protein as a major host protective antigen has been attracted the most attention in vaccine development (Bentley et al., 1994). Several recombinant expression systems have been successfully afforded for the production of immunogenic and protective VP2 (in table I.I some of them are summarized). Self-assembly has also been reported in some of the heterologous expression systems. Oral delivery is preferred over parenteral immunization due to less stress for the vaccine recipient, the possibility to induce

Page 24: Expression and Characterization of Infectious Bursal

Chapter I Introduction

18

mucosal immunity and the low cost and ease of administration. VP2, which was produced in plant systems such as Arabidopsis thaliana (Wu et al., 2004) and rice (Wu et al., 2007) was able to induce protective immune response when administered orally but no assembly to VLPs was reported. Due to the low accumulation of the recombinant vaccine in the plant cells, high volumes of plant materials with several repetitions should administered to reach the adequate immune-stimulating dosage. An ideal oral subunit vaccine for production in transgenic plants is the primary antigen of the infectious pathogen that accumulates in high levels and also aggregates in forms that are recognized at mucosal sites where an immune response is triggered. These include viral coat proteins such as IBD-VP2 that co-assemble to form VLPs or SVPs.

Page 25: Expression and Characterization of Infectious Bursal

Chapter I Introduction

19

Tab

le I

.1 R

ecom

bina

nt e

xpre

ssio

n sy

stem

s th

at h

ave

been

use

d fo

r pro

duct

ion

of s

ubun

it va

ccin

es fo

r chi

cken

s ag

ains

t IB

DV

by

expr

essi

on o

f IB

D-

VP2

.

Ref

eren

ces

Aza

d et

al.

1991

Om

ar e

t al.,

200

6;

Ron

g et

al.,

200

7

Aza

d et

al.,

199

1

Pitc

ovsk

i et a

l., 2

003

Mar

tinez

-To

rrec

uadr

ada

et a

l.,

2003

Lee

et a

l., 2

006b

Lai e

t al.,

200

4

Wu

et a

l., 2

004

Wu

et a

l., 2

007

NR

: not

repo

rted

Imm

unog

enic

ity

Non

-imm

unog

enic

Imm

unog

enic

but

no

t ful

ly p

rote

ctiv

e

Imm

unog

enic

Prot

ectiv

e

Prot

ectiv

e

Prot

ectiv

e

Prot

ectiv

e

Prot

ectiv

e

Prot

ectiv

e

Rou

te o

f vac

cine

de

liver

y to

the

chic

kens

Intra

mas

cula

r in

ject

ion

Intra

mas

cula

r in

ject

ion

Intra

mas

cula

r in

ject

ion

Intra

mas

cula

r in

ject

ion

Subc

utan

eous

Intra

mas

cula

r in

ject

ion

Intra

mas

cula

r in

ject

ion

Ora

l

Ora

l

Dos

e

50 µ

g

50 µ

g

50 µ

g

100

µg

3 µ

g

20 µ

g

20 µ

g

5× 5

µg

4× 2

-10

mg

Des

crip

tion

Form

atio

n of

in

clus

ion

body

Wat

er so

lubl

e pr

otei

n

Wat

er so

lubl

e pr

otei

n ag

greg

atio

n

Wat

er so

lubl

e pr

otei

n

Form

atio

n of

SV

P, 2

3 nm

in

diam

eter

Fo

rmat

ion

of

SVP,

~25

nm

in

diam

eter

Form

atio

n of

SV

P, 2

3 nm

in

diam

eter

NR

NR

Acc

umul

atio

n le

vel

NR

3.8

% T

SP

NR

NR

~ 8

mg/

l

30 m

g/l

0.4

mg/

g

0.5

- 4.8

% T

SP

0.68

- 4.

5% T

SP

Det

ails

E. c

oli

S. c

erev

isia

e

Pich

ia p

asto

ris

Spod

opte

ra

frug

iper

da (S

f9)

Hig

h-Fi

ve (H

i-5)

Larv

ae o

f Cab

bage

lo

oper

(Tri

chop

lusi

a ni

)

Ara

bido

psis

Ric

e

Hos

t

Bac

teria

Yea

st

Inse

ct c

ell l

ine

(bac

ulov

irus

expr

essi

on

syst

em)

Plan

ts

Page 26: Expression and Characterization of Infectious Bursal

Chapter I Introduction

20

I.6 Application of VLPs and SVPs in nanotechnology

Other than common vaccination approaches, VLP and SVP-based structures have also been exploited for drug targeting (Wang et al., 2002c; Remond et al., 2009; Dhart et al., 2010; Zhao et al., 2011) and for displaying of inserted foreign immunogenic epitope (chimer particles) to the immune system of the host for induction of immune response against the inserted epitopes (Veremeiko et al., 2007; Fraile et al., 2012; Remond et al., 2009). In the latter case, IBD-SVPs were successfully used in displaying foot and mouth disease virus (FMDV) epitopes (Remond et al., 2009). However, generation of chimeric VLP/SVP is empirical and there is always a chance that inserted peptides interrupt in VLP assembly or in case of assembly are not immunogenic. An important limitation for chimeric particles is the limited size and nature of the epitope that can be inserted (Trovato et al., 2012). In the nanotechnology field, viral nanoparticles (VNPs) are defined as nanoscale devices based on natural viruses (Singh et al., 2007; Destito et al., 2007; Steinmetz et al., 2007a, 2008 and 2009). They offer advantages over synthetic nanoparticles such as carbon nanotubes and quantum dots (Steinmetz et al., 2010) because they are biocompatible, biodegradable and non-toxic. They provide a robust and symmetrical structure, amenable to both genetic and chemical modifications (Lee et al., 2009); can be used for the development of novel nanoscale formulations and as nanotemplates, nanoprobes and nanoreactors (Bronstein, 2011). Importantly, VNPs are stable, symmetrical, monodisperse particles with a distinct size, atomic precision and uniform multivalency; they are also easy to prepare in large quantities (Grasso and Santi, 2010). VNPs derived from mammalian viruses are already widely used, but concerns about their potential toxicity and pathogenicity have led to the more recent development of plant VNPs (Singh et al., 2007; Rae et al., 2005; McCormick and Plamer, 2008), which are safer and can be prepared in gram quantities from a few kilograms of infected leaves (Steinmetz et al., 2007a; Nichols et al., 2002; Barnhill et al., 2007a). Spherical VNPs based on Cowpea mosaic virus (CPMV) (Wang et al., 2002a,b,c; Wang et al., 2003; Raja et al., 2003), Cowpea chlorotic mottle virus (CCMV) (Gillitzer et al., 2002) and Turnip yellow mosaic virus (TYMV) (Barnhill et al., 2007a), as well as rod-shaped VNPs based on Tobacco mosaic virus (TMV) (Schlick et al., 2005) have been investigated for this purpose. The majority of research has focused on CPMV making this virus the gold standard for stability, addressability and functionalization (Miermont et al., 2008; Kaltgrad et al., 2008; Sapsford et al., 2006; Destito et al., 2007). As an alternative to VNPs, VLPs and SVPs can be produced recombinantly in higher amount that lack the virus genome, thus providing further safety (Mason et al., 1992; Remond et al., 2009). The absence of genomic nucleic acid leaves spaces for the encapsidation of small useful molecules, e.g. drugs, imaging reagents or nucleic acids (Yamada et al., 2003; Caston et al., 2001). Furthermore, the structure of the particles can be modified by genetic engineering e.g. to make the surface more favorable for the nucleation of a specific mineral or alloy (Shah et al., 2009; Reiss et al., 2004), or to introduce one or more chemically modified amino acids to display a specific chemical group for structural stabilization or conjugation (Steinmetz et al., 2007b; Peabody, 2003; Strable et al., 2008). Modification also provides an opportunity to optimize the physicochemical properties of the particle, which must be tailored for specific applications. For example, nanoparticles designed as contrast agents for

Page 27: Expression and Characterization of Infectious Bursal

Chapter I Introduction

21

magnetic resonance imaging (MRI) should encourage mineralization whereas those used for electronics applications need to be stable in acid, ethanol and at high temperatures. VNPs used for biomedical applications such as imaging or drug delivery often need to be bioconjugated in the presence of dimethylsulfoxide (DMSO), so the particles must be stable in this organic solvent (Steinmetz et al. 2008). The potential application of either IBD-VLPs or IBD-SVPs has not yet been reported.

I.7 Aim of this thesis Infectious bursal disease virus (IBDV) serotype I is a worldwide distributed, highly contagious immunosuppressive agent for young chickens, which causes high morbidity and mortality. IBDV is stable in the environment and survives strict hygiene management and disinfection. No specific treatment is known for infectious bursal disease (IBD) and vaccination is the only adequate way of disease control (Müller et al., 2012). Currently, IBD vaccines are composed of either live attenuated or inactivated (killed) virus. Live vaccines are usually administered orally, by addition to drinking water whereas killed vaccines administered intramuscularly. Due to the emergence of variant strains in the Unites States and very virulent strains in Europe, which are speculated as a result of selection pressure from the live administered vaccines (Synder, 1990), conventional vaccines lost their efficiency to provide full protection (Rong et al., 2005). In addition, their application is accompanied with a risk of reversion of virulence. Over the last three decades, several reports have been published on recombinant expression of IBD-VP2 as the major protective IBDV antigen, responsible for induction of virus neutralizing antibodies in chicken. With the produced vaccines partial to full protection was achieved. But except the ones that are expressed in plant systems, the requirement for parenteral administration is still the limiting factor that increases the cost of administration and needs for protein purification. In contrast, oral delivery of vaccines is an attractive alternative due to the ease of administration and low costs. Also, oral delivery of the recombinant protein could reduce the production costs by almost 90% due to elimination of downstream processing (Chebolu and Daniell, 2009). The aim of the first part of this study was the development of an oral subunit vaccine for chickens against IBDV. Therefore the VP2, the coat protein of IBD virus, which carries the major host protective antigen and has self-assembly capability, was expressed in tobacco and Pichia pastoris. In this context, increasing the VP2 accumulation level through targeting to different organelles in both production systems was investigated. In addition, the self-assembly of the produced VP2 in both expression systems in vivo and in vitro was studied. The immunogenicity of the produced VP2 and its ability in induction of a protective immune response was studied when the vaccine was administered through either parenteral or oral route to the chickens. In the second part the potential application of the generated IBD sub-viral particles (IBD-SVPs) derived from self-assembly of the recombinantly produced VP2 as a nanotechnology platform was investigated. The tolerance window of these nano-particles concerning heat, pH and organic solvent stability was studied. Moreover, the addressability of solvent-exposed amine and carboxylic groups on the surface of IBD-SVPs was tested via traditional bioconjugation methods. A schematic overview of this PhD thesis is represented in the figure below.

Page 28: Expression and Characterization of Infectious Bursal

Chapter I Introduction

22

Figure I.6 Schematic overview of the work presented in this dissertation.

PCR amplification of IBD-VP2 cDNA (441 amino acids) sequence expression vectors

Stable transformation of tobacco

Screening and selection of best protein producers

Characterization of the purified IBD-VP2 • Glycosylation

1. Immunoblot 2. Enzymatic deglycosylation 3. Glyco-proteomics analysis

• Self assembly studies

1. Size exclusion chromatography 2. Electron microscopy

Immunogenicity studies for the tobacco-produced

IBD-VP2

Cloning of IBD-VP2 cDNA into the plant expression vector pTRA

Selection of the best production strategies

Cloning of IBD-VP2 cDNA into the Pichia expression

vector pPICZ_B

Purification of IBD-VP2

Study of IBD-VP2 accumulation levels in different cell compartments

Study the potential application of IBD-VP2 subviral particles as a platform in

nanotechnology

Expression and purification of IBD-VP2

Immunization of chickens and evaluation of the vaccine efficacy using the following studies: • Serum antibody response • Mucosal immune response • Health monitoring • Viral challenge • Histological assay • Virus detection in bursa

Study the temperature, pH and organic solvent tolerance window

Bioconjugation 1- Amin group coupling 2- Carboxyl group coupling

Page 29: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

23

II Materials and Methods II.1 Materials

Chemicals and consumables II.1.1All chemicals were supplied by following companies: Amersham Pharmacia Biotech (Freiburg), BioRad (München), Duchefa (Haarlem, Netherlands), Fluka (Steinheim), Gerbu (Gaiberg), Merck (Darmstadt), MWG-Biotech (Ebersberg), New England Biolabs (Schwalbach), Pharmacia (Uppsala), Pierce (Germany), Roche (Mannheim), Roth (Karlsruhe) and Sigma-Aldrich (Steinheim). The consumables were provided from: Amicon (Witten), Biozym (Oldendorf), Calbiochem (Darmstadt), Corning Inc. (Hamburg), Eppendorf (Hamburg), Gentaur (Aachen), Greiner (Solingen), Millipore (Eschborn), Pall (Dreieich), Qiagen (Hilden), Sarstedt (Nümbrecht), Sartoriusstedim (Göttingen), Schott Glaswerke (Mainz), Serva (Heidelberg), USB/Amersham (Braunschweig) and Whatman (Dassel).

Buffers, media and solutions II.1.2All standard buffers, media and stock solutions were prepared according to Sambrook and Russell standard protocols (2001) using deionized water. Compositions of non-standard solutions or buffers are listed at the end of respective method section. All media and some solutions were prepared and sterilized by autoclaving (25min/121°C/2bar). Heat-sensitive components, such as antibiotics, were prepared as stock solutions, filter-sterilized (pore size: 0.2 μm) and added to the medium after autoclaving following cooling down to 50°C. RPMI-1640 medium (Sigma) was used in hybridoma cell culture. Sabouraud agar medium (Heipha Dr Müller GmbH, Eppelheim Germany) was used as a yeast selection medium to test cell vitality.

Matrices and membranes II.1.3Ni-NTA superflow matrix (Qiagen) was used for purification of His-tag fused recombinant proteins by immobilized metal ion affinity chromatography (IMAC, II.2.2.4). Nanosep columns (Pall) with different cut-off membranes were used for ultra-filtration assays and protein concentrating (300, 100, 50, 10 kDa cut-off). Protran® nitrocellulose membrane from Whatman, HybondTM-C-nitrocellulose membrane (0.45 µm) from GE healthcare (Freiburg) and Whatman no.1 paper from Whatman were used in immunoblot analysis (II.2.3.3, II.2.3.4). Rotilabo syringe filters (0.45 and 0.22 μm) (Roth) were utilized for clearance and sterilization, respectively, in small volumes and PES bottle top filter (Sartoriusstedim) for big volumes. Vivaspin 20 ml centrifugal concentrators (Sartorius Stedim biotech) were used to concentrate the protein of interest. Analytical and preparative gel filtration matrices (Sephacryl and superdex) (GE healthcare) with different separation range were used for protein purification and analysis (II.2.2.5, II.2.3.5).

Enzymes and reaction kits II.1.4The restriction enzymes from New England Biolabs (NEB) (Schwalbach) were used for DNA digestion. Home-made Taq polymerase was used in colony-PCRs (II.2.1.20). In addition, the following kits were used:

Page 30: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

24

• Expanded high fidelity PCR system (Roche) • Nucleospin Plasmid kit (Machery-Nagel, Düren) for mini-preparation of plasmids • NucleoSpin

Extract II Kit (Machery-Nagel) for DNA fragment purification • CloneJETTM PCR cloning Kit (Fermentas, St. Leon-Rot) for fast and direct PCR product

cloning • BCA protein assay kit Pierce (Rockford, IL, USA) for protein concentration measurement • EzWayTM PAG protein elution kit (Koma Biotech, Seoul, South Korea) • Y-PER Yeast protein extraction reagent (Pierce) • DyLight 488 antibody labelling kit (Pierce) for fluorescent labelling of protein • AviPro Gumboro vac (Lohmann Animal Health GmbH & Co. KG, Cuxhaven) as

commercial IBD vaccine used as positive control • IDEXX IBD-XR Ab test (IDEXX, Westbrook, ME, USA) for antibody titer determination

in chickens

Antibodies and substrates II.1.5Rabbit anti-His polyclonal antibody (BioMol, Hamburg), mouse anti-IBD-VP2 (15F5 mAb) monoclonal antibody (Ingenasa, Madrid, Spain) and rabbit anti-IBD-VP2 polyclonal antibodies, kindly provided by prof. Wang (National Chung Hsing University, Taichung, Taiwan) and prof. Rodriguez (CNB, Spain), respectively, have been used as primary antibody in immunoassays including immunoblot (II.2.3.3, II.2.3.4), ELISA (II.2.3.1) or antibody-labelled electron microscopy (II.2.3.6). Goat anti-mouse Fc (GAM) and goat anti-rabbit Fc (GAR) antibodies, conjugated either to alkaline phosphatase (AP) or horseradish peroxidase (HRP) (Dianova, Hamburg) were used as a secondary antibody in immunoblotting and ELISA, respectively. Rabbit anti-chicken IgY (Fc)-HRP conjugate, goat anti-chicken IgA-HRP conjugate and goat anti-chicken IgM-HRP conjugate (Gentaur, Aachen) were used to study the chicken immune response (II.2.6.2). NBT/BCIP (BioRad) and ABTS (Roche) were used as substrate for detection of target immobilized proteins in immunoblot and ELISA, respectively. Goat anti-biotin gold (5nm in diameter)-labelled antibodies (Aurion, Wageningen, The Netherlands) were exploited in conjugation studies via electron microscopy (II.2.3.6). Alkaline phosphatase conjugated rabbit anti-xylose antibodies (Faye et al., 1993) was used in immunoblot for glycan analysis of proteins expressed in plants (II.2.4.1).

Special chemicals II.1.6Alkaline phosphatase conjugated Galanthus nivalis lectin (EY laboratories Inc., CA, USA) was used for glycan chain detection of the proteins expressed in yeast (II.2.4.1). Adjuvants, MM and 100 (Gerbu, Wieblingen) were used in parenteral immunization of mice and chicken, respectively. Sodium fluoride (NaF) from Sigma was also used in oral adjuvant mixture for chicken immunization.

Oligonucleotides II.1.7Oligonucleotides used for DNA amplification using polymerase chain reaction (PCR) (II.2.1.20), splicing by overlapping extension PCR (SOE-PCR) (II.2.1.20) and sequence analysis (II.2.1.21) are listed in the following tables. All oligonucleotides were synthesized by MWG (Ebersberg). In

Page 31: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

25

addition, an oligodeoxynucleotide (ODN) containing an unmethylated CpG dinucleotide with phosphorothioate backbone synthesized by Biolegio (Nijmegen, the Netherlands) was used as oral adjuvant in chicken immunization experiments (II.2.6). Table II.1 Primers used for generation of DNA fragments by PCR.

Table II.2 SOE-PCR primers for introducing extra DNA fragments into constructs or joining two separate constructs.

Primer name Restriction site at 5’ Primer sequence (5’→3’)

VP2-forward BspHI CCGTCATGACAAACCTGCAAGATC

VP2-reverse-withHis6 XbaI GCTCTAGATTTAGTGATGGTGATGGTGATGTGCTCCTGCAATCTTCAG

Solulin-for5´UT EcoRI CGCGAATTCACAACACAAATCAGATTTATAGAGAGATTTATAAAAAAAAAAAAAAC

VP2-reverse - ATTAGACTCGACCGCTCCTGCAATCTTCAG

CytB5memAnch.-for - CTGAAGATTGCAGGAGCGGTCGAGTCTAATTCC

vIL-10_XbaI-bw XbaI GCTCTAGACGGTTTAATCTTCTGC

Primer name Overhang region (5’→3’) Restriction

site at 5’

Primer sequence (5’→3’) including overhang

region

CHS-elongation5’ TTTATAAAAAAAAAA

AAAAC -

TTTATAAAAAAAAAAAAAACAATGACAAACCTG CAAGATCA

Solulin-for5´UT TTTATAAAAAAAAAA

AAAAC EcoRI

CGCGAATTCACAACACAAATCAGATTTATAGAGAGATTTATAAAAAAAAAAAAAAC

CHS-elongation5’primer-withHis6

TTTATAAAAAAAAAA AAAACAATG

-

TTTATAAAAAAAAAA AAAACAATGCATCAC CATCACCATCACACA AACCTGCAAGATCA

VP2-reverse ATTAGACTCGACCGCT

CCTGCAATCTTCAG -

ATTAGACTCGACCGCTCCTGCAATCTTCAG

CytB5memAnch.-for CTGAAGATTGCAGGAGCGGTCGAGTCTAAT

- CTGAAGATTGCAGGAGCGGTCGAGTCTAATTCC

vIL-10_XbaI-bw - XbaI GCTCTAGACGGTTTAATCTTCTGC

Page 32: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

26

Table II.3 Primers used for DNA sequencing.

Table II.4 Phosphorothioate CpG ODN used as adjuvant in oral immunization of chickens.

Vectors II.1.8Schematic maps of all plasmids used for tobacco and Pichia transformation are shown in Appendix VII-2. pUC19 (Invitrogen) was used for E. coli efficiency of transformation. The vector pJET1.2 from Fermentas was used as an intermediate vector for all sub-cloning purposes prior to sequencing. The vector contains a beta-lactamase (bla) gene for selection with ampicillin. The pPICZ_B vector (Invitrogen) was used for expression of recombinant proteins in Pichia pastoris. This vector contains a methanol inducible AOX1 promoter for high-level expression in P. pastoris, a c-myc epitope tag for detection of target proteins with an anti-myc antibody, a C-terminal poly-histidine (His6 or in brief H6) tag for rapid purification with nickel-chelating resin and/or detection with an anti-His antibody and a Zeocin™ resistance gene (Sh ble) under both EM7 (synthetic prokaryotic promoter) for positive selection of recombinant E. coli and TEF1 (Transcription elongation factor 1 gene) promoter for positive selection of yeast. The vector pTRAkc (Sack et al., 2007) was used for transformation of the tobacco plants via Agrobacterium-mediated transformation (II.2.2.3, II.2.2.4). pTRAkc is an optimized plant expression vector which in this case contains the Cauliflower mosaic virus 35S promoter, the 5’ untranslated region of the Chalcone synthase gene from Petrosinella and the 3’ pA35S untranslated region from CaMV. Scaffold attachment regions (SAR) were introduced next to the right and left border of T-DNA to improve gene expression. On the T-DNA part of this vector the kanamycin (nptII) resistance gene as a plant selection marker and on the vector backbone ampicillin (bla) resistance gene as a bacterial selection marker are present.

Bacterial, yeast and virus strains II.1.9E. coli strains DH5α and XL1-Blue were used as host cells for all intermediate cloning constructs (Table II.5). P. pastoris X-33 cell line (wild type, Mut+) from Invitrogen was used for expression of recombinant IBD-VP2.

Primer name Primer sequence (5’→3’)

VP2Primer1_forward TGGTAGCAACATGTGACA

VP2Primer1_reverse TTGAAACACGAGTTCTCC

pSS5′ ATCCTTCGCAAGACCCTTCCTCT

pSS3′ AGAGAGAGATAGATTTGTAGAGA

pJET1.2Forward CGACTCACTATAGGGAGAGCGGC

pJET1.2Reverse AAGAACATCGATTTTCCATGGCAG

Name sequence (5’→3’)

CpG ODN TCGTCGTTGTCGTTTTGTCGTT

Page 33: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

27

A. tumefaciens GV3101 (pMP90RK GmR, KmR, RifR (Koncz and Schell, 1986) was used for Agrobacterium-mediated gene transfer. For the challenge, a classical IBDV strain, MB3, from a recent outbreak in Germany was used. Table II.5 Names, suppliers and genotypes of E. coli strains used throughout the work.

Plants and animals II.1.10Nicotiana tabacum L. cv. Petite Havana SR1 was used for transient and stable protein expression (II.2.2.2, II.2.2.3) of IBD-VP2 after vacuum infiltration with recombinant agrobacteria. Six to eight week-old female BALB/c mice were used for immunogenicity studies (II.2.5). Two-week old chickens hatched from specific pathogen free eggs (Lohmann Tierzucht, Cuxhaven) were used for vaccine analysis (II.2.6).

Equipment and applications II.1.11Cameras: MP4 (Polaroid, Cambridge, MA, USA). E.A.S.Y 429K camera (Herolab, Wiesloch). Cell counter: Cell counter CASY 1 TT (Roche Innovatis)

Centrifuges: AvantiTM

30 and AvantiTM

J-25 from Beckman (California, USA), Biofuge A from Heraeus (Hanau), Eppendorf 5415D. Rotors: F0650, F2402H, JLA 10.500 and JA 25.50 from Beckman, #1140 and #11222 from Sigma, RLA-300, SS-34 and GS-3 from DuPont Instruments (Bad Homburg). Clean bench: Kojair (Nideggen). Chromatography equipment: ÄKTA explorer 10xT (Amersham Pharmacia Biotech), Gel filtration HiPrep 26/60 (column 26 mm inside diameter, 20 cm length) Superdex 200 and Sephacryl S-400 HR (GE healthcare, Freiburg, Germany) and UNICORN control, evaluation and documentation software (GE healthcare, Freiburg, Germany).

DNA-sequencing machine: 3700 DNA analyzer and BigDyeTM

cycle-sequencing terminator chemistry (V 3.0) from Applied Biosystems. DNA gel electrophoresis apparatus: Wide mini and mini cells for DNA agarose electrophoresis from BioRad, and the power suppliers from BioRad. Dynamic light scattering apparatus: Koheras (Kleinostheim) Electron microscopy: 400T electron microscope (Philips, Eindhoven, The Netherlands) Electroporation apparatus: Multiporator from Eppendorf and 0.2 cm or 0.4 cm cuvettes from BioRad. ELISA reader: Synergy HT from Bio-Tek Instruments (Bad Friedrichshall). ELISA washer: Elx405 from Bio-Tek Instruments.

Strain Source Genotype

DH5α Invitrogen F- (f80d Lac 2ΔM15) Δ(LacZYA-argF) U169end A1 rec1hsdR17(rk-

mk+) deoR thi-1 supE44 gyrA96 relA1 λ-

XL1-Blue Stratagene recA1 endA1 gyrA96 thi-1 hsdR17 supE44 relA1 lac [F′ proAB laclIq

ZΔM15 Tn10 (Tetr)]

Page 34: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

28

Freeze-drying machine (Lyophilizer): Alpha 1-4 LSC (Company Christ, Osterode) combined with vacuum pump RZ6 (Vacuubrand, Wertheim). Incubators: CO2 Incubator CB Series (Binder, Tuttlingen)

InnovaTM

4340 incubator shaker: New Brunswick scientific (Nürtingen). Microfluidizer: M-110L from Microfluidics (Newton, MA, USA). Micromass Q-TOF micro Mass spectrometer equipped with a standard electrospray unit with variable in-source pressure, MALDI source, connected to an Ultimate 3000 Cap-LC system (Biobasic HPLC C18 column, Dionex). Microscopes: Leica (Wetzlar). PCR Thermocyclers: Primus and Primus 96 plus from MWG-Biotech, Mastercycler Personal from Eppendorff, DNA Engine from MJ Research Inc (Watertown, USA). Photometers: Spectrophotometer Uvikon 930 from Kontron (Neufahrn) and Cuvettes # 67.742 from Sarstedt (Nümbrecht). NanoDrop, ND1000 from Thermo Scientific. Probe sonicator: Braun Biotech (Melsungen).

Protein gel electrophoresis equipment: Mini PROTEAN IITM

from Bio-Rad. Software: Windows XP operating system (Microsoft); Microsoft Office 2010 (Microsoft); Adobe Photoshop CS2 (Adobe); Clone manager suite 8; Vector NTI advance 10; AIDA image analyzer v.4.14; MassLynx 4.0 SP4 Software (Waters Micromass), CLC Main workbench 6.5, EndNote X1; PyMOL v 0.99 and Chemdraw 12. UV-Transilluminators: Wavelength 302nm and UVT-20M from Herolab (Wiesloch). UV-chamber from BioRad. Vacuum filtration unit: Sartorius (Göttingen).

II.2 Methods

Recombinant DNA technologies II.2.1All experiments related to genetic engineering were performed according to the regulations of “S1-Richtlinien” General recombinant DNA techniques, i.e. DNA precipitation, restriction enzyme digestion, DNA ligation, DNA agarose gel electrophoresis, were performed according to the standard protocols described in Sambrook and Russell (2001).

Preparation of heat-shock competent E. coli II.2.1.1A single colony of E. coli strain of either DH5α or XL1-Blue grown on LB-agar plate was inoculated into 5 ml LB medium in 15 ml falcon tube and incubated overnight (o/n) at 37ºC. Two ml of the o/n culture was transferred into 200 ml of LB medium. The cells were cultured at 37°C for 3-4 hours until the OD600 nm reached 0.4-0.5 and then transferred to an ice-cold tube. After placing on ice for 10 min, the cells were recovered by centrifugation (3500 ×g/4°C/3 min). The supernatants were decanted and the pellets were resuspended gently in 10 ml ice-cold 0.1 M CaCl2 and kept on ice for 20 min. The cells were recovered by centrifugation as described above and resuspended in 3 ml ice-cold 0.1 M CaCl2 /15% (v/v) glycerol. One hundred μl-aliquots of the suspension were dispensed into pre-chilled 1.5 ml microcentrifuge tubes, frozen immediately in liquid nitrogen and stored at -80°C.

Page 35: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

29

Transformation of E. coli by heat-shock II.2.1.2Frozen aliquots (100 µl) of the heat-shock competent E. coli cells (II.2.1.1) were thawed on ice for 5-10 min and mixed gently either with plasmid DNA (up to 200 ng) (II.2.1.12) or ligation products (II.2.1.19) as described by Sambrook and Russell (2001). Together with DNA, the cells were stored on ice for 30 min. The cells were then suddenly exposed to 42°C for 90 seconds and immediately placed on ice for further 2 min. Then, 800 μl of pre-warmed (37°C) LB medium were added to the tubes and incubated at 37°C for 1 h. After incubation 200 μl of cells were plated onto a LB-agar plate supplemented with appropriate antibiotics and were incubated at 37°C over night.

Preparation of electrocompetent E. coli II.2.1.3Electrocompetent E. coli were prepared from strains DH5α and XL1-blue as described by Dower et al. (1988). A single bacterial colony from an LB plate was inoculated in 5 ml LB-medium and cultured at 37°C o/n. Five ml of fresh o/n culture was transferred into 500 ml of LB medium. The cells were incubated at 37°C for 3-4 hours until the mid-log phase (OD600 nm= 0.5-0.6). Then the cells were placed on ice for 15-20 min and harvested by centrifugation (3000 ×g/4°C/10 min). Cells were washed three times with sterile cold water while the water amount was reduced to half in each washing time and resuspended in ice-cold 10% (v/v) glycerol to a 300-fold concentration from the original culture volume (at >1010 cells/ml). Sixty μl aliquots were frozen directly in liquid nitrogen and stored at -80°C.

Transformation of E. coli by electroporation II.2.1.4Transformation via electroporation was accomplished according to the protocol provided by Stratagene. Frozen aliquots of electrocompetent E. coli cells (II.2.1.3) were thawed on ice for 5-10 min and were mixed with either 100 pg of plasmid DNA or 100-200 ng of desalted ligation product (desalting was performed using desalting membrane). The cell/DNA mixture was transferred into a prechilled sterile electroporation cuvette (0.2 cm or 0.1 cm gap) and put into a safety chamber. After application of a 5 ms electric pulse (2.5 kV for 0.2 cm and 1.7 kV for 0.1 cm gap electrocuvette) the cells were immediately diluted with 900 µl of pre-warmed (37°C) SOC medium and incubated at 37°C with shaking for 1 h. Finally, 200 μl of the cells were plated onto LB-agar plates containing appropriate antibiotics and incubated at 37°C for o/n.

Preparation of electrocompetent A. tumefaciens II.2.1.5

A single colony of A. tumefaciens strain GV3101 grown on a YEB-agar plate containing 25 µg/ml rifampicin (Rif) and 25 µg/ml kanamycin (Km) (YEB-Rif-Km) was used to inoculate 10 ml of YEB-Rif-Km medium in a 100 ml Erlenmeyer flask and incubated at 28°C for two days with shaking (160 rpm). One ml of the culture was transferred into 100 ml of YEB-Rif-Km medium and cultivated at 28°C for 15-20 h with shaking (160 rpm) until the OD600 nm reached 1-1.5. After that the cells were kept on ice for 15 min and harvested by centrifugation (4000 ×g/4°C/5 min). The culture medium was decanted and the cells were sequentially washed three times with 100 ml of ice-cold ddH2O and one time with 100 ml of ice-cold sterile 10% (v/v) glycerol. After each washing step the cells were spun down by centrifugation (4000 g/4°C/5 min) and finally were resuspended in 500 µl of sterile 10% (v/v) glycerol. Around 50 µl aliquots of the suspension were

Page 36: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

30

dispensed into prechilled 1.5 ml microcentrifuge tubes, frozen immediately in liquid nitrogen and stored at -80°C. YEB-Rif-Km medium:

Nutrient broth 0.5% (w/v) Yeast extract 0.1% (w/v) Peptone 0.5% (w/v) Sucrose 0.5% (w/v)

The following compounds were added after autoclaving and cooling to 55°C: 2 mM MgSO4, 25 µg/ml rifampicin and 25 µg/ml kanamycin.

Transformation of A. tumefaciens by electroporation II.2.1.6

About 0.2-0.4 µg of plasmid DNA (II.2.1.12) in sterile ddH2O was added to a thawed aliquot of electrocompetent agrobacteria cells (II.2.1.5) and placed on ice for 5 min. The cell/DNA mixture was transferred into a prechilled electroporation cuvette (0.2 cm) and put into a safety chamber. After application of the pulse (2.5 kV, 5ms), the cells were diluted in 900 µl of SOC medium in a 15 ml falcon tube and incubated at 28°C for 1 h with shaking (160 rpm). Finally, 1-10 µl of the cells were plated on YEB-agar containing 25 µg/ml rifampicin (Rif), 25 µg/ml kanamycin (Km) and 50 µg/ml carbenicillin (Carb) (YEB-Rif-Km-Carb) and incubated at 28°C for 2-3 days. As a negative control, transformation of Agrobacterium cells with H2O was performed.

Determination of the transformation efficiency of competent bacteria II.2.1.7The efficiency of transformation for each new batch of competent cells was assessed by test transformations with known concentrations (100 pg) of supercoiled plasmid pUC19 for E. coli and pTRAkc for A. tumefaciens cells. The following equation was used to calculate the transformation efficiency (CFU/µg): CFU on control plate × 1 × 106 pg × volume of transformants × dilution factor pg plasmid DNA μg volume plated

CFU: Colony forming unit. The transformation rates were obtained; for heat-shock competent E. coli >106/µg pUC19, electrocompetent E. coli >108/µg pUC19 and A. tumefaciens >103/µg pTRAkc.

Transformation of P. pastoris by electroporation II.2.1.8Transformation of P. pastoris was accomplished according to the method published by Wu and Letchworth (2004) with some modifications. A single colony of wild type P. pastoris strain X-33 grown on YPD-agar plate was selected to inoculate 20 ml YPD medium in a 100 ml Erlenmeyer flask and incubated at 28°C for 24 h with shaking (160 rpm). Around 20 µl of yeast culture were transferred to 100 ml of fresh YPD medium and incubated at 28°C until the cell density reached OD600 nm = 1-2. The cells were collected by centrifugation (1500 ×g/4°C/5 min). The number of the

Page 37: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

31

cells was calculated according to the equation of 1 OD600 nm = 5 × 107 cells/ml (EasySelectTM Pichia expression kit user manual, Invitrogen). For each transformation, 8 × 108 cells were suspended at room temperature for 30 min in 8 ml of 10 mM Tris-HCl containing 100 mM LiAc, 10 mM DTT, 0.6 M sorbitol, pH 7.5. The cells were then pelleted by centrifugation (1500 ×g/4°C/5 min), resuspended in 1.5 ml of ice-cold 1 M sorbitol and were transferred to a 2 ml microcentrifuge tube. Then, the cells were washed three times with 1.5 ml ice-cold 1 M sorbitol and resuspended in 1 M ice-cold sorbitol at a final concentration of about 1010 cells/ml. After that 100 µl of those cells were mixed with 3-5 µg of linearized plasmid DNA (II.2.1.15, digested by SacI) in 1-10 μl of water, transferred to a prechilled electroporation cuvette (0.2 cm gap) and stored on ice for 5 min. Then the electroporation pulse (1.5 kV, 5 ms) was applied and the cells were immediately diluted in 1 ml of ice-cold 1 M sorbitol. The cells were incubated at 28ºC for 1-2 h without shaking. Finally, 50-200 μl aliquots were spread on YPDS plates containing 100 μg/ml of Zeocin™. The plates were then incubated at 28°C for 3–6 days until colonies formed. The growing colonies from the Zeocin™ containing plates were streaked on both MD (minimal dextrose) and MM (minimal methanol) plates to screen for Mut+ (methanol utilization plus) strains as described (EasySelectTM Pichia expression kit user manual, Invitrogen). YPD:

Yeast extract 1% (w/v) Peptone 2% (w/v) Dextrose (D-glucose) 2% (w/v)

YPDS: Yeast extract 1% (w/v) Peptone 2% (w/v) Dextrose 2% (w/v) Sorbitol 1 M

MD: YNB 1.34% (w/v) Biotin 4×10–5% (w/v) Dextrose 2% (w/v)

MM: YNB 1.34% (w/v) Biotin 4×10–5% (w/v) Dextrose 2% (w/v) Methanol 0.5% (v/v)

Culturing of E. coli and glycerol stock preparation II.2.1.9Individual colonies of all strains were obtained by plating the corresponding strain on LB agar plates. For recombinant bacterial selection, LB agar containing 100 µg/ml ampicillin were used. Incubation was performed at 37°C. The plates were stored at 4oC for short periods (less than 2 weeks). For long term storage, 5 ml LB medium containing the suitable antibiotics and 2% (w/v)

Page 38: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

32

glucose was inoculated with a single recombinant colony and grown overnight at 37°C with vigorous shaking (160 rpm). Glycerol stocks were prepared by mixing 800 µl of a fresh overnight culture with 200 µl of 80% (v/v) sterile glycerol in water. Bacteria glycerol stocks were stored at -80°C.

Culturing of recombinant A. tumefaciens and glycerol stock preparation II.2.1.10Positive recombinant Agrobacterium colonies containing the plasmid of interest were inoculated in 10 ml of YEB-Rif-Km-Carb medium and cultivated at 28°C for 2 days with vigorous shaking (160 rpm). The culture was transferred to 15 ml falcon tubes and cells were collected by centrifugation at 4,000 g for 10 min at 15°C. The cells were resuspended in a 1:1 volume of fresh YEB Rif-Km-Carb medium and glycerol stock media (GSM). The suspension was stored at -80°C in 500 µl aliquots and used for further experiments. Glycerol stock medium (GSM):

Glycerol 50% (v/v) MgSO4 100 mM Tris-HCl, pH 7.4 25 mM

Culturing of recombinant P. pastoris and glycerol stock preparation II.2.1.11A single colony of each recombinant strain was cultured in 20 ml YPD containing 100 µg/ml ZeocinTM for 2 days at 28°C with vigorous shaking (160 rpm). Then 1 ml of that culture was used to inoculate 100 ml of YPD containing 100 µg/ml ZeocinTM and cultivated for o/n at 28°C with shaking (160 rpm). Next day the cells were harvested by centrifugation (1500 g/4°C/5 min) and resuspended in fresh YPD containing 15% (v/v) glycerol with a final cell concentration OD600 nm of 50–100 (1 OD600 nm = 5 × 107 cells/ml). Finally, 1 ml of cell aliquots were frozen in liquid nitrogen and then stored at -80°C for further experiments.

Isolation of plasmid from recombinant E. coli II.2.1.12Plasmids were isolated in high quality from relevant recombinant E. .coli based on the alkaline lyses method (Sambrook and Russell, 2001) using Nucleospin Plasmid kit Mini and Midi (Machery-Nagel) according to the manufacturers’ instructions. Purity and yield of plasmid DNA was checked by reading the UV absorbance at 260 and 280 nm via NanoDrop spectrophotometer according to Sambrook and Russel (2001) or by analytical agarose gel electrophoresis (II.2.1.13). Isolated DNA samples were stored at -20°C and used for cloning and sequencing purposes.

Analytical agarose gel electrophoresis of DNA II.2.1.13Plasmid DNA (II.1.8), PCR fragments (II.2.1.18) and enzymatically digested DNA (II.2.1.15) were electrophoretically separated on 0.8-1.2% (w/v) agarose gels prepared in TBE buffer containing 0.1 μg/ml ethidium bromide as described by Sambrook and Russel (2001). Known amount of the DNA molecular marker such as 1 kb (Fermentas) or 100 bp (Roth) DNA ladder were used for evaluation of sample size, integrity and determination of DNA concentration. The DNA was

Page 39: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

33

visualized on an UV transilluminator by illumination at 302 nm. The gel pictures were documented by using a universal hood II for Gel Doc XR (BioRad). 10X TBE electrophoresis buffer (pH 8.3):

Tris-base 900 mM Boric acid 900 mM EDTA 25 mM

Preparative agarose gel electrophoresis of DNA II.2.1.14Preparative gel electrophoresis was used for isolation of DNA fragments after restriction enzyme digestion (II.2.1.15) or PCR-amplified DNA (II.2.1.18) prior to cloning into an appropriate vector. After electrophoresis, the desired DNA fragments were excised from the gel and purified using the “NucleoSpin

Extract II Kit l” (Machery-Nagel) according to the manufacturers’ protocol.

Restriction digest of DNA and preparative agarose gel electrophoresis II.2.1.15Restriction endonucleases, appropriate buffers and BSA solution were obtained from New England Biolabs. Single or double restrictions of DNA fragments were performed at suitable buffer and temperature according to the manufacturers’ instruction.

Quantification of DNA II.2.1.16The concentration of plasmids and DNA fragments were quantified using NanoDrop spectrophotometer according to the manufacturers’ instruction. In order to know the proper amount of DNA in ligation mixture, analytical agarose gel electrophoresis (II.2.1.13) followed by measurement of band intensity using AIDA software (according to the manufacturers’ instruction) was performed.

Ligation of the DNA fragments II.2.1.17Digested DNA fragments (II.2.1.15) including insert and vector were ligated together in 3:1 ratio in order to obtain the desired vector by using 80 U of T4 DNA ligase or 1 µl Quick T4 DNA ligase (NEB) in buffer systems recommended in the manufacturers’ protocol. The final volume of ligation mixture was adjusted to 20 µl. Sticky and blunt end ligations were carried out at 22°C for 30 min or longer (up to overnight). Ligation product was used for E. coli transformation (II.2.1.2, II.2.1.4).

PCR amplification II.2.1.18Polymerase chain reaction (PCR) was used for amplification of IBD-VP2 from the TA-IBD-VP2 kindly provided by Dr. Haleh Hashemi (National institute of genetic engineering and biotechnology, Tehran, Iran). The vector containing the cDNA sequence of IBDV-VP2 (NCBI code: AY704912). The PCR product containing the gene encoding the mature VP2 (441 aa) was used as a template in PCR for generation of other derived cDNA constructs. The DNA fragments encoding the cytochrome b5 membrane anchor was generated using pTRAkc-Cyt-m/vIL-10 (Bortesi et al., 2009) as template in PCR. DNA amplification was carried out based on the protocol of Sambrook and Russell (2001), using ExpandTM high fidelity PCR system (Roche). The reactions

Page 40: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

34

were performed in 0.2 ml PCR reaction tubes using a DNA thermal cycler (MWG-Biotech). The cycler contained a heated lid to prevent evaporation of the reaction mixture. For screening of recombinant E. coli and agrobacteria, colony-PCR was performed to detect plasmids as described by Jesnowski et al. (1995). Isolated colonies were picked using sterile tips and suspended in 20 µl of sterile distilled water in PCR tubes. The suspension was boiled for 10 min and cell debris was spun down by centrifugation (13000 g/RT/5 min). Finally, 10 µl of the supernatant was used for PCR reactions in a total volume of 25 µl. The optimal annealing temperature (Tm) of the primers was experimentally optimized or calculated based on the empirical formula of Wu et al. (1991): Tm = {22 + 1.46 [2× (G + C) + (A + T)]} PCR reactions were performed in a total volume of 25 µl as described below: Table II.6 PCR master mix.

Amplification was performed under the following conditions: Step 1: 5 min 95°C Step 2: 1 min 95°C Step 3: 1 min 54°C Step 4: 1 min 72°C Step 5: 10 min 72°C The annealing temperature (step 3) and the time for extension (step 4) were changed according to Tm value of primers and the length of the target gene respectively. SOE-PCR (Horton et al., 1989) was exploited to introduce either extra sequence to a DNA fragment or to fuse two DNA fragments together. In the first case, a primer (forward) containing a matching sequence at the 3’ end with the template DNA fragment together with the backward primer was used to amplify the template for 5 cycles as mentioned above. The forward primer had an extra sequence at the 5’ end. By this way the extra sequence was introduced to the PCR

Components Volume Final concentration

10x PCR buffer 2.5 µl 1X

50 mM MgCl2 1.0 µl 1.5 mM

10 mM dNTPs 0.5 µl 0.2 mM each

1.5 pmol forward Primer 2.0 µl 0.12 pmol

1.5 pmol backward primer 2.0 µl 0.12 pmol

Template DNA 2.0 µl 0.1-10 ng

Taq DNA polymerase (5U/ml) 0.25 µl 1.25 units

dd H2O to 25 µl

30 X

Page 41: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

35

products. Then the next primer was added to the PCR mixture. This primer had a matching sequence at the 3’ end with the forward primer and could also carry an extra sequence at the 5’ end which again leads to the introduction of extra sequences to the primary PCR product. Performing PCR using this primer and the backward primer elongated the PCR product and led to introduction of the desired sequence. For fusion of two sequences, initially two separate PCR were performed. Then, the SOE-PCR was carried out by mixing equimolar amounts of two overlapping PCR fragments in a total reaction volume of 25 µl (2.5 µl 10x PCR buffer, 1.75 mM MgCl2, 20 pmol of each dNTP, and 0.2 units ExpandTM high fidelity DNA polymerase mix) under the following conditions: an initial denaturation for 5 min at 95°C followed by 4 cycles of 95°C for 1 min, 37°C for 1 min and 72°C for 1 min. Then the reaction mixture from the SOE-PCR was mixed with 25 µl of PCR cocktails containing 2.5 µl 10x PCR buffer, 1.75 mM MgCl2, 20 pmol of each dNTP, 10 pmol of forward and backward terminal primers, 0.2 units ExpandTM high fidelity DNA polymerase mix and again another normal PCR was performed as mentioned above. The performance of each PCR reaction was checked by running 5 μl of each reaction on agarose gels (II.2.1.13) with appropriate DNA markers.

DNA sequencing II.2.1.19Dye termination sequencing based on chain termination method described by Sanger et al. (1977 was performed using the Applied Biosystems 3700 DNA analyzer and BigDyeTM terminator sequencing kit. Chromas software package (Technelysium Pty Ltd) was used for displaying the chromatogram files. For evaluation of sequencing data the Vector NTI advance10 software package and Clone manager software professional suit version 8 (Sci-Ed software) were used.

Expression and purification of recombinant protein II.2.2Recombinant proteins were transiently expressed in tobacco leaves (II.2.2.3). They were also stably expressed in tobacco (II.2.2.4) and yeast cells (II.2.2.6). Recombinant proteins were extracted, purified and characterized (II.2.2.5, II.2.3).

Growth and maintenance of tobacco plants II.2.2.1Tobacco seed plants were cultured and grown in a greenhouse in ED73 standard soil (Patzer, Sinntal-Jossa) with 0-30% (v/v) sand under supplementary illumination of 10000 Lux (plus the sun light), 70-90% humidity and 16 h light/ 8 h darkness photoperiod at 24°C. To prevent pollination from other plants, flowers were covered with plastic bags with micropores. Mature, dried seeds were stored in paper bags at RT.

Preparation of recombinant A. tumefaciens II.2.2.2Recombinant A. tumefaciens (II.2.1.10) were grown followed by vacuum infiltration of tobacco leaves according to Kapila et al. (1997) and Vaquero et al. (1999) with minor modifications. Inoculation of recombinant agrobacteria was initiated by taking 50 µl of frozen cells from the glycerol stock (II.2.1.10) into 5 ml of liquid YEB-Km-Rif-Carb medium in a 15 ml falcon tube. The culture then incubated at 28°C o/n with shaking at 160 rpm. Next day, a fraction of the culture was inoculated 1:100 into fresh YEB-Km-Rif-Carb medium in a 250 ml Erlenmeyer flask. The culture was incubated o/n as described above. The day after, filter sterile acetosyringone (200

Page 42: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

36

mM), 1 M glucose and 1 M MES (pH 5.6) were added to the culture to a final concentration of 20 µM, 10 mM and 10 mM, respectively. The incubation was continued o/n. Next day, the OD600nm

was measured; calculated amount of cells were harvested by centrifugation (4000 ×g/RT/15 min) and resuspended in infiltration medium until the OD600 nm reach 1.0. The culture was incubated at RT for 2 h. The cell suspension was used for vacuum infiltration of cultivated tobacco plant leaves (II.2.2.2). YEB-Km-Rif-Carb:

Nutrient broth 0.5% (w/v) Yeast extract 0.1% (w/v) Peptone 0.5% (w/v) Sucrose 0.5% (w/v)

2 mM MgSO4, 25 µg/ml kanamycin, 25 µg/ml rifampicin and 50 µg/ml carbenicillin were added after autoclaving and cooling to 55°C. Infiltration medium:

MS basic salt mixture 0.43% (w/v) Glucose 20 mM Sucrose Acetosyringone

10% (w/v) 200 µM

Vacuum infiltration and/or injection of tobacco leaves with recombinant II.2.2.3agrobacteria

N. tabacum cv. Petite Havana SR1 plants were cultivated in the greenhouse (II.2.2.1). Top leaves from young plants were harvested and placed into a stainless steel beaker with 200 ml of agrobacteria cell suspension (II.2.2.2) and fixed with a mortar to allow complete covering of the leaves with the cell suspension. A continuous vacuum (60-80 mbar) was applied for 25 min. After removing the vacuum, the leaves were briefly rinsed in tap water and kept on wet Whatman paper no.1 with adaxial side upwards. The plastic tray was sealed with saran wrap and placed at 22°C with a 16 h photoperiod (7500 lux) for 3-4 days. Leaves were weighed, frozen in liquid nitrogen and stored at -80°C until use. As a negative control, tobacco leaves were infiltrated with agrobacteria suspension, which harbor a pTRAkc plasmid but lack the gene of interest (empty vector control). As an alternative for small-scale expression and comparisons, agro-injection was employed. Briefly, agrobacterium suspension was applied directly into mesophyll space of leaf cells by injection through an invented spot wound using a syringe without needle. This method provides rapid expression analysis in natural form without separating leaves from plant and for longer investigation period. The injected plants were placed at 22°C with a 16 h photoperiod for 3 days.

Page 43: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

37

Recombinant Agrobacterium-mediated stable transformation of Nicotiana tabacum II.2.2.4Wild type seeds of N. tabacum L. cv. Petite Havana SR1 were grown on MS I medium in weck glasses. The youngest leaves were harvested and used for transformation. They were cut into small pieces (0.5-1 cm2) after removal of the central vein. Then, they were transferred into sterile glasses containing 100 ml of recombinant Agrobacterium suspension (OD600 nm = 1.0) and incubated at room temperature (RT) for 10-15 minutes. The leaf pieces were then transferred onto sterile water-wetted Whatman papers in Petri dishes. They were incubated at 23-25 °C in dark for 2 days. Afterwards, they were moved onto MS II plates and incubated at 23-25 °C in 16 h light and 8 h darkness photoperiod for 3-4 weeks. After shooting, shoots were cut and moved onto MS III plates and incubated at the same condition like before for 10-14 days until roots emerged. The transgenic plantlets (T0) were transferred into weck glasses containing MS III medium and incubated at the same conditions. Two weeks later, they were ready to transfer to soil. They were grown in standard cultivation soil, ED73 (Patzer KG, Sinntal-Jossa) with 0-30% (v/v) sand under the following conditions: 16 h artificial light, 25°C (or higher depending on the outside temperature), 10,000 lux (plus the sun light) and 70-90% humidity. Young leaves of regenerated plants were used for screening analysis as previously described (II.2.2.4, II.2.2.5). To prevent pollination from other plants, flowers of designated plants were covered with plastic bags with micro pores to induce self-pollination. Mature, dried seeds (T1) were stored in the seed bank in paper bags at RT. MS I medium:

MS basic salt mixture 0.44% (w/v) Sucrose 2.0% (w/v) Thiamine-HCl 0.2 mg/l

The pH was adjusted to 5.8 with 1 N NaOH (for preparation of solid medium, 0.8% (w/v) agar was added). The medium was autoclaved and 500 μl of vitamin solution I were added upon cooling to 55 °C. MS II medium: MS medium supplemented with:

BAP 1 mg/l NAA 0.1 mg/l Kanamycin Claforan Betabactyl Agar

100 mg/l 100 mg/ml 200 mg/ml 0.8% (w/v)

MS III medium: MS medium supplemented with:

Kanamycin 100 mg/l Claforan Betabactyl Agar

200-250 mg/l 200-250 mg/l

0.8% (w/v)

Page 44: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

38

Vitamin solution I: Glycine 0.4% (w/v) Nicotinic acid Pyridoxine

0.1% (w/v) 0.1% (w/v)

Filter sterilized and stored at 4°C.

Expression and purification of IBD-VP2 from tobacco leaves II.2.2.5Extraction of either transiently produced IBD-VP2 from vacuum infiltrated tobacco leaves (II.2.2.3) or transgenic tobacco plants (II.2.2.4) were performed by grinding the leaves in a kitchen blender in companion with 2 ml of extraction buffer per gram leaf material. Cell debris was removed by centrifugation (15000 ×g/4°C/20 min) and the supernatant was used in SDS-PAA gel electrophoresis followed by either Coomassie staining (II.2.3.2) or immunoblot (II.2.3.3 and II.2.3.4). For purification, the leaves were grinded as above in companion with purification buffer with pH 4.0. After centrifugation as above, the clarified extract was passed through double layer of miracloth filter. Then the pH was adjusted to 8.0 and NaCl was applied up to a final concentration of 1 M. Following gently stirring at 4°C for 1 h, one more round of centrifugation was performed as above. Then the supernatant was used for purification of IBD-VP2 via IMAC. The clarified extract was loaded on a pre-equilibrated Ni-NTA resin column. Pre-equilibration of the column resin was performed by washing 1 ml of resin with 20 column volumes of PBS (phosphate buffer saline). The clarified extract was passed through the column with 1 ml/min flow rate. The resin was washed with 20-30 column volumes of PBS and the protein was eluted by 2-3 ml of elution buffer (PBS) containing 250 mM imidazole. The elution fractions were mixed and separated 12% (w/v) SDS-PAA gel. The target band for the protein of interest was excised from the gel and the protein content was recovered using EzWayTM PAG protein elution kit (Koma Biotech.) according to the manufacture’s instruction. Protein extraction buffer:

NaPO4 pH 7.0 50 mM NaCl 500 mM Na2S2O5

Tween 20 10 mM

0.5% (v/v) Purification buffer (pH 4.0):

Acetate buffer 100 mM Ascorbic acid 50 mM β-mercaptoethanol 5 mM Tween 20 0.5% (v/v)

As an alternative purification method, ultracentrifugation was exploited. About 15 gram transiently transformed tobacco leaves were grinded while mixed with 45 ml (1:3 ratios) extraction buffer (25 mM NaPO4 pH 7.0, 100 mM NaCl, 50 mM ascorbic acid, pH 5.3). The extract was passed through

Page 45: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

39

two layers of Mira-cloth filter. Two tablets of complete EDTA-free protease inhibitor cocktail (Roche) were added for each 45 ml of extract. The sample were incubated at RT for 1 hour and then centrifuged at 27,261 ×g for 30 min to remove the plant debris. To concentrate the extracts, 40% ammonium sulphate (226 g/l) was gradually added to dissolve rapidly avoiding accumulation of undesirable high local salt concentrations. Then centrifugation at 50000 ×g for 30 min at 4°C was carried out. The pellet were resuspended in 4 ml buffer (25 mM NaPo4 pH 7.0, 100 mM NaCl, pH 6.8) and loaded on prepared sucrose gradient tubes. The sucrose gradient was generated by tandem layering of 4.7 ml from 60, 50, 40, 30, 20 and 10% (w/v) sucrose dissolved in extraction buffer (25 mM NaPo4 pH 7.0, 100 mM NaCl pH 6.8). Tubes were incubated for 2 h at 4°C to allow diffusion and generation of 10-60% (w/v) continuous sucrose gradient column. Three ml of extracts were layered on the top of the column. Centrifugation was performed at 100000 ×g for 3 h at 4°C. Thirty-three fractions were generated by carefully removing 1 ml from the top to the bottom of the tube and analysed by ELISA (II.2.3.1), Coomassie staining (II.2.3.2) and immunoblot (II.2.3.3).

Expression and purification of IBD-VP2 from P. pastoris II.2.2.6The yeast P. pastoris was used as an alternative system for production of IBD-VP2. After transformation, ten recombinant P. pastoris (X-33) colonies were selected from YPDS plates (II.2.1.8) for each construct and the production level of IBD-VP2 was compared at small scale (10 ml of culture medium) between various colonies by extracting the cellular soluble protein content using Y-PER yeast protein extraction reagent (Pierce) according to the manufactures’ instruction followed by performing an immunoblot (II.2.4.3). A colony which produced relatively more amount of protein (IBD-VP2) was designated for glycerol stock preparation (II.2.1.11). For medium-scale expression and purification, 20 ml of YPD medium containing 20 µl of Zeocin was inoculated with the glycerol stock and was cultured for 2 days at 28°C with vigorous shaking. The day after, 2 ml of the main culture was transferred to 200 ml of YPD containing 200 µl Zeocin™ and incubated at the same condition up to the following day. Then the OD600 nm was measured, the cells were harvested by centrifuge (3000 ×g/RT/5 min) and resuspended in 1 L of BMMY medium containing 0.5% (v/v) methanol and the OD600 nm was adjusted to ~ 1.0. The culture was incubated in baffled Fernbach flasks for overnight at 28°C with vigorous shaking and then the methanol concentration was gradually increased from 0.5%-1% (v/v) during additional 3 days of growth. The cell-free medium was collected following centrifugation of the 1 L culture at 3000 ×g for 10 min at 4°C and was concentrated to 50 ml by passing through a 30 kDa cut off membrane (Schleicher & Schuell) with the help of the cross-flow technique. For protein extraction, the harvested cells were resuspended in breaking buffer and disrupted by five passes in a microfluidizer (Newton, MA, USA). The supernatant was collected after centrifugation at 13,000 ×g for 30 min at 4°C. For purification, the IBD-VP2 content of the cell extract and concentrated medium was precipitated by addition of 50% ammonium sulfate and resuspended in 5 ml phosphate-buffered saline (PBS). The extract was purified by size exclusion chromatography (SEC) on a Hiprep 26/60 Sephacryl S-400 HR column (GE HealthCare, Freiburg) and the SVP elution fractions were concentrated using a Vivaspin 20 spin column with a 300-kDa cut-off membrane (Sartoriusstedim). As a negative control, 500 ml of wild type X-33 cell culture was

Page 46: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

40

processed as the transformed cells (without Zeocin™) and the medium and cell extract was extracted and concentrated as explained. The purity of IBD-SVPs was determined by densitometric analysis of gels stained with Coomassie brilliant blue using the AIDA software (Raytest isotope mass devices GmbH). BMMY medium (pH 6.0):

Potassium phosphate 0.1 M YNB 1.34% (w/v) Biotin 0.4% (w/v) Methanol 0.5% (v/v) Yeast extract 1% (w/v) Peptone 2% (w/v)

YNB is yeast nitrogen base without amino acids. Breaking buffer (pH 4.0):

Sodium acetate pH 4.0 0.1 M PMSF 1 mM EDTA 1 mM Glycerol 5% (v/v)

Protein analysis II.2.3

Quantification of proteins II.2.3.1The concentration of purified protein was determined by BCA assays. Concentration measurements were performed in duplicate according to the manufacturer’s instructions, using BSA as standard in low binding plate. To measure the concentration of IBD-VP2 in tobacco leaves, two leaf disks were cut out, weighted and mixed and grinded together with 2 µl of extraction buffer (II.2.2.5) per milligram leaf material. Cell debris was removed by centrifugation (13,000 g/4°C/20 min) and the supernatant was analyzed in an ELISA. Briefly, cell extracts were coated on polyvinyl chloride 96-well ELISA plates serially diluted two-fold with starting dilution factor of 1:50 in PBS and incubated o/n at 4°C (wild type tobacco leaf extract and 2 µg of purified IBD-VP2 in wild type tobacco leaf extract were used as negative and positive control, respectively). The plates were washed three times with PBST and blocked with 2.5% (w/v) skim milk in PBST for 1 h at RT. After washing as above, rabbit anti-IBD-VP2 (1:10000 in PBS) was added. Following 1 h incubation at RT and wash, horseradish peroxidase-conjugated goat anti-rabbit (final concentration: 0.12 µg/ml in PBS) was added. The plates were incubated at RT for 1 h. After washing the plate, the color was developed by adding ABTS substrate solution. OD405 nm was measured using a microtiter plate reader. IBD-VP2 content was calculated according to the ratio of mean of OD405 nm at the saturation point of the sample to the mean of OD405 nm of the positive control (known concentration) at the saturation point. To measure the concentration of IBD-VP2 in Pichia cell cultures, the cells were harvested from 1 ml of the culture by centrifugation (3000 g/4°C/5 min) and the total soluble protein was extracted

Page 47: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

41

using Y-PER yeast protein extraction reagent (Pierce) according to the manufacturer’s instruction. Cell debris was removed and the quantification was performed as described above for tobacco leaves using ELISA. Wild type X-33 culture was treated in the same manner and used as negative control.

SDS-PAA gel electrophoresis and Coomassie brilliant blue staining II.2.3.2Following protein extraction and purification (II.2.2), and quantity of the recombinant proteins were studied by discontinuous SDS-polyacrylamide gels (for the stacking gel: T = 4%, C = 2.6%, pH 6.8; for the separating gel: T = 12%, C = 2.6%, pH 8.8) (Ausubel et al., 1995) under reducing and/or non-reducing conditions (Laemmli, 1970) using a BioRad minicell II apparatus. Before loading onto the gel, protein samples were mixed with appropriate 5x sample buffer depending on the requirement for reducing or non-reducing conditions. For reducing conditions, samples were denatured by boiling for 5 min. The proteins were separated electrophoretically with constant 180 V for 1 h. Protein bands were visualized by staining with Coomassie brilliant blue or transferred to nitrocellulose membrane for immunoblotting analysis (II.2.3.3). Staining and de-staining of the gels were performed with Fairbank solutions following boiling by microwave and constant rocking (Wong et al., 2000). The purity of protein was quantified by densitometry of the Coomassie brilliant blue stained gels using AIDA image analysis software. Reducing SDS-PAGE sample buffer:

Tris-HCl (pH 6.8) Glycerol

62.5 mM 10% (v/v)

β-mercaptoethanol 5% (v/v) SDS Bromophenol blue

2% (w/v) 0.05% (w/v)

Non-reducing SDS-PAGE sample buffer (pH 8.3):

Tris-HCl (pH 6.8) Glycerol

62.5 mM 10% (v/v)

SDS Bromophenol blue

2% (w/v) 0.05% (w/v)

SDS-PAGE running buffer (pH 8.3):

Tris base 25 mM Glycine 192 mM SDS 0.1% (w/v)

Immunoblot analysis II.2.3.3Separated proteins (II.2.3.2) were transferred from an SDS-PAA gel to a PVDF or Hybond™-C nitrocellulose membrane (0.45 μm) electrophoretically using the BioRad Mini Protean II tank blot device. After blotting the membrane was blocked with 1x PBST buffer containing 5% (w/v) skim milk powder. As primary antibody either rabbit anti-His6 tag polyclonal antibody or rabbit- or mouse anti-IBD-VP2 (0.2 and 0.1 µg/ml in 1x PBST) were used to detect his-tagged protein and

Page 48: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

42

IBD-VP2, respectively. Binding of the primary antibody was detected by addition of diluted (1:5000 in 1x PBST) alkaline phosphatase (AP)-conjugated secondary polyclonal antibodies, either goat anti-rabbit or goat anti-mouse (final concentration: 0.2 µg/ml). Bound secondary antibodies were finally revealed by addition of substrate BCIP/NBT in AP buffer. PBST buffer (pH 7.4):

NaCl 137 mM KCl 2.7 mM Na2HPO4 8.1 mM KH2PO4 1.5 mM Tween 20 0.5-1.0% (v/v)

Transfer buffer (pH 8.3):

Tris-HCl 25 mM Glycine 192 mM Methanol 20% (v/v)

AP buffer (pH 9.6):

Tris-HCl, pH 9.6 100 mM NaCl 100 mM MgCl2 5 mM

Dot blot analysis II.2.3.4Around 5 µl of either tobacco or Pichia cell extracts producing IBD-VP2 (II.2.2.5, II.2.2.6) was immobilized on a nitrocellulose membrane. Immobilized IBD-VP2 was detected using rabbit anti-IBD-VP2 polyclonal antibody (final concentration: 0.2 µg/ml) followed by AP-labeled goat anti-rabbit polyclonal antibodies (final concentration: 0.12 µg/ml). Binding of AP-conjugated secondary antibodies was revealed by adding the BCIP/NBT substrate. This method was used for screening of transgenic plants and recombinant yeast colonies.

Size exclusion chromatography (SEC) II.2.3.5Gel filtration was performed using superdex 200 and Hiprep 26/60 Sephacryl S-400 HR column (GE HealthCare). The columns were equilibrated with 1x PBS. Purified tobacco- (II.2.2.4) and Pichia-derived samples were loaded and run at 1 ml/min on the first and second column, respectively. The eluent was collected in 2-ml fractions. For calibration, Ovalbumin (43 kDa), Conalbumin (75 kDa), Aldolase (158 kDa), Ferritin (440 kDa) and thyroglobulin (669,000 Da) were run as molecular weight standards. The eluate fractions were monitored by UV 215 nm, 225 nm and 280 nm. The IBD-VP2 containing fractions were evaluated by indirect ELISA. Briefly, 100 µl of the fractions were coated into 96-well ELISA plate overnight at 4 ºC. The plates were blocked using 2.5% (w/v) skim milk in PBST for 1 h at RT. Immobilized IBD-VP2 was detected using rabbit anti-IBD-VP2 polyclonal antibody (final concentration: 0.2 µg/ml) followed by AP-labeled goat anti-rabbit polyclonal antibodies (final concentration: 0.12 µg/ml). Washing was performed three times with PBST following each step. The bound antibodies were detected by

Page 49: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

43

ABTS as substrate. The OD value was measured at 405 nm with a microplate reader and OD values >0.2 above background were considered as positive.

Immunogold labelling and transmission electron microscopy (TEM) II.2.3.6The morphology of the IBD-SVPs was investigated by applying 30-µl aliquots of each sample for 15 min onto discharged 400-mesh carbon-coated nickel grids (Plano, Wetzlar). Excess particles were removed by rinsing with 1x PBS. For conjugation analysis, the grids were blocked for 30 min with 1% (w/v) BSA containing 0.1% (w/v) sodium azide. Goat anti-biotin (final concentration: 0.2 µg/ml in 1x PBS) antibodies labeled with 6-nm gold particles (Aurion, Wageningen, The Netherlands) were used for detection. The grids were incubated for 1 h with the antibody and washed three times each with 1x PBST and then with distilled water. Finally, the grids were negatively stained with a 2% (w/v) aqueous uranyl acetate and observed under a 400T electron microscope (Philips, Eindhoven, The Netherlands) operated at 60 kV accelerating voltage. Digital images were captured with an Olympus camera (MORADA) and processed using iTEM software (Olympus Soft Imaging System GmbH, Münster).

Glycan analysis II.2.4

Immunoblot II.2.4.1To analyze the glycosylation of the recombinant purified (II.2.2.5, II.2.2.6) IBD-VP2 protein, lection blots were performed following SDS-PAGE (II.2.3.2) and immunoblotting (II.2.3.3). For this purpose, rabbit anti-xylose (1 µg/ml in 1x PBS) antibody was used as primary antibody in immunoblot analysis for detection of tobacco-produced and purified glycoproteins followed by an alkaline phosphatase-conjugate goat anti-rabbit secondary antibody (0.2 µg/ml in 1x PBS). NBC/BCIP was used as substrate according as described above (II.2.3.3). For glycan analysis of purified Pichia-produced IBD-VP2, alkaline phosphatase conjugated Galanthus nivalis lectin (0.2 µg/ml) was used for detection of the glycoproteins followed by addition of NBC/BCIP as substrate.

Enzymatic deglycosylation II.2.4.2N-glycan chains of purified tobacco- and Pichia-produced IBD-VP2 proteins were removed by the peptide N-glycosidase F (Roche). Around 10 µg of purified IBD-VP2 was deglycosylated by incubating overnight at 37°C with 2 U of N-glycosidase F in PBS (containing 10 mM EDTA, pH 7.4) to a final volume of 100 µl. The cleavage of the glycan branches resulting in a lower molecular weight and increased mobility was monitored by SDS-PAGE (II.2.3.2).

Glyco-proteomics analysis II.2.4.3Purified tobacco- and Pichia-produced IBD-VP2 proteins were separated by SDS-PAGE and Coomassie-stained were cut out of the gel and put in distilled water and stored at 4°C. After the pieces were de-stained, carbamidomethylated, concomitantly digested with sequencing-grade trypsin and chymotrypsin, proteins were extracted from gel pieces as described (Kolarich et al., 2006). The protein was taken to dryness in a Speed Vac concentrator and reconstituted with water before subsequent LC-MS analysis. Mass spectrometric analysis was performed on a Q-TOF

Page 50: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

44

Ultima Global (Waters Micromass) equipped with a standard electrospray unit, connected to an Ultimate 3000 Cap-LC system (Dionex) as previously described (Stadlmann et al., 2008). Briefly, the tryptic (glyco)-peptides were separated using a Biobasic C18 column (100 x 0.18 mm, Thermo Electron), applying an acetonitrile gradient from 5 to 80% (v/v) acetonitrile at a flow-rate of 2 µl/min, and analyzed in the ES+ MS mode. Data analysis was performed with MassLynx 4.0 SP4 Software (Waters Micromass).

Immunization of mice II.2.5Five female mice (BALB/c) each was immunized with 50 µg of purified tobacco-derived IBD-VP2 protein (II.2.2.5) mixed with 40 µl GEBRU adjuvant MM (Gerbu, Wieblingen) via intraperitoneal injection. Six further boosts were given with 2 weeks intervals with the same amount of antigen and 20 µl GEBRU adjuvant MM per mouse. One mouse was used as a control in the same schedule but received just adjuvant without antigen. The final boost was performed 4 days prior to sacrifice. Blood samples were collected one week after each immunization and prior to sacrifice. Blood was taken from the tail vein making a wound with scissors on top of the tail and taking 5 µl of blood. The blood was diluted 100 fold with 1x PBS and stored at -20°C for estimation of antibody titers.

Determination of antibody titer II.2.5.1Determination of polyclonal antibody titers in sera of mice was performed by direct ELISA using purified IBD-VP2 protein (II.2.2.5) as antigen. Briefly, 1 µg antigen/ml PBS was coated on polyvinyl chloride 96-well microtiter plates over night at 4°C. The following day, after washing, plates were incubated in 2.5% (w/v) skim milk in 1x PBST again for overnight at 4°C. After washing, serially diluted two-fold down of mouse sera, from 1:1,000 to 1:204,800 dilutions, were added to the plates. Test sera were also analyzed in wells lacking antigen in order to analyze background binding. After one hour incubation at 37°C and washing with 1x PBST, peroxidise-conjugated goat anti-mouse antibodies were added at 0.2 µg/ml. After 1 h incubation at RT, plates were washed and stained with ABTS (2, 2- azino-bis-ethylbenzthiazolin-6-sulfonic acid) substrate solution. Extinction was measured at 405 nm after 20 min stain development.

Immunization of chickens II.2.6

Vaccine component preparation II.2.6.1Sixteen grams of the Pichia cells harvested from 1 litre culture was applied to freeze-drying. Freeze-drying was performed after inactivation of Pichia cells by applying to two thermo-cycles; heating to 70°C for 5 min directly after harvest followed by incubation for 5 min at 15°C. The freeze-drying machine was switched on for 30 min until the pump warmed-up and the condensation trap cooled down to -55°C. Then the frozen samples were placed into the machine and the vacuum was adjusted to 0.31 mbar pressure. The process was continued until complete dryness of the samples (48 h for about 8 gram of fresh weight cells in a 50 mL falcon tube). Purified IBD-SVPs (II.2.2.6) was used in immunization, as well.

Page 51: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

45

Immunization plan II.2.6.2Sixty-five chickens were hatched (day 0) from specific pathogen free (SPF) eggs (Lohmann Tierzucht, Cuxhaven) and divided randomly between 13 experimental groups, 5 for each group, according to the table II-7. Chickens were housed in separate isolators as below; Isolator 1: groups 1, 2, 3 and 4; Isolator 2: groups 5, 6, 7 and 8; Isolator 3: groups 9, 10, 11; Isolator 4: group 12; Isolator 5: Group 13, under negative-pressure ventilation and filtrated air positive pressure. They were given free access to food and drinking water. Immunization started at day 14 and repeated 4 times with one-week intervals. A mixture of 50 µg CpG ODN (II.1.7, table II.1.4) plus NaF (100 mg per kg of chicken weight at the time of vaccination (based on the graph shown in figure II.1)) was used as oral adjuvant and Gerbu adjuvant 100 (Gerbu) was used as parenteral adjuvant in the immunization procedure. Oral immunization was administrated by oral gavage into the crop. Chickens in groups 1 and 2 received orally high dose (250 mg) of lyophilized Pichia cells producing IBD-SVP with and without adjuvant mixture, respectively. Chickens in groups 3 and 4 received orally low dose (25 mg) of lyophilized Pichia cells producing IBD-SVP with and without adjuvant mixture, respectively. Chickens in groups 5 and 6, orally received low dose (50 µg) of purified IBD-SVP (II.2.2.6) with and without adjuvant mixture, respectively. Chickens in group 7 and 8 orally received high dose (500 µg) of purified IBD-SVP (II.2.2.6) with and without adjuvant mixture, respectively. Chickens in group 9 orally received lyophilized non-transformed Pichia cells (250 mg) and were used as Pichia control. Individuals in group 10 received 20 µg of purified IBD-SVP (II.2.2.6) plus 20 µl Adjuvant 100 intramuscularly (i.m). Birds in groups 11 and 13 were not vaccinated and used as challenge and normal control, respectively (table II-7). Birds in group 12 orally received AviPro Gumboro vac (Lohmann Animal Health GmbH & Co. KG) and used as positive control of oral immunization. All the orally administrated vaccines were dissolved in distilled water. Then adjuvants were added. The final volume applied orally was 1 ml. The final volume of parenterally administrated vaccine was 0.1 ml. Blood samples (from the wing vein) and cloaca swab were collected from each chicken prior each immunization, challenge and day of euthanization. Cloaca swabs were rinsed in 500 µl PBS containing complete, EDTA-free Protease Inhibitor cocktail tablets from Roche (1 tablet for each 25 ml of 1x PBS) for one hour. The samples were centrifuged (5000 ×g/5 min/RT) and after removing the insoluble part, stored at -20°C for further studies. One week after the last boost, chickens (except chickens from group 13) were challenged with a classical IBDV strain, MB3, from a recent outbreak in Germany. Because the strain did not grow in well in embryonated SPF eggs and was not cell culture adapted, the birds were infected ocularly with 0.1 ml homogenized and filtrated (0.2µm) bursa fabricious from naturally infected chickens kindly provided by Dr. Hermann Block (Poultry Practice Mayer-Block, Uelsen). A week after challenge, blood and cloaca samples were collected from all survived birds, chickens were sacrificed and bursa fabricious was isolated, divided into two segments and stored in -20ºC for virus clearance studies (II.2.6.4) and viral tissue damage analysis by histology tests (II.2.6.5).

Health status monitoring II.2.6.3The health of chickens was monitored every week during immunization and every day following challenge. The weight of chickens was recorded at the beginning of immunization and every week

Page 52: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

46

before next round and right after death. All the observations of clinical sign were also recorded every day following challenge for 5 days before euthanization.

Figure II.1 Weight development in Specific pathogen free (SPF) chickens. The data was provided by Dr. Rüdiger Hauck (Institute of Poultry Diseases, Freie Universität Berlin). Table II.7 Experimental design for chicken immunization.

Antibody titer determination II.2.6.4The titer of IBD-VP2-specific antibodies (IgY, IgM and IgA) in blood and (for IgA) in feces cloak swab) samples was measured by indirect ELISA. Microtiter plates (96-well) were coated with purified Pichia-produced IBD-SVPs (final concentration: 10 µg/ml) in PBS overnight at 4°C. The plates were blocked using 2.5% (w/v) skim milk in PBST for 1 h at RT. After 3 times washing with PBST, 100 µl of serially twofold dilutions of serum samples starting from 1:50 dilution in 1x PBS were applied. For IgA detection in the sera, 1:5 dilutions were used. Cloak swab was rinsed in 500 µl of 1x PBS containing complete, EDTA-free Protease Inhibitor cocktail tablets from Roche (1 tablet for each 25 ml of 1x PBS) for 2 hours at 4ºC. Hundred microliters of the extract was applied to the ELISA plate following centrifugation at 3000 ×g for 5 min at RT. After 2 hours

Page 53: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

47

incubation at RT and 3 times washing as described above, secondary antibodies were applied. They were horseradish peroxidase-conjugated rabbit anti-chicken IgY (2 µg/ml), goat anti-chicken IgM (5 µg/ml) and goat anti-chicken IgA (5 µg/ml) for detection of chicken IgY, IgM and IgA, respectively. Bound secondary antibodies were detected by ABTS as substrate. The OD value was determined at 405 nm using a microplate reader. The anti-IBD-VP2 titer was considered as the last dilution showing an OD405 nm higher than 0.2.

IBDV antibody test kit II.2.6.5The anti-IBD-VP2 titer was also assessed by the commercial antibody ELISA test kit, IDEXX IBD-XR Ab test (IDEXX, Westbrook, ME, USA). This kit is commonly used in chicken farms to test the serum antibody titer against IBDV for evaluation of immunity in chickens. The method is based on indirect ELISA and individuals show titers above 396 are considered as resistant.

Detection of IBDV in bursa tissue II.2.6.6The presence of the IBDV antigen in the bursa of Fabricius was analyzed using a sandwich ELISA as previously described (Rong et al., 2007) with some modifications. Briefly, wells of a flat bottom 96-well microtiter plate were coated with mouse anti-IBD-VP2 antibody (15F5, final concentration 100 µg/ml) for o/n at 4°C. Then, wells were washed with 1x PBS containing 0.05% (v/v) Tween 20 (PBST). The plate was blocked by 150 µl/well of 2.5% (w/v) skim milk dissolved in PBST and incubated at RT for 1 h. Following the washing procedure as described above, the bursa of Fabricius was ground and diluted with 1x PBS at 1:1 (w/v). One hundred microliter of the homogenate was added to the wells. The plate was incubated o/n at 4°C. After washing as above, 100 µl of rabbit anti-IBD-VP2 diluted at 1:5000 in PBST, was added to each well. Following incubation at RT for 1 h and wash, 100 µl of HRP-conjugated goat anti-rabbit (final concentration: 0.12 µg/ml) was added as secondary antibody. The bound antibodies were detected by ABTS as the substrate. The OD value was determined at 405 nm with a microplate reader and the values higher than 0.2 were considered as positive.

Histological bursa lesion II.2.6.7Bursa Fabricius was collected after euthanization and dissection of the birds from all 65 chickens and fixed in 10% (v/v) phosphate buffer formalin, were paraffin embedded, cut and stained with haematoxylin and eosin. Bursa lesion scores were determined using a light microscope and comparison between groups (Block et al., 2007). The bursal lesion scoring system was previously described by Li et al. (2004) and is as follows: score 0, no bursal damage and lesion in any follicle, clear demarcation of medulla and cortex; score 1, mild necrosis and mild lymphocyte depletion of a few follicles with overall bursal architecture maintained; score 2, moderate atrophy or lymphocyte depletion in 1/3 to 1/2 of the follicles, aggregation of heterophils, macrophages, and hyperplasia of epithelial reticular cells with some vacuole-like structures; score 3, more than 1/2 of follicles with severe necrosis, atrophy and lymphocyte depletion, and loss of the outline of follicular architecture such that it is replaced by proliferated connective tissue and fibroplasias.

Page 54: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

48

Statistical analysis II.2.6.8Statistical analyses were carried out using SPSS 19.0 software (SPSS Inc., Chicago, IL, USA). For all analyses, chicken was considered as an experimental unit. The significance level (α) was set to 0.05. The normality of the data and the homogeneity of variances were assessed using Shapiro-Wilk’s and the Levene’s tests, respectively. The collected data were subjected to one-way ANOVA analysis of variance followed by Duncan’s test at significance level of p< 0.05.

Bioconjugation analysis II.2.7

Stability analysis of IBD-sub viral particles (IBD-SVPs) II.2.7.1The solvent stability of the IBD-SVPs was tested by dissolving purified particles (II.2.2.6) in 5, 10, 20 or 50% (v/v) ethanol/water or DMSO/water to a final concentration of 1 mg/ml, with IBD-SVPs diluted to the same concentration in 1x PBS as a control. All samples were incubated at room temperature for 24 h then centrifuged at 13000 ×g for 20 min at RT prior to analysis. To test temperature stability, 50-µl aliquots of purified IBD-SVPs (1 mg/ml) were incubated at 25, 40, 50, 60, 65, 68.5, 72, 75, 80 and 90°C for 1 h prior to centrifugation as above. To test pH stability, purified IBD-SVPs were dissolved in 50 mM acetate buffer pH 2.5, 50 mM acetate buffer pH 5.5, PBS pH 7.4, 50 mM Tris-HCl pH 8.0 or 50 mM Tris-HCl pH 9.0, to a final concentration of 0.5 mg/ml. After overnight incubation at room temperature, the samples were centrifuged as above. In all three cases, the stability, conformation and integrity of the IBD-SVPs were determined by NAGE (II.2.7.2), spectroscopy (II.2.7.3), DLS (II.2.7.4) and TEM (II.2.3.6) as described.

Native agarose gel electrophoresis (NAGE) II.2.7.2Treated samples were separated by 1% (w/v) native agarose gel electrophoresis at 50 V for 1 h. The agarose gel was prepared in buffer A (25 mM Tris-HCl pH 8.15, 19.2 mM glycine) as previously described (Kim et al., 2000). The protein bands were visualized by staining with Coomassie brilliant blue.

Spectroscopy II.2.7.3After solvent exposure, temperature and pH testing, each VP2 sample was centrifuged at 13000 ×g for 20 min at RT, and the absorbance spectrum (UV/Vis, 220–310 nm) was measured using a NanoDrop spectrophotometer (PeQlab Ltd., Erlangen). The total quantity of soluble protein was estimated from the A280 value (mean from three replicates). After dye molecule coupling, the absorbance was measured using the “protein and labels” option of the spectrophotometer.

Dynamic light scattering (DLS) II.2.7.4The radius of IBD-SVPs in solution (0.5-ml aliquots, 1 mg/ml) was determined by DLS in self-beating mode on an ALV set-up comprising a 473-nm solid-state laser (Koheras, Kleinostheim) operating at a 60º angle. Experimentally measured autocorrelation functions (ACFs) of the intensity fluctuations g (2)(t) were determined using the CONTIN algorithm (Provencher, 1982a,b) yielding the distribution of the relaxation time t.

Page 55: Expression and Characterization of Infectious Bursal

Chapter II Material and methods

49

Chemical bioconjugation II.2.7.5Biotin was conjugated to the amine group of lysine residues or the carboxyl group of aspartic and glutamic acid residues of purified IBD-SVPs (II.2.2.6) exposed to the solvent. IBD-SVPs display five lysine residues (Lys 81, 192, 309, 316 and 381), four glutamic acid residues (Glu 300, 311, 355 and 378) and nine aspartic acid residues (Asp 51, 83, 138, 190, 213, 267, 279, 287 and 323) per coat protein; IBD-SVPs are formed by 60 copies of identical coat protein units. Alexa Fluor 488 N-hydroxysuccinimide (NHS) ester and the DyeLightTM antibody labeling kit (Pierce) were used to determine the number of accessible amines. Conjugation to amine groups was achieved using 1 mg/ml IBD-SVPs and biotinamidohexanoic acid NHS ester (Sigma) dissolved in DMSO to a concentration of 2.1 mM, and mixed with 1x PBS to a final DMSO concentration of 10% (v/v). The ratio of biotin to IBD-SVP was 6000:1. The reaction was allowed to proceed for 2 h at 4°C with gentle agitation. Conjugation to carboxyl groups was achieved by dialyzing IBD-SVPs in 1x PBS against 0.1 M MES buffer (pH 5.5) using a Nanosep centrifugal device with a 100 kDa MWCO (Pall, Dreieich). IBD-SVPs in MES buffer at a final concentration of 0.5–3.5 mg/ml were incubated with 6.5 mM EDC (1-[3-(Dimethylamino) propyl]-3ethylcarbodiimide methiodide) and 2 mM biotin hydrazide in DMSO (Sigma). The biotin hydrazide was used at a molar excess of 7800–15,600:1 biotin: IBD-SVP and the final DMSO concentration was 10% (v/v). The reaction was allowed to proceed overnight at room temperature with gentle agitation to prevent precipitation. Modified particles were purified on a Hiprep 26/60 Sephacryl S-400 HR column (GE HealthCare) and the SVP peak was concentrated using a Vivaspin 20 with 300-kDa cut-off membrane (Sartorius).

Page 56: Expression and Characterization of Infectious Bursal

Chapter III Results

50

III Results The result section contains two major parts: The first part describes the expression, purification and characterization of IBD-VP2 in tobacco plants to study protein accumulation in different cell compartments, stability, functionality, glycosylation, immunogenicity and self-assembly capability either in vivo or in vitro. Accordingly, the relevant cDNA of IBD-VP2 was cloned into the plant expression vector pTRAkc (III.1.1) and then expressed either transiently (III.1.2) or stably (III.1.3) in N. tabacum cv. Petite Havana SR1 leaves. The recombinant protein accumulation levels were evaluated using four sub-cellular targeting strategies (cytosol, apoplast, membrane-anchored and chloroplast) exploiting proper targeting peptide sequences. The target protein was purified (III.1.4) and characterized (III.1.5 and III.1.6). Furthermore, the immunogenicity of tobacco-derived proteins was analyzed in mice (III.1.7). Moreover, the yeast, Pichia pastoris, was used as an alternative expression system. The IBD-VP2 was cloned into the Pichia expression vector pPICZ_B (III.2.1) and expressed as either cytosolic or secreted protein (III.2.2). The produced proteins were purified (III.2.3) and characterized (III.2.3 and III.2.4) in the same manner as for the plant expression and moreover used as a vaccine for immunization of chickens in both oral and parenteral administrations (III.3). In the second part of the result section, the sub-viral particles (IBD-SVPs) derived from self-assembly of recombinant IBD-VP2 monomers produced in Pichia, were investigated as a potential nanotechnology platform and characterized concerning organic solvent (III.4.1), heat (III.4.2) and pH (III.4.3) stability. Finally, the addressability of solvent-exposed amine and carboxylic groups on the surface of IBD-SVPs was tested via traditional bioconjugation methods (III.4.4).

III.1 Expression and characterization of IBD-VP2 in N. tabacum

Cloning of IBD-VP2 gene sequence into pTRAkc vector for generation of III.1.1tobacco expression vectors

For recombinant expression of IBD-VP2 in tobacco, the corresponding cDNA was cloned into the plant expression vector pTRAkc containing the CaMV 35S promoter with the duplicated enhancer region (II.2.1.17). The cDNA of the mature IBD-VP2 sequence (strain IR01, DNA accession number: AY704912) was used as a template in PCR using high-fidelity Taq polymerase. The corresponding sequence of the first 441 amino acids (mature IBD-VP2) fused to the C-terminal His-tag was amplified using the primers VP2-forward and VP2-reverse-withHis6 (table II.1) containing BspHI and XbaI restriction recognition sites, respectively. Following direct cloning of the PCR product (~1.3 kb) into the pJET1.2/blunt vector, the sequence was verified by sequencing using the pJET1.2 forward and reverses primers and reconfirmed by the inner primer set VP2Primer1_forward and VP2Primer1_reverse (table II.3) (II.2.1.19). The IBD-VP2 encoding region was double digested by BspHI and XbaI and either cloned into the vector pTRAkc-GFPapo containing the sequence for the signal peptide LPH of the murine antibody 24 heavy chain (Vaquero et al., 1999) or into the vector pTRAkc-rbcs-cTP containing the Rbcs encoding for the chloroplast targeting signal peptide of Ribulose-1,5-bisphosphate carboxylase oxygenase

Page 57: Expression and Characterization of Infectious Bursal

Chapter III Results

51

(RuBisCO) small subunit from Solanum via the restriction sites NcoI and XbaI, resulting in generation of the final expression vectors pTRAkc-VP2apo and pTRAkc-VP2chlo, respectively. For cytosolic accumulation, SOE (splicing by overlap extension) PCR was exploited as described (II.2.1.19) using the CHS-elongation5’ and VP2-reverse-withHis6 primers for elongation and Solulin-for5’UT and VP2-reverse-withHis6 for amplification of the chalcone synthase 5’ UT fused to the IBD-VP2 encoding sequence. The resultant 1.4 kb construct (CHS-VP2) was double digested with EcoRI and XbaI and cloned into the corresponding sites in the pTRAkc-GFPapo vector. The final vector was named pTRAkc-VP2cyto. For creation of the plant expression vector encoding for the membrane retained VP2, two separate fragments were constructed and fused together. To make the His-tag accessible for recombinant protein detection and purification, the tag was fused to the N-terminal end of IBD-VP2. For this, the fragment CHS-VP2 was generated using SOE-PCR. Briefly, IBD-VP2 cDNA was used as template in companion with CHS-elongation5’-withHis6 and VP2-reverse for elongation, Solulin-for5’UT and VP2-reverse for amplification. The resulting fragment was 1.4 kb long. The second fragment was 128 bp long containing an ORF of 96 bp cytochrome b5 membrane anchor peptide encoding sequence (CytB5-MA), amplified by PCR using CytB5 cDNA as template and CytB5memAnch.-for and vIL-10_XbaI-bw primer sets. Eventually, the two generated fragments were fused together in a SOE-PCR using the generated overlapping region of the two fragments and the joint sequence was amplified using the primers Solulin-for5’UT and vIL-10_XbaI-bw. Finally, the generated construct was double digested with EcoRI and XbaI and subsequently cloned into the corresponding sites of the pTRAkc-GFPapo vector. The final construct was named pTRAkc-VP2mem. All PCR and cloning procedures as well as vector maps are illustrated in Appendix VII.2. All harboring plasmids were checked by restriction enzyme digestion following sequencing (data not shown). All four generated vectors carrying the correct sequence were transferred to Agrobacterium via electroporation (II.2.1.6). Recombinant agrobacteria were checked by colony PCR and positive clones used for transient and stable transformation of tobacco (II.2.2.2, II.2.2.3, II.2.2.4). The schemes of the constructs were demonstrated in figure III.1.

Page 58: Expression and Characterization of Infectious Bursal

Chapter III Results

52

Figure III.1 Schematic drawing of the generated pTRAkc plant expression vectors for IBD-VP2 expression in tobacco; P35S: CaMV 35S promoter with duplicated enhancer region; CHS: 5' untranslated region of the Petroselinum chalcone synthase; LPH: plant codon optimized leader sequence derived from the murine heavy chain of the TMV-specific mAb24; Rbcs: Rubisco small subunit chloroplast targeting signal; CytB5-MA: cytochrome b5 membrane anchor; H6: IBD-VP2: cDNA of infectious bursal disease viral protein 2 corresponding to the first 441 amino acids; H6: his-6 tag for detection and purification; pA35S: terminator from CaMV; SAR: scaffold attachment region; Pnos: nopaline synthase gene promoter; nptII: Neomycin phosphotransferase II (kanamycin resistance gene); pAnos: nopaline synthase gene terminator; LB: left border; RB: right border; (not drawn to scale).

Transient expression of IBD-VP2 in tobacco leaves III.1.2For fast screening of the best targeting strategies resulting in the highest accumulation of IBD-VP2 (or in brief VP2), transient expression experiments via agro-infiltration were performed (II.2.2.3). Three tobacco leaves for each construct were infiltrated with recombinant agrobacteria. After three days of incubation, soluble proteins were extracted (II.2.2.5) and analyzed by immunoblot (II.2.3.3) to determine the productivity and integrity for each of the four different VP2 constructs. The calculated molecular mass for VP2 fused to the His-tag is ~48 kDa. The fused membrane anchor (CytB5-MA) adds 3.7 kDa to the protein size. The protein was named based on the cellular accumulation site. Therefore, VP2apo, VP2chlo, VP2cyto and VP2mem stand for VP2 accumulated in the apoplast, chloroplast, cytosol and cell membrane, respectively. Immunoblot analysis revealed a distinct band of ~40 kDa for the constructs pTRAkc-VP2cyto and pTRAkc-VP2chlo (figure III.2). While the corresponding band for pTRAkc-VP2apo was ~47 kDa, VP2mem was not detectable by immunoblot (figure III.2) under both non-reducing and reducing conditions. Interestingly, in contrast to most of the known proteins, the movement pattern of VP2 under reducing condition is smeary whereas under non-reducing condition a distinct band was observed. Accordingly, detection and amount evaluation of VP2 was performed based on immunoblot under the non-reducing condition. Since equal amounts of total soluble proteins were

Page 59: Expression and Characterization of Infectious Bursal

Chapter III Results

53

loaded onto the gel, the accumulation level of VP2 varied considerably between the tested expression constructs. Higher level of accumulation was observed for apoplastic localization of the VP2 protein followed by accumulation in the cytosol and chloroplasts. The size difference for VP2apo when compared to the cytosolic forms VP2cyto and VP2mem can be explained by glycosylation since only the VP2apo protein is transported to the secretory pathway.

Figure III.2 Comparison of VP2 accumulation in various cell compartments using immunoblot. Total soluble leaf proteins were extracted by grinding a leaf disk from a transiently transformed tobacco leaf in two volumes extraction buffer (II.2.2.5), 10-12 µl of the extract containing equal amount of total soluble protein (TSP) was separated by 12% (w/v) SDS-PAGE either under non-reducing or reducing condition (II.2.3.2) and blotted onto nitrocellulose membrane (II.2.3.3). Immuno-detection was carried out using rabbit anti-IBD-VP2 (1:10000) as primary followed by phosphatase-conjugated goat anti-rabbit as a secondary antibody (0.12 µg/ml). Detection was performed with NBT/BCIP for 5 min at RT. M: prestained protein marker. Accumulation levels of the transiently produced four versions of VP2 recombinant protein were determined more precisely by ELISA (II.2.3.1). As standard, purified VP2 protein derived from transiently transformed tobacco leaves with pTRAkc-VP2cyto (III.1.4) was mixed with total soluble protein extract from wild type tobacco leaves and applied in serial dilution to the microtiter plate. Figure III.3 shows that the highest yield was obtained from the apoplastic located VP2 (~260 µg/g FLW). VP2cyto (~160 µg/g FLW) and VP2chlo (~70 µg/g FLW) accumulated to lower levels. The ELISA confirmed the result from the immunoblot analysis because the level of VP2mem was close to the detection limit.

Page 60: Expression and Characterization of Infectious Bursal

Chapter III Results

54

Figure III.3 Determination of VP2 content accumulated in different cell compartments of transiently transformed N. tabacum cv. Petite Havana SR1 leaves. Recombinant protein levels were determined by ELISA (II.2.3.1). 2.5 µl of tobacco extract was mixed with PBS and applied to 96-well ELISA plates in two-fold serial dilutions. 2.5 µl of wild type plant extract alone and with 2 µg of purified VP2cyto derived from transiently transformed tobacco leaves (III.1.4), were also applied into serial dilutions, used as negative and positive standards, respectively. Rabbit anti-VP2 antibody (1:10,000 µl in PBS) and GARHRP antibody (0.12 µg/ml) were used as primary and secondary antibodies, respectively. Detection was performed using ABTS as substrate after 10 min incubation at RT. The measurement was performed in three independent transient experiments (n=3) and error bars represents standard deviations.

Generation and screening of transgenic tobacco plants expressing IBD-VP2 III.1.3Based on transient expression results, the apoplast-targeted (pTRAkc-VP2apo) and cytosolic expression (pTRAkc-VP2cyto) constructs were used for stable transformation of N. tabacum cv. Petite Havana SR1 plants via Agrobacterium-mediated transformation (II.2.2.3). Thirty transgenic lines (designated VP2apo_1→30) and thirty-five transgenic lines (VP2cyto_1→35) were regenerated from transformation experiments, respectively. The T0 generation showed a normal phenotype and was screened for recombinant protein accumulation by immunoblot (II.2.3.3) (data not shown). To achieve higher expression levels via genetic homogeneity, the top four producer plants for each construct were led to produce seeds upon self-fertilization. In total, 47 and 28 T1 plants were analyzed by immunoblot for accumulation of VP2apo and VP2cyto, respectively. The best three elite VP2 producers was selected for each construct and the VP2 levels were determined by ELISA (II.2.3.1). These plants were led to self-pollination to generate seeds. Seed were stored as backup materials in the seed bank of the Fraunhofer IME. The productivity in VP2apo producing T1 plants was reaching to 20-30 µg/g FLW, whereas VP2cyto producing T1 plants showed levels of 10-20 µg/g FLW. Similar to the transient produced recombinant proteins, VP2apo and VP2cyto extracted from T0 and T1 plants were intact on 12% (w/v) SDS-PAA gels under non-reducing condition and had a molecular weight of ~47 and ~40 kDa, respectively (data not shown).

Purification of IBD-VP2 from tobacco leaves III.1.4Both plant-produced proteins, VP2apo and VP2cyto, contain a C-terminal His-tag facilitating purification by immobilized metal ion affinity chromatography (IMAC) using Ni-NTA columns.

0

50

100

150

200

250

300

Apoplast Chloroplast Cytosol Membrane-anchored

µg/g

fres

h le

af w

eigh

t n=3

n= 3

Page 61: Expression and Characterization of Infectious Bursal

Chapter III Results

55

Upon harvesting and vein removal, leaves from transiently producing VP2apo or VP2cyto were used for the purification of VP2 (II.2.2.5). The eluent was collected in 0.2 ml fractions using PBS containing 250 mM imidazole. To monitor the protein content during the each purification step, samples were collected and analyzed by 12% (w/v) SDS-PAGE followed by Coomassie staining and immunoblot using rabbit anti-His and phosphatase-conjugated goat anti-rabbit as primary and secondary antibodies. NBT/BCIP was used as a substrate for color development (figure III.4). Distinct ~40 and ~47 kDa bands were detected for VP2cyto and VP2apo, respectively. In case of VP2cyto, the protein is intact whereas in the case VP2apo the presence of fuzzy bands smaller and bigger than the intact protein size correlates with minor degradation and either aggregation or glycosylation, respectively. The purity of VP2 was determined 35% by densitometric analysis of Coomassie stained gel.

Figure III.4 SDS-PAGE analysis of VP2 purified by IMAC form transiently transformed tobacco leaves using pTRAkc-VP2apo (A, B) and pTRAkc-VP2cyto (C, D). VP2apo and VP2cyto with His-tag fused at the C-terminal ends were purified using Ni-NTA (II.2.2.5). Twelve-microliter samples of each purification step (including extract, solution part of extract following pH shift (loading), flow through from Ni-NTA column, dead volume, elution fractions 1-4) were separated by 12% (w/v) SDS-PAA gel under non-reducing condition. The VP2 corresponding bands were detected in immunoblot (II.2.3.3) recruiting rabbit anti-His and phosphatase-conjugated goat anti-rabbit as primary and secondary antibodies, respectively. Color was developed using NBT/BCIP as substrate following 5 min incubation period. Distinct ~47 (A) and ~40 kDa (C) bands were detected for VP2apo and VP2cyto, respectively. Proteins content were visualized by Coomassie staining (II.2.3.2) for VP2apo (B) and VP2cyto (D). The purity was determined about 35% using densitometric method. M: prestained protein marker. To improve the purity, the VP2 bands were excised from the gel and the protein content was recovered using EzWayTM PAG protein elution kit (II.2.2.5). The recovered samples were concentrated using nanosep column with 10 kDa MWCO. The purity of the protein was determined >95% by densitometric method using Coomassie stained gel (figure III.5). Total protein content was measured using the BCA kit. The protein yield was estimated about 100 and

Page 62: Expression and Characterization of Infectious Bursal

Chapter III Results

56

80 µg/g FLW for VP2apo and VP2cyto, respectively. The purified protein was stored at -80°C in 50 µg fractions.

Figure III.5 SDS-PAGE analysis to check the VP2 purity. The corresponding VP2 bands obtained from Ni-NTA purification (figure III.4) was recovered from the Coomassie stained gel using EzWayTM PAG protein elution kit and concentrated using 10 kDa MWCO. Twelve µl of the samples were separated using 12% (w/v) SDS-PAA gel under non-denaturing condition. The purity of the proteins were evaluated >95% using densitometric method. Small degradation fractions, less than 10 kDa in size, are also observable.

Glycosylation analysis of IBD-VP2apo III.1.5According to the SDS-PAGE analyses (figure III.5), the tobacco-produced full-size VP2cyto has mass of ~40 kDa whereas the size of VP2apo is ~47 kDa. This variation (~7 kDa) in addition to the smeary movement of VP2apo on SDS-PAA gels (figure III.5) can be explained by glycosylation. The IBD-VP2 contains three potential N-glycosylation sites (verified by the NetNGlyc 1.0 server used for N-glycosylation predictions) (figure III.6). Full glycosylation of all three sites with plant specific extracellular glycan adds additional ~7 kDa to the calculated mass of the protein increasing it to ~54 kDa which is in agreement with the size observed on the stained SDS-gel (figure III.5). To characterize the glycosylation pattern of the transiently produced VP2apo in tobacco leaves, the purified protein (III.1.4) was analyzed by different glycan analysis procedures.

Figure III.6 Prediction of potential N-glycosylation sites of the IBD-VP2 protein predicted by the NetNGlyc 1.0 server (http://www.cbs.dtu.dk/services/NetNGlyc/). Three recognized N-glycosylation positions (aa 46, aa 121, aa 396) are underlined.

Immunoblot analysis III.1.5.1To determine if the tobacco-produced and IMAC purified VP2apo is glycosylated; an immunoblot analysis was performed using glycan-specific antibodies (figure III.7). Following SDS-PAGE and

Page 63: Expression and Characterization of Infectious Bursal

Chapter III Results

57

blotting, the xylose residues were detected by using a rabbit anti-xylose antibody (1 µg/ml in 1x PBS) and detection was carried out using alkaline phosphatase-conjugate goat anti-rabbit as a secondary antibody. NBC/BCIP was used as substrate. VP2cyto and recombinant phl p 1 (timothy grass (Phleum pratense) pollen major glycoprotein allergen produced in transgenic tobacco) were used as negative and positive controls, respectively. The result indicates that VP2apo as well as the apoplastic targeted recombinant phl p1 in contrast to VP2cyto reacted with the anti-xylose antibody, confirming the glycosylation of VP2apo.

Figure III.7 SDS-PAGE for glycosylation analysis of the tobacco-produced VP2 followed by (A) Coomassie brilliant blue staining and (B) glycan detection by immunoblot. Purified proteins (III.1.4) were separated by 12% (w/v) SDS-PAA gel under non-reducing condition (II.2.3.2). Rabbit anti-xylose (1 µg/ml in 1x PBS) and phosphatase-conjugated goat anti-rabbit (0.2 µg/ml in 1x PBS) were used as primary and secondary antibodies. Detection was performed with NBT/BCIP. VP2cyto and phl p 1 (timothy grass (Phleum pratense) pollen major glycoprotein allergen produced in transgenic tobacco were used as negative and positive (Ctrl+) controls, respectively. M: prestained protein marker.

Enzymatic deglycosylation of VP2apo III.1.5.2To demonstrate the addition of glycan chains to the secreted recombinant protein, the purified plant-produced VP2apo (III.1.4) was subjected to deglycosylation (II.2.4.2) with PNGase F and the resulting product was separated by SDS-PAA gel followed by Coomassie staining using VP2cyto as non-glycosylated control (figure III.8). After glycan removal through enzymatic digestion and electrophoretic separation VP2apo showed a molecular weight of 40 kDa, the same size as VP2cyto which accumulates in the cytosol, demonstrating that glycosylation was responsible for the mass differences of ~7kDa.

Page 64: Expression and Characterization of Infectious Bursal

Chapter III Results

58

Figure III.8 SDS-PAGE analysis of the deglycosylated tobacco-produced and affinity-purified VP2apo using peptide N-Glycosidase F (PNGase F). VP2apo after enzymatic deglycosylation (II.2.4.2) has the same size as tobacco-produced and affinity purified VP2cyto (III.1.4) on 12% (w/v) SDS-PAA gel stained with Coomassie brilliant blue (II.2.3.2).

Glyco-proteomics analysis III.1.5.1The VP2 contains three potential glycosylation sites (aa 46, aa 121, aa 396). To determine if all three sites contain glycan chains and to analyse the corresponding glycan composition of each site, the purified tobacco-produced VP2apo (III.I.4) was separated by SDS-PAGE. The Coomassie brilliant blue stained band was excised from the gel digested with trypsin and chemotrypsin and finally analyzed by LC-MS (Dr. F. Altmann, J. Stadlmann and BoKu Vienna, Austria). The result of the LC-MS is shown in figure III.9 and summarized in table III.1. Complex type N-glycans such as GnGnXF and GnGnF were the most prominent glycan types and accounted for 55.7% of the total VP2 glycosylation profile. GnMXF and GnMX were present at lower levels (~20%), while the other types of glycan were found in only few amounts (about 2% or less each). The obtained glycan data showed that the leader peptide (LPH) was fully functional in leading VP2 to the secretory pathway and finally to the apoplast. All three N-glycan sites were glycosylated with variation in glycan compositions and their frequencies are described in more detail in table III.1

Page 65: Expression and Characterization of Infectious Bursal

Chapter III Results

59

Table III.1 Glycan compositions and their ratio at the three N-glycan sites (1, 2 and 3) of the plant-produced and affinity-purified VP2 was determined by liquid chromatography-mass spectrometry (LC-MS).

Glycan type Relative frequency (%) Site 1 Site 2 Site 3

GnGnXF (M3Gn4X1F1) 33.8 30.1 19.6 GnGnF (M3Gn4F1) 30 29.3 24.4 GnMXF (M3Gn3X1F1) 6.5 9.1 13.1 GnMF (M3Gn3F1) 7.4 7.5 17.5 GnUF(Man2Gn3F1) 2 2.9 7.6 GnM (M3Gn3) 2 2.4 3.2 AGnX (A1M3Gn4X1) 0 2.2 0 AGnF (A1M3Gn4F1) 0 2.2 0 AGn (A1M3Gn4) 0 3 0 MU (M2Gn2) 2.2 0 0 GnMX (M3Gn3X1) 1.9 1.8 2.1 Other types 13.3 12 6.6

Figure III.9 Liquid chromatography-mass spectrometry (LC-MS) of the three N-glycosylation sites of plant-produced and affinity-purified VP2apo. Purified VP2apo (III.1.4) separated by 12% (w/v) SDS-PAGE (II.2.3.2) was excised from the gel. Following protein recovery, it was digested with trypsin and chemotrypsin and the resultant peptides were applied to LC-MS analysis (II.2.4.3). The spectrum of glycosylated peptides are shown in A (site 1), B (site 2) and C (site 3) (see also Figure III.6).

Page 66: Expression and Characterization of Infectious Bursal

Chapter III Results

60

Analysis of plant-produced VP2 assembly III.1.6To study if the purified tobacco-produced VP2cyto and VP2apo assembled into SVPs, the recombinant proteins were analyzed by size exclusion chromatography using Superdex 200 having a separation range of 10-600 kDa for analysis of globular proteins (II.2.3.5). The chromatographic profiles were obtained by measuring the absorption at 215 nm (figure III.10). The first two peaks showed the highest content of intact VP2apo and VP2cyto, respectively, as determined by immunoblot. The corresponding size of each peak was calculated based on the elution volumes using a calibration curve equation obtained by running the molecular weight standards: ovalbumin (43 kDa), conalbumin (75 kDa), aldolase (158 kDa), ferritin (440 kDa) and thyroglobulin (670 kDa). As indicated in the figure, the first two peaks are matching with trimers and monomers, respectively. The shift in peak profile between VP2apo and VP2cyto are due to the glycan moieties, which are covalently attached to VP2apo and are absent in VP2cyto. The two latter peaks which are in size of 5 and less than 1 kDa belong to degradation fractions and salt impurities, respectively. This result shows that the trimer form is the maximum assembly level observed in IMAC-purified VP2 samples.

Figure III.10 Chromatographic profile obtained by separation of purified (III.1.4) VP2cyto (red) and VP2apo (blue) in two separate runs using a Superdex 200 column (II.2.3.5). To estimate the VP2 content of each peak, the elution fractions corresponding to the tip of the peaks were separated by 12% (w/v) SDS-PAA gel followed by immunoblot using rabbit anti-IBD-VP2 (1:10000) and phosphatase-conjugated goat anti-rabbit (0.12 µg/ml) as primary and secondary antibodies, respectively. Color was developed using NBT/BCIP as substrate following 5 min incubation period. The intact VP2 was eluted in trimeric and monomeric forms in about 12 and 14 ml of the elution volume. The degradation fractions (~5 kDa) and salt impurities (>1 kDa) were also eluted at about 19 and 24 of the elution volume. Standard calibration curve equation obtained by running the molecular weight standards (black triangles): ovalbumin (43 kDa), conalbumin (75 kDa), aldolase (158 kDa), ferritin (440 kDa) and thyroglobulin (670 kDa).

Page 67: Expression and Characterization of Infectious Bursal

Chapter III Results

61

As shown previously, it is possible to purify IBD-subviral particles produced by recombinant expression of VP2 with His-tagged using IMAC (Wang et al., 2000). Based on structural analysis it was indicated that His-tags fused either to the N- or C-terminal end of VP2 monomers are located on the interior face of fully assembled particles and are not accessible on the surface (Lee et al., 2006; Garriga et al., 2006). Even though, it was still possible to purify IBD-SVPs using IMAC even after his tag removal (Lee et al., 2006). The affinity of IBD-SVPs to Ni+2 is due to two His residues (His 249 and His 253) located on the outer surface of the IBDV particles (Doong et al., 2007). Sequence alignment between the IBD-VP2 sequence described by Doong et al. (2007) with the peer one used in this study showed that the two mentioned His residues are replaced by Glu residues (Appendix VII.3), which do not bind to the Ni+2 column. As described above (III.1.4) the majority of plant-produced VP2apo and VP2cyto was able to adsorb to the Ni+2 column (figure III.4). This fact reveals that either the assembled particles disrupted during extraction/ purification steps due to the pH shifts or high salt concentration or VP2 monomers to not assemble efficiently in tobacco cells. VP2cyto was selected to study the in vitro assembly post extraction and purification because VP2apo carries superimposed glycan chains that may interfere in assembly. Several conditions have been tested during extraction such as lower salt concentration, detergent removal in extraction buffer and keeping the pH constantly neutral in all extraction and purification steps to support assembly of SVPs. In all the efforts, VP2cyto could be purified by IMAC indicating the accessibility of the His-tag but this means also that assembly of VP2 monomers did not occurred (data not shown). Finally, ultracentrifugation as an alternative purification method was exploited (II.2.2.4). The total soluble protein was extracted from transiently transformed leaves and applied to the 10-60% gradient sucrose column (II.2.2.5). Following ultracentrifugation, one milliliter fractions of the sucrose column were collected from the top to the bottom of the sucrose gradient and were analyzed by indirect ELISA (II.2.3.1) to estimate the VP2 content. As depicted in the figure III.11, only one major peak representing the sucrose factions 8 to 17 was observed. The presence of the VP2cyto protein in these fractions was confirmed by immunoblot (data not shown). Fractions 8 to 17 containing the major part of the purified VP2cyto were concentrated using nanosep 10 kDa MWCO devices and analyzed by immunogold labeling and electron microscopy (II.2.3.6). However, no SVPs could be detected (data not shown). Furthermore, efforts to detect SVPs in stably transformed leaf tissues using immunogold labeling and electron microscopy did not succeed (data not shown). All these observations indicated that VP2cyto does not form complexes larger than trimers when produced in tobacco.

Page 68: Expression and Characterization of Infectious Bursal

Chapter III Results

62

Figure III.11 ELISA analysis (II.2.3.1) for the VP2 content of the tobacco-produced VP2cyto separated by 10-60% sucrose gradient ultracentrifuge (II.2.2.5). One milliliter fractions were collected from top to the bottom of the sucrose column and the corresponding VP2cyto content was determined by indirect ELISA (II.2.3.1) using polyclonal anti-VP2 as primary (1:10,000) followed by goat anti-rabbit HRP-labeled as a secondary antibody (0.2 µg/ml). Wild type tobacco cell extract separated by the same procedure was used as a negative control. Detection was performed with ABTS as a substrate for 5 min at RT. The fractions 8 to 17 containing high amounts of VP2cyto were isolated, concentrated using nanosep column with 10 kDa MWCO devices and used for electron microscopy. To facilitate in vitro assembly, fractions 11-15 ml of the superdex 200 column (figure III.10) containing VP2cyto monomers and trimers were collected and mixed. The trimer-monomer mixture was dialyzed overnight against PES buffer containing Ca+2. Although Ca+2 is known as an essential factor for IBD-VP2 assembly (Garriga et al., 2006), it may induce incorrect protein aggregation and precipitation. To inhibit non-proper subunit interactions, detergents such as dithiothreitol (DTT) and/or Tween-20 were added to the mixture and removed gradually stepwise while the Ca+2 concentration was gradually increased using dialysis as described (Lipin et al., 2006). Even though, protein precipitation could not be avoided. As an alternative approach, the trimer-monomer VP2 mixture was completely disassembled using 8 M urea and 5 mM DTT followed by stepwise removal of urea through dialysis to facilitate re-assembly (Klikova et al., 1995; Acosta-Rivero et al., 2004). The urea content was reduced to 2 M, 1 M and 0 M in three steps through dialysis using PES buffer containing Ca+2. As shown in figure III.12, addition of urea not only destroyed the trimer structure, but also increased the degradation of the protein as shown by SEC. Even after stepwise dialysis, the trimer assembly did not reproduce (data not shown) and consequently higher assembly could not take place, meanwhile, the monomer content remained intact after urea treatment and dialysis. In summary, SVP assembly did not occur in tobacco and the alternative approach to initiate SVP assembly after VP2 purification was also not successful. Consequently, it was decided to investigate production of VP2 in an alternative system such as Pichia (III.2). Nevertheless, immunogenicity of the VP2 monomer-trimer mixture was tested in mice.

0

0.5

1

1.5

2

2.5

3

3.5

4

0 5 10 15 20 25 30 35

OD

405

nm

Fractions (1ml each, collected from top to bottom)

VP2cyto tobacco extract

wild type tobacco extract

Page 69: Expression and Characterization of Infectious Bursal

Chapter III Results

63

Figure III.12 Chromatographic profile obtained by separation of VP2cyto before (blue) and after incubation with urea (brown) in two separate runs using a Superdex 200 column (II.2.3.5). The degradation fractions (~5 kDa) and salt impurities (>1 kDa) were also eluted at about 19, 21 and 24 of the elution volume. Standard calibration curve equation obtained by running the molecular weight standards (black triangles): ovalbumin (43 kDa), conalbumin (75 kDa), aldolase (158 kDa), ferritin (440 kDa) and thyroglobulin (670 kDa).

Immunogenicity of tobacco-produced VP2 in mice III.1.7To check the immunogenicity of the tobacco-produced VP2, two groups of mice each containing five individuals were immunized either with purified (III.1.4) VP2apo (group 1) or VP2cyto (group 2), respectively. As indicated in figure III.15, immunization was repeated seven times, intraperitoneally at two-week intervals (II.2.5). After each boost, polyclonal antisera were collected from each mouse from the tail vein and the titer was determined by direct ELISA (II.2.5.1). The antibody titer refers to the highest dilution at which VP2-specific binding was detectable above background binding to mock- vaccinated mouse antisera as negative control (OD405 nm >0.2 was considered as positive signal). To estimate the cross-reactivity of antibodies with the blocking agent, a PBS sample (serum-free) was also applied to the test. All obtained quantified signals were subtracted from the latter sample to remove cross-reactivity effects and increase the accuracy. After the seventh boost, the final polyclonal antibody titer was higher than 1:500,000 at least in one mouse for each group; mouse 1 for VP2cyto and mouse 5 for VP2apo (figure III.14). This experiment indicated that the tobacco-produced monomeric and trimeric VP2 accumulating either in the cytosol or apoplast is immunogenic in mice.

Page 70: Expression and Characterization of Infectious Bursal

Chapter III Results

64

Figure III.13 Schematic diagram shows mice immunization and blood sampling plan.

Figure III.14 Determination of polyclonal IgG antibody titers in the sera of five mice immunized with (A) VP2apo or (B) VP2cyto, respectively, by indirect ELISA (II.2.5.1). Purified tobacco-derived VP2 (1 µg/ml) was coated to ELISA plate (II.2.5.1). Mouse 2 from the VP2apo group died after the second boost and was removed from the experiment. Serial dilutions of the collected sera were added to the coated plates and bound antibodies were detected by addition of GAMHRP polyclonal antibody (0.2 µg/ml). ELISA reading was performed at OD405 nm after 20 minutes incubation with ABTS substrate at 37°C. X represents the antiserum dilutions. The arrows are indicating the titers above 1:500,000 in the immunized mice.

III.2 Expression and characterization of IBD-VP2 in P. pastoris Since the level of transiently and stably plant-produced VP2apo and VP2cyto was relatively low to perform oral vaccination experiments and, more important, did not assemble to SVPs, which is required to induce a robust immune response, the VP2 was produced in the alternative eukaryotic expression system P. pastoris.

Cloning of IBD-VP2 gene sequence into pPICZ_B vector for generation of III.2.1Pichia expression vectors P. pastoris transformation was performed using pPICZ_B (Invitrogen) as yeast expression vector. The IBD-VP2 encoding fragments were excised from the plant expression vectors pTRAkc-VP2apo and pTRAkc-VP2cyto using EcoRI and XbaI and cloned into the same restriction sites of pPICZ_B, resulting in the final expression vectors pPICZ_B-VP2sec and pPICZ_B-VP2cyto, respectively (figure III.15). As a result, VP2, which was fused to the C-terminal His-tag was expressed under the control of the methanol-inducible alcohol oxidase 1 (AOX1) promoter and terminator, downstream of the CHS 5’ UT sequence with and without the targeting signal sequence

Page 71: Expression and Characterization of Infectious Bursal

Chapter III Results

65

LPH of the murine antibody 24 heavy chain (Vaquero et al., 1999) in pPICZ_B-VP2sec and pPICZ_B-VP2cyto, respectively (vector maps in appendix VII.2B).

Figure III.15 Schematic presentation of the final yeast pPICZ_B based expression vectors. 5’ AOX1 and AOX1 TT: alcohol oxidase 1 gene promoter (methanol-inducible) and terminator, respectively; CHS: 5' untranslated region of the Petroselinum chalcone synthase; LPH: plant codon optimized targeting signal sequence derived from the murine heavy chain of the TMV-specific mAb24; IBD-VP2: cDNA of infectious bursal disease viral protein 2 corresponding to the first 441 amino acid; H6: his-6 tag for detection and purification; pTEF1: transcription elongation factor 1 gene promoter from S. cerevisiae that drives expression of the Sh ble gene in Pichia conferring zeocin resistance; pEM7: constitutive synthetic prokaryotic promoter that drives expression of the Sh ble gene in E. coli conferring zeocin resistance; Sh ble: Streptoalloteichus hindustanus bleomycin resistance gene; cyc1 TT: CYC1 transcription termination region (GenBank accession number M34014), 3’ end of the S. cerevisiae CYC1 gene that allows efficient 3’ mRNA processing of the Sh ble gene for increased stability; (not drawn to scale).

Pichia transformation, screening and expression III.2.2Following positive selection and amplification of both vectors pPICZ_B-VP2sec and pPICZ_B-VP2cyto in E. coli XL1-blue, the IBD-VP2 sequences and reading frames were confirmed by sequencing. Around 3 µg of the purified plasmid DNA was linearized with SacI by overnight incubation and introduced into P. pastoris X-33 cells by electroporation. The transformed X-33 cells were initially screened on YPG plates containing 100 µg/ml zeocin. All of the resulted colonies showed the Mut+ (methanol utilization plus) phenotype on minimal methanol medium based on the screening method described in the EasySelect Pichia Expression kit instruction (Invitrogen, II.2.1.8). Ten positive colonies per construct were selected for analysis of recombinant protein expression (II.2.2.6). After cultivation and induction of VP2 expression by addition of 0.5- 1% (v/v) methanol, yVP2sec and yVP2cyto levels were evaluated in cell extracts and culture media by dot blotting (figure III.16). Glycerol stocks were prepared from clones with the highest yVP2sec level in the media (colony 8) and yVP2cyto level in the cell extract (colony 9).

Page 72: Expression and Characterization of Infectious Bursal

Chapter III Results

66

Figure III.16 Dotblot analyses for screening of yVP2sec or yVP2cyto in recombinant P. pastoris cultures (II.2.3.4). Colonies were inoculated in 3 ml YPD medium and grown o/n at 28 ˚C. At the following day, the expression of recombinant protein was induced by switching the medium to BMMY containing 0.5% (v/v) methanol while the OD600 nm was adjusted to 1.0. Twenty-four hours after expression induction, the cells were harvested by centrifugation at 5,000 ×g for 5 min. Total soluble proteins of the cells was obtained using Y-PER yeast protein extraction reagent (Pierce) according to the manufacture’s instruction. A) Five µl of total soluble proteins of the cell extracts and B) five µl of culture medium were coated on nitrocellulose membrane, following by blocking and immuno-detection using polyclonal anti-VP2 as primary (1:10,000) followed by phosphatase-conjugate goat anti-rabbit (0.2 µg/ml) as a secondary antibody. Detection was performed with NBT/BCIP for 5 min at RT. Wild type Pichia (X-33) extract was used as a negative control (Ctrl-). To provide larger quantities of the recombinant protein for subsequent assays, the best induction time for yVP2 production was evaluated in 0.5 l culture. The production level of yVP2cyto#9 and yVP2sec#8 (figure III.16) was monitored each 24 h during the 96 h expression induction period as recommended by the manufacture’s instruction (EasySelect Pichia Expression kit, Invitrogen). For this purpose, 1 ml of the culture was collected at each day and the presence of yVP2 was determined in the medium culture of yVP2sec and in the cell extract of yVP2cyto using ELISA (II.2.3.1). The accumulation level of yVP2sec in the culture medium increased from about 22 mg/l at 24 h to about 65 mg/l at 96 h of expression induction whereas cytosolic accumulation of yVP2cyto increased from about 20 mg/l after 24 h to 76 mg/l at 96 h expression induction (figure III.17). The highest expression levels were detected at an induction of 96 h, therefore in all subsequent experiments an induction time of 96 h was used.

Page 73: Expression and Characterization of Infectious Bursal

Chapter III Results

67

Figure III.17 Time-course study for evaluation of production levels of Pichia-produced (A) yVP2sec and (B) yVP2cyto accumulating in the medium and inside the cell, respectively. Evaluation was performed using ELISA (II.2.3.1). The expression of the recombinant proteins was induced in 500 ml of the Pichia culture at OD600 nm=1.0 by adding 0.5% (v/v) methanol for the first two days and 1% (v/v) for the following two days until 96 h (II.2.2.6). One milliliter of the cultures was collected at each day. Cells were separated from the medium. Total soluble protein was extracted from the harvested cells. Two microliter of the obtained medium and cell extract were used in ELISA to measure the VP2 content. Two µl of wild type yeast extract alone and in companion with affinity purified VP2cyto (III.1.4) with final concentration of 2-10 ng/µl were used as negative and positive standards, respectively. Rabbit anti-VP2 antibody (1:10000) and GARHRP antibody (0.2 µg/ml) were used as primary and secondary antibodies, respectively. Detection was performed using ABTS as substrate after 10 min incubation at RT. The experiment was performed in biological triplicates. To verify the productivity and integrity of yVP2sec and yVP2cyto, culture medium samples without processing and cell extract samples after extraction using Y-PER protein extraction reagent, obtained from 96 h of expression induction were analyzed by SDS-PAA gel followed by Coomassie staining and immunoblot. As shown in figure III.18, cytosolic expression of yVP2cyto in Pichia resulted in intact VP2, which showed the same size as the purified tobacco-produced VP2cyto (~40 kDa). Small amounts of the protein were also detectable in the medium of yVP2cyto, which probably is originated from disrupted cells during cultivation. The upper band on top of the blot, which was also observable in plant-derived VP2cyto, was due to non-systematic hydrophobic interactions between the monomers. The smeary movement pattern of yVP2sec obtained either from cells or medium in addition to its higher molecular mass compared to VP2cyto indicates the presence of glycan chains attached to yVP2sec. Hyper-mannosylation is common for secreted proteins in Pichia. Interestingly, yVP2sec fractions from cell extract and medium have the same separation pattern and size in the immunoblot. This observation indicates that yVP2sec located inside the cells is glycosylated like yVP2sec secreted to the medium. Since the medium contains less host protein when compared to cell extracts, purification of yVP2sec from medium is easier and faster. In summary, the result demonstrates that yVP2 can be produced in Pichia and accumulates to high levels, both inside the cells or when secreted to the medium. As a next step, the assembly of the

Page 74: Expression and Characterization of Infectious Bursal

Chapter III Results

68

VP2 inside and outside the cell as well as the role of glycosylation on protein assembly was investigated. Further studies on glycan analysis are described below (III.2.4.1).

Figure III.18 SDS-PAGE analysis of yVP2sec and yVP2cyto obtained from selected Pichia colonies (III.2.2) after 96 h expression induction followed by Coomassie brilliant blue staining (A and C) and immunoblot (B and D). Twelve-microliter samples were separated by 12% (w/v) SDS-PAA gel (II.2.3.2): (1) yVP2cyto cell extract, (2) yVP2cyto medium culture, (3) yVP2sec cell extract, (4) yVP2sec medium culture, (5) wild type Pichia (X-33) cell extract and (6) wild type medium culture as negative controls, (7) tobacco-derived and affinity purified VP2cyto and (8) VP2apo as controls (III.1.4). The experiment was performed under reducing (A and B) and non-reducing (C and D) condition. The VP2 corresponding bands were visualized in immunoblot (II.2.3.3) recruiting rabbit anti-VP2 antibody (1:10000) and phosphatase-conjugated goat anti-rabbit (0.2 µg/ml) were used as primary and secondary antibodies, respectively. Detection was performed with NBT/BCIP for 5 min at RT.

Expression and purification of IBD-VP2 and analysis of SVP assembly III.2.3Production of yVP2sec and yVP2cyto was performed on a 500-ml culture with 96 h induction of recombinant protein expression (II.2.2.6). For small-scale protein extraction from the cells (up to 5 ml culture), Y-PER protein extraction reagent was used whereas for larger cell amounts (> 100 ml culture) the extraction was performed in 0.1 M acetate buffer pH 6.0 containing 1 mM PMSF, 1 mM EDTA and 5% (v/v) glycerol. Following extraction of the soluble proteins, the pH was decreased to 4.0. Then the extract was incubated at 4 ºC for 1 h while stirring to precipitate the host proteins. Precipitated proteins were pelleted at 13,000 ×g for 20 min at 4 ºC. The soluble fraction and the pellet were collected. The pellet was resuspended in PBS to the equal initial volume before the pH shift. A sample of the soluble fraction and the pellet was analyzed for VP2 content (figure III.19). Although dropping the pH to 4.0 led to a loss of about 40% of VP2 content the purity was improved about 75%. Therefore the acetate buffer pH of 4.0 containing the same components (breaking buffer; II.2.26) was used for direct extraction of yVP2cyto and yVP2sec from the cells.

Page 75: Expression and Characterization of Infectious Bursal

Chapter III Results

69

Figure III.19 First step purification of yVP2cyto using pH-shift from 6.0 to 4.0. Following total soluble protein extraction from the cells using acetate buffer pH 6.0, the pH was shifted to 4.0 by adding 1 M HCl. The solution was centrifuged at 13,000 ×g/ 20 min/ 4 ºC. The pellet was resuspended in PBS to the equal initial volume before pH shift. Twelve-microliter of the supernatant and the resuspended pellet were separated on 12% (w/v) SDS-PAA gel under non-denaturing condition followed by Coomassie brilliant blue staining (II.2.3.2) (A) and immunoblot (II.2.3.3) (B). Rabbit anti-VP2 antibody (0.12 µg/ml) and goat anti-rabbit AP-labeled (0.2 µg/ml) were used as primary and secondary antibodies, respectively. Purified tobacco-derived VP2cyto was used as a positive control. Densitometric quantification revealed that although this procedure led to the loss of 40% of VP2 content, it increases the purity up to 75%. The data were shown for yVP2cyto but a compatible result with respect to purity and integrity was obtained for yVP2sec extracted from the cells. Due to the low total protein content in the Pichia medium culture, the pH shift strategy was not administered for the purification of yVP2sec from the medium. Hereafter yVP2sec-c and yVP2sec-m were chosen to show yVP2sec from the cell and medium, respectively. As the second purification step, precipitation of yVP2sec-c and yVP2cyto as well as yVP2sec-m was performed by gradual increasing the concentration of ammonium sulfate salt to 50% followed by resuspension of the pellet into 10 ml PBS resulting in the recovery of almost all yVP2 content (60% of the total content) with 90% purity (data not shown). To polish the purified protein and coincidently analyze the protein assembly through studying the molecular mass, the protein samples were applied to a Hiprep 26/60 Sephacryl S-400 HR (GE HealthCare) size exclusion chromatography column with 20- 8000 kDa separation range (II.2.3.5). The eluent was collected in 2 ml fractions and samples (10 µl) from each fraction were used in indirect ELISA (II.2.3.1) to determine the yVP2 content (figure III.20). As depicted (A1) applying yVP2cyto purified from cell extracts onto SEC led to the formation of two major peaks at 152 and 250-ml elution volume. The ELISA response (figure III.20, A2) showed that both peaks contain the major yVP2cyto contents. The same peaks were also obtained by analyzing yVP2sec-c (figure III.20, B1) whereas in case of yVP2sec-m, only a 250-ml peak was observed (figure III.20, B2). Both peaks were absent in the wild type Pichia X-33 cell extract treated by the same purification process (figure III.20, B3). ELISA confirmed that in case of yVP2sec-c and yVP2cyto, both 152-and 250-ml peaks contained the yVP2 protein (data not shown) indicating that cell extracted yVP2

Page 76: Expression and Characterization of Infectious Bursal

Chapter III Results

70

is present in the form of high (152-ml peak) and low molecular mass (250-ml peak) while the yVP2sec-m sample purified from the medium contained only the low molecular mass fraction. For yVP2cyto and yVP2sec-c, 152-ml peak fractions were collected, mixed and concentrated using nanosep centrifugal device with 300 kDa MWCO. Fractions of 250-ml peak were collected separately from yVP2cyto, yVP2sec-c and yVP2sec-m samples, mixed and concentrated using 10 kDa MWCO devices.

Figure III.20 Separation of Pichia-produced yVP2 using size exclusion chromatography (A, B) and further electron microscopy studies (C). The yVP2sec-c and yVP2cyto proteins were produced and purified from Pichia cells (II.2.2.5) using breaking buffer pH 4.0 and 50% (w/v) ammonium sulfate precipitation. Similarly purified Pichia X-33 wild type cell extract was used as a negative control in the purification procedure. yVP2sec-m was extracted from the culture medium directly, using 50% (w/v) ammonium sulfate precipitation. The purified protein was separated by size exclusion chromatography (SEC) using a S-400 HR column with a size separation range 20-8000 kDa: (A1) purified yVP2cyto, (B1) purified yVP2sec-c, (B2) purified yVP2sec-m and (B3) Pichia wild type similarly purified cell extract. The eluted fractions were tested for VP2 content by indirect ELISA (II.2.3.1). ELISA confirmed that both 152-ml and 250-ml peaks containing yVP2(A2) (data shown for yVP2cyto). Analysis of VP2sec-m (B2) as well as X-33 wild type cell extract (B3) as negative control showed that 152 ml-peak is absent in the medium extracted yVP2sec. Electron microscopy for the concentrated 152-ml and 250-ml peaks revealed that only 152-ml peak contained fully assembled IBD-SVPs. Electron micrograph for the 152-ml peak for the purified yVP2cyto named as IBD-SVPcyto (C1) and yVP2sec-c named as IBD-SVPsec (C2) was shown. The conformational changes in IBD-SVPsec are due to the glycosylation. Scale bar represents 50 nm. The concentrated samples were studied by TEM (II.2.3.6). Electron microscopy revealed that the 152-ml peak contained fully assembled IBD-SVPs (figure III.20C) whereas no symmetrical

Page 77: Expression and Characterization of Infectious Bursal

Chapter III Results

71

structure was detected in the 250-ml peak (data not shown), although it contained yVP2 according to ELISA (figure III.20 A2). This shows that the partially purified and concentrated cell extracted yVP2cyto and yVP2sec-c both contained a mixture of two types: fully assembled symmetrical IBD-SVPs with 23 nm in diameter, T= 1 lattice, based on previous studies (Caston et al., 2001; Coulibaly et al., 2005) and not-fully assembled yVP2. Determination of the assembly level in the 250 ml-peak was not possible due to the low resolution of the column at low molecular weight range. The 152 ml-peak representing IBD-SVPs was present both in yVP2cyto and yVP2sec-c samples derived from cell extracts but was absent in the wild type purified cell extract and yVP2sec-m purified from medium (figure III.20B3 and 4), while the second peak eluted at 250 ml was present in the purified yVP2cyto, yVP2sec-c as well as yVP2sec-m samples, obviously containing only VP2 monomers and/or less assembled VP2 proteins. This fact revealed that yVP2cyto and yVP2sec was present as SVPs inside the Pichia cells but not in the medium. This observation may be explained by the pore size of the Pichia cell wall, which prevents the passage of big structures such as SVPs into the medium; consequently, SVP structures were entrapped in the cell space between the cell wall and cell membrane. As it is shown in figure III.20C, SVPs derived from yVP2cyto (IBD-SVPcyto) have regular doughnut-like shape with a central hole whereas SVPs derived from yVP2sec-c (IBD-SVPsec) are less homogeneous and showed a stretched conformation. Also the putative central holes appear filled in IBD-SVPsec. These conformational differences when compared to IBD-SVPcyto are due to glycosylation of IBD-SVPsec, which influences the appearance of yVP2sec SVPs (Figure III.20C1 and C2), which is analyzed in more detail in section III.2.4. Purified yVP2cyto separated into two distinct bands on SDS-PAA gel, one at about 40 kDa, matching with the mass of the VP2 monomer, and the other one with a high molecular mass appear on top of the blot (figure III.21A). Purified yVP2sec-c could be also separated into two bands on a SDS-PAA gel, one at about 60 kDa and one with high molecular mass and a smear between the two bands (figure III.21B). The size difference for yVP2cyto when compared to the yVP2-sec-c and also the smeary band pattern of yVP2sec-c on the SDS-PAGE can be explained by glycosylation of yVP2sec-c. The purity of purified of SVPs derived from yVP2cyto and yVP2sec-c were determined by SDS-PAGE and Coomassie staining followed by densitometric calculation of the protein bands (II.2.2.6). They were calculated about 96.2% and 90%of intact proteins, respectively (figure III.21A, B). In contrast to the SVPs structure that hides the His-tag inside making it inaccessible, yVP2sec-m, which was present in the non-fully assembled form (figure III.20 B2) has an accessible C-terminal His-tag that facilitates purification using IMAC. Therefore, the culture medium of yVP2sec was applied to Ni-NTA column after pH shifted to 8.0. The purification steps were studied by SDS-PAGE. As shown in the figure III.21C, yVP2sec-m binds to the column as expected and elutes by applying immidazole. The eluent showed a smeary movement pattern on the SDS-PAA gel, which extends form the top to about 60 kDa with a purity of about 90% (figure III.21 C). The glycosylation may explain the smeary band pattern since yVP2sec is transported to the secretory pathway in Pichia.

Page 78: Expression and Characterization of Infectious Bursal

Chapter III Results

72

IBD-SVPcyto isolated from the 152-ml fractions of yVP2cyto after SEC were used for subsequent vaccination (III.3) and nanobiotechnology (III.4) studies. The total amount isolated from one-liter culture volume, was 38 mg as determined by the BCA kit (II.2.3.1) with a purity of 96.2%. Since the total expression level of yVP2cyto was 76 mg/l (figure III.17), by using the explained purification procedure, half of the yVP2cyto content was yielded as purified fully assembled IBD-SVPcyto. Purified IBD-SVP derived from yVP2cyto were distributed in separate vials each containing 1 mg and stored at -80°C for the subsequent studies.

Figure III.21 SDS-PAGE analyses of yVP2. Purified IBD-SVPcyto (A) and IBD-SVPsec (B) obtained by pH shift and salt precipitation and polished by SEC: one microgram of the proteins were separated on 12% (w/v) SDS-PAA gel followed by Coomassie brilliant blue staining (cbbs) (II.2.3.2) and immunoblot (ib) (II.2.3.3). Two bands were detected for yVP2cyto, a ~40 kDa-band and a band on top of the gel. Also two bands observed for yVP2sec-c, a ~60 kDa-band and a band on top of the gel. In addition a smear between the bands was visible. The purity was determined about 96.2% for yVP2cyto and 90% for yVP2sec-c using densitometry method (B) Analysis of yVP2sec-m purified by IMAC: A Twelve-microliter sample of each purification step (including culture medium, supernatant after pH shift to 8.0, loading, flow through from Ni-NTA column, dead volume and elution fraction) of culture medium of yVP2sec-m was studied by 12% (w/v) SDS-PAA gel. Further analysis was performed using Coomassie brilliant blue staining (II.2.3.2) and immunoblot under non-reducing condition. For immunoblots (II.2.3.3), rabbit anti-His antibody (0.12 µg/ml) and goat anti-rabbit AP-labeled (0.12 µg/ml) were used as primary and secondary antibodies, respectively. Detection was performed with NBT/BCIP for 5 min at RT. Like yVP2sec-c, yVP2sec-m was smeary which extends form top of the gel to about ~60 kDa. The purity was determined about 90% using densitometric method.

Glycosylation analysis of yVP2sec III.2.4As described above (figure III.6), VP2 has three N-glycosylation sites. The smeary and slower movement of yVP2sec–m and IBD-SVPsec compared to IBD-SVPcyto on SDS-PAGE (figure III.21) indicated glycosylation of the secreted yVP2. Hyper-glycosylation, more specifically, hyper-mannosylation is often observed for secreted recombinant proteins produced in P. pastoris (Hellwig et al., 1999; Promdonkoy et al., 2009). To verify glycosylation of purified IBD-SVPsec, isolated form the cells by pH shift and salt precipitation and polished by SEC (III.2.3; figure

Page 79: Expression and Characterization of Infectious Bursal

Chapter III Results

73

III.21B) as well as purified yVP2sec-m isolated form the culture medium by IMAC (III.2.3, figure III.21C) was applied to further glycan analysis.

Lectin blot III.2.4.1The potential glycosylation of IBD-SVPsec and yVP2sec-m was first analyzed by a lection blot (II.2.4.1) using mannose-specific Galanthus nivalis lectin. Following SDS-PAGE and blotting onto a nitrocellulose membrane, IBD-SVPsec and yVP2sec-m, both having a purity of about 90% (figure III.21B, C), were detected by alkaline phosphatase-conjugated Galanthus nivalis lectin. IBD-SVPcyto (figure III.21A) and tobacco-derived VP2apo (III.1.4) were used as negative and positive control, respectively. Figure III.22 shows that IBD-SVPsec as well as yVP2sec reacted with mannose-specific lectin and consequently were glycosylated. Although VP2apo should contain plant type glycan containing mannose residues in the structure, obviously the low frequency and reduced availability of mannose residues limits the proper reaction with Galanthus nivalis lectin resulting in no visible band. In other words, mannose residues in complex plant glycans are buried by other residues such as GlcNaC whereas in the complex Pichia glycans a large number of mannose residues (9-17 residues) are present. In contrast IBD-SVPcyto, which accumulated in the Pichia cytosol, did not show any binding to the lectin demonstrating that this protein is not glycosylated, as expected.

Figure III.22 Lectin blot analysis of yVP2 produced in Pichia. One microgram of purified yVP2sec-m isolated from the culture medium, IBD-SVPcyto isolated from the cells, IBD-SVPsec isolated from the cells (figure III.21) and tobacco-derived VP2apo (III.1.4) were separated by 12% (w/v) SDS-PAGE followed by blotting onto nitrocellulose membrane. The membrane was blocked using 2.5% (w/v) skimmed milk in PBST. The detection was carried out using mannose specific AP-labelled Galanthus nivalis lectin (0.2 µg/ml) and NBT/BCIP as a substrate.

Enzymatic deglycosylation of yVP2sec-m III.2.4.2As was demonstrated by the lectin blot analysis, both yVP2sec-m and IBD-SVPsec are glycosylated. Since they are both composed of the same protein and passed through the secretory pathway, they only differ in their level of assembly. Therefore, hereafter only yVP2sec-m was studied due to its more simple structure and ease of reaction with the deglycosylation and digestive enzymes. yVP2-sec-m was subjected to deglycosylation using PNGase F (II.2.4.2). IBD-SVPcyto as well as non-deglycosylated yVP2-sec-m was used as controls. The resulting products were

Page 80: Expression and Characterization of Infectious Bursal

Chapter III Results

74

separated by SDS-PAA gel electrophoresis followed by Coomassie staining and lectin blot analysis (figure III.23). After glycan removal through enzymatic digestion from yVP2sec-m and electrophoretic separation, yVP2sec-m had the same size of ~40 kDa as the non-glycosylated IBD-SVPcyto on the Coomassie stained gel (figure III.23A), demonstrating that glycosylation was responsible for the observed mass differences. Lectin blot was also performed with the same samples using Galanthus nivalis lectin (II.2.4.1). Only intact yVP2sec-m reacted with the lectin whereas the deglycosylated yVP2sec-m as well as IBD-SVPcyto showed no reactivity with the lectin indicating that they do not contain any attached glycans. These results showed that the difference in size between yVP2 accumulated in cytosol and yVP2 passed through secretory pathway as well as smeary movement of yVP2sec on SDS-PAA gel is due to the glycosylation.

Figure III.23 SDS-PAGE analysis of the deglycosylated yVP2sec-m using peptide N-glycosidase F (PNGase F) and lection blot. One microgram of (1) deglycosylated purified yVP2sec-m, (2) purified IBD-SVPcyto (3), purified yVP2sec-m without deglycosylation were separated by 12% (w/v) SDS-PAA gel electrophoresis followed by Coomassie brilliant blue staining (A) and lectin blot (B) using AP-labelled Galanthus nivalis lectin (0.2 µg/ml) and NBT/BCIP as a substrate (II.2.4.1).

Glyco-proteomic analysis of yVP2sec III.2.4.3The glycan composition of purified yVP2sec-m isolated from the Pichia culture medium (figure III.21B) was analyzed by LC-MS. Following IMAC purification and dialysis of the elution fraction against PBS, yVP2sec-m was digested with trypsin and chemotrypsin and finally analyzed by LC-MS (II.2.4.3). As described above VP2 contains three potential glycosylation sites (figure III.6; aa 46, aa 121, aa 396). The result of LC-MS is shown in figure III.24 and summarized in table III.2. Mass spectrometry revealed that high mannose type glycan are abundant at all three sites of yVP2sec-m. A monosaccharide (such as mannose) with six carbon atoms was shown as Hex thus HexNac2Hex11 with the 33.71% average abundance between the three sites of followed by HexNac2Hex10 (31.67 %) are the most prominent glycan compositions. HexNac2Hex9 (with 8.4% average abundance) is the simplest recognized glycan-type and more complex-types such as HexNac2Hex12-17 are also present. Glycosylation analysis of yVP2sec-m confirmed that the signal peptide of the murine heavy chain antibody (LPH) is also functional in Pichia in leading yVP2 to the secretory pathway and finally to the medium. All three N-glycan sites were glycosylated with variation in glycan compositions and their frequencies are described in Table III.2 in more detail.

a

Page 81: Expression and Characterization of Infectious Bursal

Chapter III Results

75

Table III.2 Glycan composition and their ratio at the three N-glycan sites (1, 2 and 3) of IMAC-purified yVP2sec-m (III.2.3) studied by liquid chromatography-mass spectrometry (LC-MS).

Glycan type Relative frequency (%) Site 1 Site 2 Site 3

HexNAc2HeX9 3.39 9.33 12.68 HexNAc2HeX10 34.13 38.59 22.30 HexNAc2HeX11 39.86 35.51 25.78 HexNAc2HeX12 9.83 10.14 16.31 HexNAc2HeX13 6.28 6.43 10.03 HexNAc2HeX14 3.28 0.00 5.57 HexNAc2HeX15 2.59 0.00 3.97 HexNAc2HeX16 0.66 0.00 2.09 HexNAc2HeX17 0.00 0.00 1.25

Figure III.24 Liquid chromatography-mass spectrometry (LC-MS) of the three N-glycosylation sites of Pichia-produced and affinity-purified yVP2sec-m. yVP2sec-m was purified by IMAC (III.2.3) from the medium of the Pichia culture, dialyzed against PBS and digested with trypsin and chemotrypsin (II.2.4.3). The resultant peptides were applied to LC-MS analysis (II.2.4.3). The spectrum of glycosylated peptides are shown in figure A (site 1), B (site 2) and C (site 3) based on their position in the yVP2sec-m sequence (figure III.6).

Page 82: Expression and Characterization of Infectious Bursal

Chapter III Results

76

III.3 Chicken immunization with yVP2cyto Aglycosylated yeast-produced VP2 either in purified (IBD-SVPcyto) (III.2.3) or cell-capsulated (Pichia yVP2cyto) form (III.2.2) was used in chicken immunization experiments against IBDV. The experiments were performed at Institute of Poultry Diseases, faculty of Veterinary Medicine, Freie Universität Berlin. To diminish regulatory issues regarding the release of genetically modified organisms (GMOs) during transport and immunization, Pichia cells (cell-capsulated yVP2cyto) containing the antigen were subjected to inactivation procedure. For this purpose, the freshly harvested cells were applied into two thermal cycles of 70 °C and 15 °C each for 5 min. About 16 g of the cells was harvested by centrifugation from 1 l culture and resuspended in distilled water to a final volume of 40 ml in a 50-ml tube and subsequently applied to the thermal cycles. To confirm proper inactivation, a vitality test was performed on yeast selection solid medium, Sabouraud agar (Heipha Dr Müller GmbH), in three cell concentration and three repetitions (data not shown). The result showed that about 99.3 % of the cells were inactivated after the first cycle and applying the second cycle inactivated the rest. The inactivated cells were stored at -80 °C for one day and then freeze-dried. To test the stability and integrity of the antigen after cell inactivation and freeze-drying, a sample of the cells was subjected to purification of yVP2 as described before (III.2.3). The resulting sample was studied by SDS-PAGE and SEC using purified IBD-SVPcyto as a control. As illustrated in the figure III.25, the movement pattern of yVP2cyto on SDS-PAA gel and SEC remains intact after inactivating and freeze-drying and are identical to the purified IBD-SVPcyto (III.2.3). The elution pattern of yVP2cyto in SEC derived from inactivated Pichia is also comparable with the purified IBD-SVPcyto (III.2.3). The major fractions eluted at 152-ml peak, which corresponds to IBD-SVPcyto (figure III.20). This observation indicates that the heat inactivation does not impair the structure of the antigen; yVP2 remains intact and can be purified in the form of SVPs. The lyophilized inactivated Pichia yVP2cyto cells as well as purified IBD-SVPcyto were used in oral immunization of the chickens. To evaluate the dosage of the cell-capsulated vaccine, the yVP2 content inside the cells was measured using SDS-PAGE followed by immunoblot and densitometric method using AIDA software. The known concentrations of purified IBD-SVPcyto (II.2.3) measured using BCA kit were applied to the same blot and used as standards. Calculations indicated that in one milligram of freeze-dried inactivated Pichia yVP2cyto powder about 16 µg of yVP2 exists. Two-week old SPF white Leghorn chickens were immunized using either the freeze-dried inactivated Pichia yVP2cyto or purified IBD-SVPcyto. The immunization and challenge scheme as well as immunization groups are shown in figure III.26. Both antigen vaccine types were applied four times, weekly: each in two different doses, high and low: 250 mg or 25 mg of freeze-dried inactivated Pichia yVP2cyto and 500 µg and 50 µg of purified IBD-SVPcyto. Each vaccine dose also was tested in companion with oral adjuvant. 50 µg CpG ODN (table II.1.4) plus an adequate amount of NaF (100 mg per kg of chicken weight) was used as an oral adjuvant mixture. As a negative control, 250 mg of freeze-dried inactivated wild type Pichia was applied to the group 9 (mock-immunized) in order to study the side effects of the expression platform on the chickens.

Page 83: Expression and Characterization of Infectious Bursal

Chapter III Results

77

Figure III.25 Analysis of stability and integrity for IBD-SVPcyto after Pichia cell inactivation using SDS-PAGE and SEC. Twelve microliter of: (1) Purified yVP2cyto derived from freeze-dried inactivated Pichia cells, (2) Purified yVP2cyto derived from active freeze-dried Pichia cells, (3) Purified IBD-SVPcyto (III.2.3) as a positive control, were separated on 12% (w/v) SDS-PAA gel followed by Coomassie brilliant blue staining (A) and immunoblot (B) using rabbit anti-VP2 (1:10,000) and GARAP (0.2 µg/ml) as primary and secondary antibodies, respectively, and NBT/BCIP as a substrate (C) Chromatographic profile obtained by separation of the corresponding samples 1, 2 and 3 using SEC S-400 HR column and Äkta Explorer (II.2.3.5). The Pichia cells and the purified antigen were administered in one-ml and 0.5-ml volumes, respectively. Oral immunization was carried out by oral gavage into the chickens’ crop. AviPro Gumboro vac (Lohmann Animal Health) vaccine composed of attenuated virus was also applied orally according to the manufacturer’s instruction as a positive control. To test the ability of IBD-SVPcyto to induce anti-IBDV antibody production, the most direct method with confirmed efficacy is to use the purified antigen and deliver by injection. Therefore, 20 µg of purified IBD-SVPcyto were delivered intramuscularly in combination with adjuvant 100 (Gerbu) in 100 µl total volume for the chickens in group 10. Two groups of the chickens not receiving any immunization treatment were used as negative controls of virus challenge and vaccine treatment, respectively. All chickens except those of group 13 (vaccine control) were challenged with a classical IBDV strain, MB3, inoculated ocularly to the chickens at 28 day post immunization (dpi). Blood and cloakal samples were collected according to table II.7; prior to each immunization, at viral challenge as well as death/euthanization days. In addition weight of the birds was recorded and monitored as a health index each week during the whole period of the experiment. During a week following challenge, the observed clinical signs were recorded and at the last day (35 dpi) the surviving chickens were euthanized, bursa Fabricious was extracted, the bursa weight to body weight (BF/BW) ratios were determined. Also, the bursal damage score, viral clearance and mucosal antibody response were investigated.

Page 84: Expression and Characterization of Infectious Bursal

Chapter III Results

78

Figure III.26 Immunization of the chickens randomly allocated between 13 immunization groups. (A) Immunization scheme: Two-week old chickens were immunized at the indicated times (schematized as black bars) by either orally (groups 1-9) or intramuscularly (group 10). Oral immunization was carried out using 250 mg and 25 mg of freeze-dried Pichia (groups 1-4) or 500 µg and 50 µg of purified (III.2.3) Pichia-derived IBD-SVPcyto (groups 5-8) with and without oral adjuvant mixture. The yeast P. pastoris X-33 and Avipro Gumboro vac (Lohmann Animal Health, Cuxhaven, Germany), which composed of attenuated virus, were applied as mock-immunized (group 9) and positive (group 12) controls, respectively. Intramascular immunization was carried out using 20 µg of purified Pichia-derived IBD-SVPcyto mixed with adjuvant 100 (Gerbu). Two groups were remaining unimmunized, one was applied to challenge experiment and named as a challenge control (group 11) and the other was kept unchallenged as a vaccine control (group 13). The gray bar indicates the time interval following viral challenge; arrow indicates the time point of euthanization of the survivorss.

Health monitoring, clinical signs III.3.1The weight gaining of the chickens as a health index was monitored during the experiment. No significant difference (p> 0.05) in weight increase was observed between all groups during the immunization, which indicates that the vaccine was not interfering in growth rate (figure III.27). The typical IBD clinical signs including ruffled feathers, depression, feces form and color as well as death were investigated daily after the viral challenge. Clinical signs of IBDV usually appear at day 2 post infection and start to disappear at day 4 due to rapid recovery of the survivors

Page 85: Expression and Characterization of Infectious Bursal

Chapter III Results

79

(Mahgoub, 2012). All the detailed clinical sign observations are summarized in table VII.2 in appendix VII.6.

Figure III.27 Weight of the chickens was monitored as a health index during immunization. The variations among the groups (II.2.6.8) of vaccinated and non-vaccinated are not significant (p> 0.05). Briefly, at the 2nd day post challenge (dpc), the moderate to severe clinical signs appeared among the birds in the groups of mock-immunized (group 9) and challenge control (group 11). Most of the birds in the other groups developed slight clinical signs. At the 3rd dpc, the mortality started among mock-immunized (group 9) and challenge control (group 11) as well as birds in groups 5 and 6 that received 25 mg of Pichia yVP2cyto without and with oral adjuvant, respectively. The other chickens in these groups also showed severe clinical signs. Other groups that orally immunized with cell-capsulated antigens showed slight to moderate clinical signs. The mortality started among the latter groups at day 4th dpc with lower frequency (two in group 1 and one in group 4) compared to mock-immunized and challenge control. At this day slight clinical signs appeared in birds of group 7 and 8, which received 500 µg of the purified IBD-SVPcyto without and with oral adjuvant, but they recovered until the next day. In contrast, among birds of groups 5 and 6, which received 25 mg of Pichia yVP2cyto, similar to mock-immunized (group 9) and challenge control (group 11) mortality as well as moderate to severe clinical signs was observed. At the 5th dpc, the survivors showed the first signs of recovery. Chickens in group 10 that received the vaccine parenterally, group 12 that received the live-vaccine showed no clinical signs following viral challenge. These observations indicate that oral immunization either with cell capsulated or purified antigen is immunogenic but the potency of conferring immunity is dose and formulation-dependent. 500

Page 86: Expression and Characterization of Infectious Bursal

Chapter III Results

80

µg of purified IBD-SVPcyto more efficaciously inhibits clinical signs compared to 50 µg of purified IBD-SVPcyto and even 250 mg of Pichia yVP2cyto that containing 4 mg of yVP2cyto.

Immune response and protection analysis III.3.2Mucosal immunization, in contrast to parenteral immunization, has the advantage of inducing both systemic and local mucosal immune response. In response to antigen application, secretory immunoglobulin A (SIgA) is generated at stimulated mucosal surfaces. This antibody isotype is produced by B cells localized underneath of the mucosal surface. Since secretory IgA has high affinity to the mucus, excreted mucus is an enriched source of IgA. Following oral immunization, secretion of IgA launches in gut lamina proporia. Thus feces samples have been used as a source for mucosal immune response analysis. In addition to SIgA, some IgA molecules without secretory component pass through paracellular passive transfer and also reach to the secretions (Johansen et al., 1999 a, b). Since oral immunization has the ability to induce systemic immune responses in peripheral lymphoid organs, the antigen-specific serum IgY, IgM and IgA antibody responses, other than mucosal immune response in feces, should be studied to evaluate the efficacy of the vaccine. For this purpose, peripheral blood and feces samples were collected weekly before each immunization at 0, 7, 14, 21 dpi, before viral challenge at 28 dpi and at the death/euthanization day and were studied using indirect VP2-based ELISA (II.2.6.2). Pichia-derived purified IBD-SVPcyto was used as an antigen coated in microtiter plate to study the specific immune response against IBDV in the chickens. To study systemic and local IgA immune response the 1:5 diluted sera and feces extract (II.2.6.4) were applied to the coated antigen in microtiter plate (II.2.6.4). For preparation of feces extract or cloakal swab sample were used (II.2.6.2). The detection was carried out using horseradish peroxidase-conjugated goat anti-chicken IgA (5 µg/ml). Finally, ABTS was used as substrate. The mean OD405 nm values over that the mean absorbance plus two times standard deviation of the pre-immune sera (> 0.2) was considered as positive antibody responses. IgA response was undetectable (<0.2) in all samples collected before challenge among all chickens (data not shown). But about a week after challenge IgA response appeared in the sera and most feces samples of vaccinated chickens (figure III.28). This response was absent in challenge (group 11) and vaccine (group 13) controls. However, the mock-immunized group (group 9) that received 250 mg of Pichia wild type cells also was seropositive. This unexpected observation might be explained by the reaction of generated chicken antibodies against Pichia proteins since the coated IBD-SVPcyto derived from Pichia. The strength of the signals is not significantly differing from the mock-immunized group and other orally immunized groups. Only in group 12 that orally immunized with attenuated virus, the response is higher than in the mock-immunized and other groups. As a conclusion, secretory IgA response was not strongly stimulated regardless of route of immunization except in the group 12 which received live attenuated virus.

Page 87: Expression and Characterization of Infectious Bursal

Chapter III Results

81

Figure III.28 Serum IgA and intestinal IgA response in chickens immunized with freeze-dried inactivated cell-capsulated and purified IBD-SVPcyto after viral challenge. Oral immunization was carried out using 250 mg and 25 mg of freeze-dried Pichia (groups 1-4) or 500 µg and 50 µg of purified (III.2.3) Pichia-derived IBD-SVPcyto (groups 5-8) with and without oral adjuvant mixture in all cases. The yeast P. pastoris X-33 and Avipro Gumboro vac (Lohmann Animal Health, Cuxhaven, Germany), which composed of attenuated virus, were applied as mock-immunized (group 9) and positive (group 12) controls, respectively. Intramascular immunization was carried out using 20 µg of purified Pichia-derived IBD-SVPcyto mixed with adjuvant 100 (Gerbu). Two groups were remaining unimmunized, one was applied to challenge experiment and used as a challenge control (group 11) and the other was kept unchallenged as a vaccine control (group 13). All chicken sera were diluted 1:5 in PBS and feces samples were prepared by rinsing the cloak swab in 500 µl PBS containing protease inhibitor cocktail for one hour and used undiluted in VP2-based ELISA (II.2.6.4). The detection was carried out using horseradish peroxidase-conjugated goat anti-chicken IgA (5 µg/ml). Finally, ABTS was used as substrate. The mean OD405 nm values over that the mean absorbance plus two times standard deviation of the pre-immune sera (> 0.2) was considered as positive antibody responses. Each bar is the average of OD405 nm reading from five individual serum samples or five individual fecal samples with standard error indicated by error bars. The IgM and IgY immune response were also investigated by ELISA as described (II.2.6.2). The OD405 nm values above the mean absorbance plus two times standard deviation of the pre-immune sera (> 0.2) were considered as positive antibody responses (seropositive). As it is illustrated in the figure III.29A and B, the IgM and IgY antibody levels at day 0 dpi of all chickens and the vaccine control (group 13) showed that all chickens were seronegative for IBDV antibodies prior to vaccine administration. Following immunization but before viral challenge, IgM response (figure III.29B) was only detectable in the chickens of group 10 that received the purified antigen intramuscularly. Subsequent to viral challenge, all groups were stimulated and started to produce IgM upon the virus entrance. The levels of IgM in the chickens only in group 10 drops down after viral challenge. This observation might be explained by quick class switch from IgM to IgY in this group indicating that intramascular immunization triggers the immune system more efficacious in the context of affinity maturation and isotype switching.

Page 88: Expression and Characterization of Infectious Bursal

Chapter III Results

82

Figure III.29 Immunization of chickens with freeze-dried inactivated cell-capsulated and purified IBD-SVPcyto. Reactivity of serum IgM (A) and IgY (B) of the chicken sera with IBD-SVPcyto were determined by ELISA (II.2.6.4). The serum samples were collected before each immunization, before viral challenge and before dead/euthanization. The 1:50 diluted sera were applied to the coated antigen in microtiter plates. The detection was carried out using either horseradish peroxidase-conjugated goat anti-chicken IgM (5 µg/ml) or horseradish peroxidase-conjugated goat anti-chicken IgY (2 µg/ml). Finally, ABTS was used as substrate. The mean OD405 nm values over that the mean absorbance plus two times standard deviation of the pre-immune sera (> 0.2) was considered as positive antibody responses. The mean of absorbance at 405 nm was shown for five chickens in each group with standard error indicated by error bars. The 0.2 value as a cut off value was shown by blue line; chickens with higher antibody titer were considered as seropositive.

Page 89: Expression and Characterization of Infectious Bursal

Chapter III Results

83

In contrast to IgM, IgY responses was observed in groups 8, 10 and 12 prior to viral challenge (figure III.29B). However, it is predominantly observed in chickens of group 10 that received the purified antigen intramuscularly followed by group 12 that received attenuated virus. After viral challenge, chickens of groups 10 and 12 increased their IgY titer. Also chickens of groups 2 that received 250 mg Pichia yVP2cyto with adjuvant and 8 that received 500 µg of purified IBD-SVPcyto with adjuvant showed a potent IgY response leading to full protection (table III.4). Although only 80% of the chickens are IgY seropositive in group 3, which received 25 mg Pichia yVP2cyto and group 7, which received 500 µg IBD-SVPcyto, all were fully protected against the virus. It was speculated that in these cases IgM provides protection. This was also observed among the chickens of groups 4 and 5. In field studies, the anti-IBDV antibody titer in the serum is determined by using the commercial IDEXX IBD-XR Ab test to assess the status as well as the potency of the serum antibodies in identification of IBDV. In other words, it evaluates the protective antibody titer against IBDV. In this experiment, all the blood samples collected from the chickens were studied using the IDEXX IBD-XR Ab kit according to the manufacture’s instruction. Chickens with titer above 396 were considered as seropositive (Rong et al., 2007). Among all the chickens, only chickens in group 12 immunized with live attenuated virus and three birds in the group 10, which received the purified antigen intramuscularly and two birds in group 8, which orally received 500 µg purified IBD-SVPcyto showed titers over 396 before viral challenge. After the challenge all the survivors were IDEXX IBD-XR seropositive. The results are summarized in the table III.4. The result confirmed the previous speculation that IgM could be protective antibody as IDEXX IBD-XR shows the protective antibody titer irrespective whether it originates from either IgY or IgM. The best method for evaluation of vaccine efficacy is to characterize the survivors after challenge with the infectious virus. In this study, the mortality rate was recorded during one week post challenge (table III.4 and III.5). All chickens that died before the 7th dpc were considered as mortality affected by the virus. After this period, all survivors were euthanized at the 7th dpc. Accounting the survivors in each group one week after viral challenge indicated that orally immunized chickens in group 2 (immunized with 250 mg Pichia yVP2cyto with adjuvant), group 3 (immunized with 25 mg Pichia yVP2cyto), group 7 (immunized with 500 µg of purified IBD-SVPcyto), group 8 (immunized with 500 µg of purified IBD-SVPcyto with adjuvant), group 12 (immunized with attenuated virus) as well as group 10 (immunized intramuscularly) were fully protected. This result indicates that oral administration of yVP2cyto both in the purified or cell-capsulated form is protective. However as previously mentioned (III.3.1) the potency of conferring immunity is dose and formulation-dependent. 500 µg of purified IBD-SVPcyto regardless of adjuvant effect gives full protection whereas 250 mg of Pichia yVP2cyto regardless of adjuvant effect gives 80% protection. Applying 50 µg of purified IBD-SVPcyto regardless of the presence of adjuvant gives only 50% protection.

Page 90: Expression and Characterization of Infectious Bursal

Chapter III Results

84

Tab

le II

I.4 T

he fr

eque

ncy

of Ig

M, I

gY a

nd ID

EX

X IB

D se

ropo

sitiv

e ch

icke

ns b

efor

e an

d af

ter

vira

l cha

lleng

e an

d th

e pr

otec

tion

rate

.

Page 91: Expression and Characterization of Infectious Bursal

Chapter III Results

85

A successful vaccination other than conferring full protection should prevent virus replication inside the body and clear the contaminated host cells. Therefore the vaccination should induce CTLs in order to clear the virus from the vaccine recipient following viral challenge. There is a chance that survivors induce protective immune response but are unable to completely remove the virus particles from the body. In such cases, although the host survives, it can act as a virus carrier to the other chickens in the flock. Therefore, the presence of virus in the bursa of dead/euthanized chickens was analyzed by ELISA (II.2.6.6) and the corresponding results are shown in table III.5. Oral vaccination with either purified or cell-capsulated antigen partially prevented virus replication inside the body with various successes. The viral prevention rates among all orally immunized groups are only between 0-10% except group 8 that immunized with 500 µg of purified IBD-SVPcyto with adjuvant which showed predominant viral clearance potency with 60% prevention (table III.5). The bursa of Fabricius (BF)/body weight (BW) ratio is an index to evaluate the viral morbidity and shows the immunity level. It is calculated using the formula: BF weight (g)/BW (Kg). In this study, the significant difference between the mean of this ratio was revealed using one-way ANOVA test followed by Duncan’s test with alpha level of 0.05 (table III.5). The ratios for the survived chickens in groups 1-7 and those of mock-immunized (group 9) and unvaccinated (group 11) were significantly different (p< 0.05) from groups 8, 10, 12 and unchallenged group 13. This observation indicated that among orally vaccinated groups only 500 µg of the antigen in the purified form in companion with adjuvant (group 8) worked as efficacious as either attenuated virus or parenteral vaccine to confer protection. However, the 250 mg of Pichia yVP2cyto with adjuvant (group 2), 25 mg of Pichia yVP2cyto(group 3) as well as 500 µg of the purified IBD-SVPcyto even without adjuvant gives full protection but less success in virus removal (maximum 10%) and prevention of bursal atrophy. The bursal lesion was also studied according to the previously described scoring system based on histopathological and morphological studies (II.2.6.7). The result of the observations and the resulting scoring in case of bursa extracted from chickens of each group is summarized in table III.5 and in more detail described in the appendix VII.6. All chickens in vaccine control group (group 13) as well as chickens that received attenuated virus (group 12) and those that received the purified antigen intramuscularly (group 10) showed score 0 in the histopathological study with no bursal damage. The unvaccinated and challenged chickens (group 11) and mock-immunized chickens (group 9) as well as those that received 50 µg of the purified IBD-SVPcyto, showed the highest lesion score (= 2.2). Other groups showed moderate bursal lesion score (1-1.6) but chickens of group 8 that received 500 µg of the purified IBD-SVPcyto with adjuvant and group 2 that received 250 mg Pichia yVP2cyto with adjuvant showed score 1 which is the minimum among orally immunized chickens.

Page 92: Expression and Characterization of Infectious Bursal

Chapter III Results

86

Tab

le I

II.5

The

viru

s cl

eara

nce

abili

ty b

y th

e im

mun

e sy

stem

of t

he im

mun

ized

bird

s w

as e

valu

ated

7 d

pc. D

etec

tion

of th

e vi

rus

in th

e ex

tract

ed

burs

a fa

bric

ious

is a

n in

dica

tion

of th

e pr

esen

ce o

f viru

s. Fo

r bet

ter c

ompa

rison

, the

pro

tect

ion

rate

is in

dica

ted

in th

e la

st c

olum

n.

Page 93: Expression and Characterization of Infectious Bursal

Chapter III Results

87

III.4 IBD-SVPcyto as a nanoplatform SVPs are a type of virus nanoparticles (VNPs) that are genome-free with a huge potential application as nano-platforms. In this section the tolerance of the produced IBD-SVPcyto will be studied against heat, pH and organic solvent, which are commonly applied to the production procedure of functionalized VNPs. Moreover, the addressability of solvent-exposed amine and carboxylic groups on the surface of IBD-SVPcyto will be tested via traditional bioconjugation methods (Taghavian et al., 2012).

Solvent stability of IBD-SVPcyto III.4.1The stability of nanoparticles in solvents is an important requirement because bioconjugation is usually carried out in an environment containing up to 20% (v/v) DMSO, whereas mineralization (e.g. with silica) requires up to 20% (v/v) ethanol as a solvent. Therefore, the IBD-SVPcyto (III.2.3) was dissolved in ethanol/water and DMSO/water solutions, with PBS as a control. The electrophoretic mobility of IBD-SVPcyto was tested under native conditions following O/N incubation at RT. As it is shown in figure III.30A, particles prepared in up to 20% (v/v) solutions of either ethanol or DMSO exhibited the same mobility as those prepared in PBS. SVP solubility decreased gradually as the ethanol concentration increased from 5 to 20% (v/v) (figure III.30A, B) and complete precipitation occurred in a 50% (v/v) solution. In contrast, there was little change in SVP solubility as the DMSO concentration increased to 20% (v/v), and only partial precipitation occurred at 50% (v/v) along with an increase in electrophoretic mobility (figure III.30A, B). Although the particles were stable in up to 20% (v/v) of DMSO, amine and carboxylate coupling worked also using a 10% (v/v) solution (data not shown). Conformational changes in the particles were monitored by Dynamic light scattering (DLS), reflecting changes in the mean particle hydrodynamic radius. Two peaks were observed, the first (~ 12 nm) corresponding to the hydrodynamic radius of single intact particles and the second (~ 100 nm) corresponding to particle aggregates (figure III.30C). Increasing the ethanol concentration to 20% (v/v) appeared to have no impact on conformation, most of the particles still gathering in the first peak. In contrast, some aggregation was apparent at 20% (v/v) DMSO because the particles were distributed evenly between the two peaks (figure III.30C). At 50% (v/v) DMSO, there was a significant shift towards the second peak as most of the particles aggregated. TEM analysis confirmed the changing conformation of particles at high solvent concentrations (figure III.30D). These results show that IBD-SVPcyto can tolerate up to 20% (v/v) ethanol or 20% (v/v) DMSO, confirming their suitability for bioconjugation and mineralization reactions under standard conditions.

Page 94: Expression and Characterization of Infectious Bursal

Chapter III Results

88

Figure III.30 Stability of IBD-SVPcyto in ethanol–water and DMSO–water mixtures determined by NAGE (A) (II.2.7.2), A280 spectroscopy (B) (II.2.7.3), DLS (II.2.7.4) (C) and TEM (D) (II.2.3.6). Particles were dissolved in 5, 10, 20 and 50% (v/v) ethanol–water and DMSO–water, respectively. After 24 h the samples were centrifuged at 13000 ×g for 20 min and the supernatant was analyzed. All the protein precipitated in 50% ethanol so in this case the pellet was used for TEM. Scale bar = 100 nm. As a control (ctrl), SVPs were dissolved to the same concentration in PBS. Data shown in panel B are derived from three independent experiments; error bars show standard deviations.

Temperature stability of IBD-SVPcyto III.4.2The electrophoretic mobility of the IBD-SVPcyto under native conditions remained constant after incubation for 1 h at temperatures between 25 and 60°C (figure 31A). No precipitation or conformational changes were observed within this temperature range (figure 31B, C, D). This indicates that IBD-SVPcyto are relatively heat resistant, with temperature stability comparable to that of CPMV particles (Strable and Finn, 2009). At temperatures above 60°C, extensive precipitation was observed along with significant changes in electrophoretic mobility and conformation (figure 31A–D).

Page 95: Expression and Characterization of Infectious Bursal

Chapter III Results

89

Figure III.31 Stability of IBD-SVPcyto at different temperatures determined by NAGE (A) (II.2.7.2), A280 spectroscopy (B) (II.2.7.3), DLS (II.2.7.4) (C) and TEM (D) (II.2.3.6). SVPs were dissolved in PBS to a final concentration of 1 mg/ml and incubated for 1 h at 40, 50, 60, 65, 68, 72, 75 and 80°C in a gradient thermocycler. The samples were then cooled on ice, centrifuged at 13000 ×g for 20 min at 4°C, and the supernatant was used for analysis. Scale bar = 100 nm. SVPs incubated at room temperature were used as the control. Data shown in panel B are derived from three independent experiments; error bars show standard deviations.

Stability of IBD-SVPcyto in different pH environment III.4.3SVP preparations were dispersed in buffer solutions with pH values of 2.5, 5.5, 8.0 and 9.0. PBS (pH 7.4) was used as the reference buffer. The IBD-SVPcyto remained stable after incubation in these buffers at RT for 24 h. The electrophoretic mobility of the SVPs was similar over the pH range 5.5–9.0, but increased slightly at pH 2.5 probably reflecting changes to the net charge on the particle surface (figure III.32A). There was no significant difference in particle solubility over the pH range 2.5–9.0, suggesting there was no aggregation or precipitation (figure III.32B) and in agreement there was no significant shift in the particle hydrodynamic radius (figure III.32C). TEM revealed slight conformational change at pH 5.5 and pH 9.0 but this did not affect the particle radius (figure III.32D).

Figure III.32 Stability of IBD-SVPcyto in different pH environments determined by NAGE (A) (II.2.7.2), A280 spectroscopy (B) (II.2.7.3), DLS (II.2.7.4) (C) and TEM (D) (II.2.3.6). Particles were dissolved in buffers at pH 2.5, 5.5, 7.4, 8.0 and 9.0, to final concentration of 0.5 mg/ml. The samples were incubated overnight at room temperature and centrifuged at 13000 ×g for 20 min at 4 °C, and the supernatant was used for analysis. Scale bar = 100 nm. SVPs incubated at pH 7.4 were used as the control. Data shown in panel B are derived from three independent experiments; error bars show standard deviations.

Page 96: Expression and Characterization of Infectious Bursal

Chapter III Results

90

Addressability of IBD-SVPcyto III.4.4IBD-VP2 contains two cysteine residues that do not participate in disulfide bond formation, but the side chains are not exposed to the solvent and are therefore poor targets for bioconjugation (figure III.33). However five lysine residues present accessible amine groups (Lys 81, 192, 309, 316 and 381) and there are 13 acidic residues that present accessible carboxyl groups (Asp 300, 311, 355, 378 and Glu 51, 83, 138, 190, 213, 267, 279, 287 and 323). Each IBD-SVP comprises 60 VP2 monomers, resulting in a total of 300 potentially addressable amines and 780 carboxyl groups per particle.

Figure III.33 Structural analysis of IBD-SVPcyto. (A) Surface Lys residues shown in yellow and reactive amine side chains highlighted in red. (B) Surface Glu and Asp residues shown in orange and pink, respectively. The reactive carboxyl side chains of both residues are highlighted in blue. (C) Surface Cys residues are shown in purple, but the potentially reactive thiol groups are hidden inside and not exposed to the solvent. (D) The IBD-SVP asymmetric unit (VP2) is shown in green with five solvent-exposed amine residues in yellow. (E) Nine and four solvent-exposed Glu and Asp residues rendered in orange and pink respectively. All the figures of protein coordinates were rendered using the program PyMOL using the file with PDB code 2DF7 (Lee et al., 2006a).

Page 97: Expression and Characterization of Infectious Bursal

Chapter III Results

91

Addressability of amine groups III.4.4.1The number of addressable amines on each IBD-SVPcyto was determined by coupling DyLight 488 NHS ester to the particles using the DyLightTM Antibody Labeling Kit (Pierce). The amount of 1 mg of IBD-SVPcyto (2 mg/ ml) per dye batch and the dye concentration (c) was determined by measuring its absorbance (A) at 493 nm and using the dye-specific molar extinction coefficient (e) of 70 000 (A = cde, with d = pathlength). This showed that on average 63 dye molecules were attached to each particle. Biotinamidohexanoic acid NHS ester has been used to determine whether biotin could be installed on the accessible amine groups. The reaction was carried out at pH 7.4 because under these conditions only the amine groups of lysine residues exposed to the solvent are expected to react with the ester (figure III.34).

Figure III.34 Labeling of amine groups on the surface of IBD-SVPcyto using biotinamidohexanoic acid N-hydroxysuccinimide ester. Schematic IBD-SVP refers to Protein Data Bank reference 2DF7 (Lee et al., 2006a). The coupling of biotin to IBD-SVPcyto was confirmed by Western blot using alkaline phosphatase-conjugated streptavidin (figure III.35A). SEC showed that the particle mass had increased after biotinylation (figure III.35B). Staining with gold-labeled anti-biotin antibodies followed by TEM also confirmed that the particles were labeled with biotin (figure III.35 C).

Figure III.35 Biochemical analysis of biotinylated IBD-SVPcyto by Western blot (A) (II.2.3.3), SEC (B) (II.2.3.5), TEM (II.2.3.6) after amine conjugation (C) (II.2.7.5) and TEM after carboxyl conjugation (D)

Page 98: Expression and Characterization of Infectious Bursal

Chapter III Results

92

(II.2.7.5). Western blot was performed either by subsequent incubation with rabbit anti-IBD-VP2 (1: 10,000) and phosphatase-conjugated goat anti-rabbit antibody (2 µg/ml) each for 1 h at room temperature (A1) or by incubating with alkaline phosphatase-conjugated streptavidin (final concentration 0.2 mg/ ml in PBS) for 1 h at room temperature (A2). The signal was detected with NBT/BCIP (Sigma); IBD-SVPcyto (lane 1), biotinylated IBD-SVPcyto via amine and carboxyl chemistry (lane 2 and 3 respectively) were analyzed. SEC was carried out using a Hiprep 26/60 Sephacryl S400 column and Äkta Explorer (II.2.3.5). Samples were analyzed at a flow rate of 0.5 ml min, and elution profiles were detected by spectrophotometry (A215). TEM micrographs show biotinylated SVPs detected with a gold-labeled goat anti-biotin antibody (1: 20 in PBS). Scale bar = 50 nm.

Addressability of carboxyl groups III.4.4.2Next, it was attempted to label exposed carboxyl groups with biotin, first by priming with EDC to produce the active intermediate O-acylisourea ester. The amine group of biotin hydrazide can then attack this intermediate to form an amide bond (figure III.36). Biotin conjugation was confirmed by Western blot, SEC and TEM as described above (figure III.35).

Figure III.36 Labeling carboxyl groups on the surface of IBD-SVP. Reaction with EDC (1-[3-(dimethylamino)propyl]-3ethylcarbodiimide methiodide) produces the intermediate O-acylisourea which reacts with the amine group of biotin hydrazide. Schematic IBD-SVP refers to Protein Data Bank reference 2DF7 (Lee et al., 2006a). This result demonstrated that IBD-SVPcyto can be functionalized on exposed carboxyl as well as amine groups by decorating the particles with biotin.

Page 99: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

93

IV Discussion Infectious bursal disease virus (IBDV) is a highly contagious agent that causes acute immunosuppressive disease in young chicken flocks. It specifically attacks and replicates in developing B-lymphocytes, cause apoptosis (Rodriguez-Lecompte et al., 2005) and due to lack of potent antibody response leads to vaccination failure and immunosuppression to other infectious diseases. The primary method for controlling the virus is vaccination. Currently flocks are vaccinated with conventional inactivated or attenuated live virus. The most common method strategy for vaccination in breeder flocks is priming vaccination using attenuated live vaccine at 2-4 weeks of age followed by a boost with oil-emulsified inactivated virus at 10-18 weeks of age. This boost induces high titers of antibody, which not only lead to a long-lasting immunity for the recipients but also transfers to the offspring as maternal antibody and gives protection to the newborn chickens in the early stage of their life when their immune system is still immature (Wyeth et al., 1992; Eteradossi and Saif, 2008). In contrast, broilers may be vaccinated once to induce active immune response against IBDV. However the correct time of vaccination is critical to avoid the inactivation of the vaccine by maternal antibodies (Tsukamoto et al., 1995). In spite of vigorous vaccination strategies, the available vaccines are not fully protective against emerging very virulent IBDV (vvIBDV) strains, which is of constant concern for the poultry industry worldwide. Although inactivated vaccines have been produced against vvIBDV, the production requires infection and sacrificing a large number of birds to obtain the desired amount of antigen. Adaptation of vvIBDV to cell culture-based systems requires extensive passages in embryonated chicken eggs before the resulting attenuated virus that can be propagated in cell culture (van Loon et al., 2002). Site-direct mutagenesis that has been introduced to adapt the virus had led to significant drop in virus replication (Lim et al., 1999; Eteradossi and Saif, 2008). But the use of inactivated or attenuated vaccine is accompanied with the risk of remaining the pathogenicity and reversion to virulence. Consequently, the development of new vaccination strategies with improved safety and effectiveness as well as low price is a market demand (Chen et al., 2012). Recombinant vaccines have been receiving great interest since the last decades. Recombinant vaccines are composed of the immunogenic part of the virus, which upon administration to the host induce neutralizing antibodies against the target virus and provide protection. In case of IBDV, the VP2 coat protein is the major host-protective immunogen and contains the conformational epitope which is responsible for induction of neutralizing antibodies. In addition, IBD-VP2 proteins can aggregate to more immunogenic subviral particles (SVPs). Many efforts have been made in producing IBD-VP2 as a subunit vaccine in different expression systems. It has been expressed in E. coli (Chen et al., 2005), Saccharomyces cerevisiae (Garriga et al., 2006; Dey et al., 2009), and Sf9 and H5 insect cell lines (Dybing and jackwood, 1997; Wang et al., 2000; Caston et al., 2001; Lin et al., 2007; Luque et al., 2007) resulting in SVP assembly. Chimeric SVPs have been also used to display VP2 epitope and mimotopes to induce protective immunity against IBDV (Chen et al., 2012; Wang et al., 2012). Also several viral expression systems such as fowlpox virus (Heine and Boyle, 1993; Tsukamoto et al., 2000), Marek’s disease virus (Tsukamoto et al., 1999), Semliki forest virus (Phenix et al., 2001) and Newcastle disease virus (Huang et al., 2004) have been recruited for expression of IBD-VP2

Page 100: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

94

leading to induction of protective immune response. However many of those expression systems have not yet found practical usage mainly due to low productivity, high production costs, low immunity and the potential risk such as integration to the host genome, viral reversion, recombination and reassortment to virulent phenotype in case of viral expression systems (Phenix et al., 2001). Plants have been introduced as a reliable alternative for expression of subunit vaccines (Tiwari et al., 2009). They are the main part of human and animal diet, which makes them more advantageous for mucosal vaccine approaches. The plant-produced IBD-VP2 conferred protection to the immunized chickens although protein expression levels of up to 4.8% of TSP were achieved in Arabidopsis thaliana as reported by Wu et al. (2004). In their study, dried, powdered leaves were resuspended in water for oral immunization. One-week old chickens received five oral doses (~5 µg IBD-VP2) at 3 days intervals. The protection rate was 80% after challenge with an IBDV virulent strain. The IBD-VP2 protein was also expressed in transgenic rice seeds up to 4.5% of TSP, which corresponds to 40 µg per seed (Wu et al., 2007). Anti-VP2 of the sera was increased in orally immunized 2 week old chickens that received four times 5 g (~10 mg IBD-VP2) seed weekly and led to 83% protection after challenge with an IBDV virulent strain. Whereas admiration 3 g (~3.3 mg IBD-VP2) and 1 g (~2 mg IBD-VP2) seed led to only 33% and 16% protection, respectively. The experiment showed that the IgG and protection levels correlate positively with the dosage. The protection efficiency also revealed that the higher dosage administrated, the more protection obtained. IBV-SVPs are known as highly immunogenic structure compared to monomeric and other aggregation forms of IBD-VP2 like tubular form. It was demonstrated that 3 µg of IBD-SVPs is adequate to induce fully protective immunity when administered subcutaneously (Martinez-Torrecuadrada et al., 2003). The necessity of applying ~10 mg rice-produced IBD-VP2 to confer protection raising the question that whether or not plant-produced IBD-VP2 assemble into SVPs. IBD-SVP assembly has not been reported in plant expression systems. In this study, the accumulation levels of tobacco-produced IBD-VP2 were studied upon various protein compartmentalization. The produced VP2 was characterized concerning self-assembly and also glycosylation.

IV.1 Characterization of tobacco-produced IBD-VP2 IBD-VP2 (495 aa) produced in Arabidopsis thaliana (Wu et al., 2004) under the control of octopine synthetase promoter and translational leader sequence had apparent 45 kDa mass on SDS-PAGE. Rice-producd IBD-VP2 (510 aa) under the control of rice endosperm-specific glutelin promoter (Gt 1) promoter and nopaline synthase terminator showed apparent 50 kDa mass. In this study, the corresponding sequence of mature IBD-VP2 protein (441 aa) was cloned into pTRA vector downstream of Cauliflower mosaic virus (CaMV) 35S constitutive promoter and chalcone synthase 5’ untranslated region with and without proper targeting signal to facilitate apoplastic, cytosolic, plastid and membrane anchored accumulation (figure III.1). The protein was transiently expressed in tobacco (Nicotiana tabacum L. cv. Petite Havana SR1) using agro-infiltration. In immunoblot, using non-reducing condition, a distinct band in size of ~40 kDa was detected in case of cytosolic and chloroplast accumulation. No band was detected by membrane integration strategy whereas a fuzzy/diffused band pattern which is sharper in size of ~47 kDa

Page 101: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

95

was observed in case of apoplast accumulation (figure III.2). Apparently the fussy pattern is due to different degree of glycosylation, which resulted in the variation of molecular mass of the protein and poor electrophoretic mobility of the sugar containing molecule. This result is in accordance to the fact that there are three potential N-glycosylation sites predicted by using NetNGlyc 1.0 server (figure III.6). Glycosylation of the accumulated protein in apoplast was also confirmed by immunoblot using rabbit anti-xylose antibody (III.7) as well as deglycosylation analysis performed on purified protein (figure III.8). The type and proportion of the glycan branches were studied in more detail using LC-MS (figure III.9). For N-glycosylation, the asparagine (Asn) residue should be in tandem tripeptide order Asn-X-Ser/Thr which X could be any amino acid except proline. However, it was reported that only 66% of the potential glycosylation sites on secreted proteins are actually glycosylated (Apweiler et al., 1999; Petrescu et al., 2004). Following trypsin and chemotrypsin digestion of tobacco-produced VP2apo, analysis of the respective peptides bearing potential N-glycosylation site confirmed all three sites were fully N-glycosylated. The N-glycans attached to these peptides exhibited plant-typical N-glycosylation profiles (table III.1, figure III.9), comprising of plant-specific complex-type N-glycans (containing α (1, 3)-linked fucose and β (1, 2)-linked xylose) mainly with GnGnXF and GnGnF with frequency of 55.7% of the total VP2 glycosylation profile. The prediction of final glycosylation profile for a particular protein is not yet possible. The glycosylation pathway and the final destination/accumulation of the heterologous protein as well as its intrinsic characters determine the glycan profiles (Bosch et al., 2013). The amino acids in and around of the glycan attachment site, the folding pattern of he protein that determines the accessibility of the site and the availability of polysaccharide precursors, all may influence the efficiency and the profile of the glycosylation. This fact may explain the variation of glycan profile in three N-glycosylation sites of VP2. The hydrophobic nature of IBD-VP2 (figure I.4) is its common characteristic with membrane proteins and the explanation of its aggregation under denaturing condition (figure III.2). Under denaturing condition, the protein must be heated at high temperatures, which might introduce artifacts to dissolution of existing aggregates or formation of new aggregates, as in the case of membrane proteins (Thomas and McNamee, 1990). In addition, this characteristic affects its movement pattern on SDS-PAA gel any may explain the variation between calculated mass (~ 47 kDa) and the mass that protein marker shows (~40 kDa). Wu et al. (2004) also reported smeary movement of VP2 on SDS-PAGE. Similar observation was previously reported by Jagadish et al., 1998. It was suggested that hydrophobic feature of VP2 is responsible for conformation dependent distortion on SDS-PAGE (Wu et al., 2004). In this study, immunoblotting was performed under non-reducing condition which facilitates the detection of a distinct band of IBD-VP2. The protein expression levels were compared by an ELISA-based assay. The highest protein levels were observed by apoplast (~260 µg/g of FLW) and cytosolic (~160 µg/g of FLW) accumulation, respectively (figure III.3). Therefore, these to accumulation strategies were chosen for further studies. Stable transformation of tobacco (Nicotiana tabacum L. cv. Petite Havana SR1) resulted in lower accumulation levels compared to transient expression as expected. The

Page 102: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

96

accumulation levels were 20-30 µg/g of FLW for apoplast and 10-20 µg/g of FLW for cytosolic accumulation. The VP2 accumulation levels in rice seeds reached to 2 mg/g of seed (Wu et al., 2007). Seed specific promoter (Gt1) that enables high VP2 expression, low protease activity and low water content in rice seeds explains such a high accumulation level. In previous studies, it was shown that 3 µg of fully-assembled VP2 produced in baculovirus expression system and Sf9 cells elicits protective immunity and confers comparable protection to that induced by inactivated vaccine when administered subcutaneously (Martinez-Torrecuadrada et al., 2003). The expression levels in tobacco plant are comparable to what was observed in Arabidopsis thaliana (Wu et al., 2004). But the format of the antigen and the assembly level needed to be studied in plant systems. The purification of IBD-VP2 was achieved by IMAC. It was previously shown that assembled IBD-SVPs produced by recombinant expression of rVP2H (fused to C-terminal his-tag) in baculovirus expression system were able to be purified by IMAC (Wang et al., 2000). By solving the 3D structure of IBD-SVP, it was revealed that His-tags fused to the C or N terminus of VP2 are located on the interior face of fully assembled particles and is not accessible to the solvent (Lee et al. 2006b, Garriga et al. 2006). Nonetheless, it was possible to purify intact IBD-SVPs by IMAC. Lee et al. (2006b) had speculated that the affinity of IBD-SVPs to Ni-NTA as due to the negative charge present on their outer surface. Ultimately Doong et al. (2007) proved that this affinity is due to two histidine residues (His249, His253) on the surface of SVPs. Sequence alignment between the IBD-VP2 sequence used by Doong et al. (2007) and the counterpart sequence used in this dissertation showed that the two mentioned His residues were replaced by Glu residues (Appendix VII.4); therefore these residues are not able to participate in the affinity to Ni-NTA. Therefore the observed affinity of tobacco-produced VP2 to the IMAC column is a hint for a deficiency in assembly. The assembly level analysis using SEC confirmed this speculation (figure III.10). The purified tobacco-produced IBD-VP2 showed high amounts of trimers and a significant portion of unassembled monomers. The separation of VP2-producing tobacco extract using ultracentrifugation (figure III.11) followed by TEM studies as well as TEM studies using VP2-producing tobacco leaf sections (data not shown) indicated that the tobacco system is not a method of choice for in vivo production of IBD-SVPs. That may contribute to the intracellular conditions of tobacco such as pH, ion strength and availability of chaperonin. Although, according to thermodynamic modeling, self-assembly of IBD-VP2 and formation of SVP is highly favorable, the SVP formation depends on VP2 concentration and is proportional to the protein synthesis (Hu and Bentley, 2000). Therefore, low accumulation of IBD-VP2 in tobacco cells obviously hampers trimer assembly and SVP formation. A similar result was reported when the human papillomavirus L1 protein was produced in Nicotiana benthamiana even though a TMV-based viral vector was used for expression (Varsani et al., 2006). In this case the assembly level did not achieve higher than pentamers, the structural unit of HPV (Biemelt et al., 2003; Varsani et al., 2003). Formation of self-assembled proteins to VLPs and SVPs is not always favored. The ionic strength, pH and the relationship of the structural units, are the cooperative factors in interactions and free energies of association between the proteins (Hu and Bentley, 2000).

Page 103: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

97

Salunke et al. (1986) showed that disassembled polyomavirus pentamers could only reassemble at high ionic strength. Similarly, Budgerigar fledgling disease virus capsid protein (VP1) forms pentamers in vitro but for further assembly to VLPs, addition of Ca2+ and the removal of chelating and reducing agent are essential (Rodgers et al., 1994). Addition of Ca2+and increasing the ionic strength was not helpful to induce proper self-assembly of the purified tobacco-produced VP2 into SVPs and led to protein aggregation and precipitation (III.1.6). Addition of detergent to disperse unfavorable aggregation followed by stepwise removal of detergent in companion with Ca2+ increasing to facilitate the VP2 assembly also led to non-proper subunit interactions and protein precipitation. Addition of urea to denature the protein and facilitate the proper refolding and assembly in vitro was also not successful. Trimeric capsomers are the main structural unit for IBDV, IBD-VLP and IBD-SVP assembly (Böttcher et al., 1997; Pous et al., 2005; Garriga et al., 2006; Luque et al., 2007). On the other hand, urea alters water structure and dynamics. The protein in the solution unfolds rapidly under 8 M urea conditions (Bennion and Daggett, 2003). As shown in figure III.12, urea destroys the trimers, which are structural units and due to rapid protein unfolding breaks them into small fractions (~ 5 kDa and ~ 1 kDa) without any influence on the monomers (figure III.12). It was demonstrated that some virus-like protein subunits such as Human papillomavirus-like particles could be renatured from denatured forms in vitro into virus-like particles without the presence of chaperons (Zhang et al., 1997). In case of IBD-VP2, it seems that for proper folding and subsequent assembly to SVPs, the activity of chaperons is essential. It was also reported that denatured and renatured IBD-VP2 is also unable to induce neutralizing antibodies in chickens (Öpplin et al., 1991) which manifests that in vitro folding occurs in non-proper form. Immunogenicity of tobacco-produced VP2 was investigated in mice and led to high serum titer IgG induction observable after the first boost (III.1.7; figure III.14). The generated mAbs (III.1.7; data not shown) against the tobacco-derived mixture of VP2 monomer and trimer mixture showed high reactivity against tobacco-produced recombinant VP2, but no detectable reactivity against Pichia produced IBD-SVPs (data not shown). This fact showed that the selected clones screened against epitopes that are hidden (cryptotope) when the protein is assembled into the subviral particle.

IV.2 Characterization of Pichia-produced IBD-VP2 Generally, low accumulation levels are the major drawback for plant-produced vaccines especially when the vaccine should be applied orally since oral immune system stimulation usually needs higher dosages of the antigen compared to parenteral immunization methods. Moreover, tobacco leaves contain toxic alkaloids such as nicotine, which restricts the application of the produced protein without extensive purification. On the other hand, the multi-immunogenic form of VP2, IBD-SVP could not be generated in tobacco. Expression of VP2 in E. coli expression system has led to contradictory results. Jagadish et al. (1998) reported that VP2 produced in E. coli is not protective. However, Rong et al., (2005) showed that administration of E. coli producing VP2 is immunogenic and protective.

Page 104: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

98

Alternatively, methylotrophic yeast Pichia pastoris is an important organism for recombinant protein production at large scales owing to its high expression levels, low production costs and high cell density growth in bioreactors (Jiang et al., 2011). It was demonstrated that the protein production yield in Pichia could achieve up to 14.8 g/l (Werten et al., 1999). Pichia has also been widely used for production of therapeutic proteins. The main concern about the Pichia expression system is the N-linked glycosylation of the high-mannose type. However, the length of the oligosaccharide chains added post-translationally to proteins in Pichia (average 8-14 mannose residues per side chain) is much shorter than those in Saccharomyces cerevisiae (50-150 mannose residues) (Grinna and Tschopp, 1989; Tschopp et al., 1987b) and the protein expression level is 10-100 fold higher. Interestingly, P. pastoris in contrast to S. cerevisiae is not capable of adding the immunogenic α (1, 3)-terminal mannose to oligosaccharides. Very little O-linked glycosylation has been observed in Pichia. However fungal-like glycans such as β-mannose linkages, which exist in produced glycoprotein may stimulate an immune response or binding to mannose receptors and consequently reduce the efficiency of the produced proteins such as enzymes or antigen vaccines (Hopkins et al., 2011). To avoid such an effect, either the glycan sites should be genetically eliminated by introducing point mutations or the enzymes involved in transfer of immunogenic glycans residues during glycosylation pathway, should be eliminated or suppressed. Another alternative is the production of the non-glycosylated form of the target protein by cytosolic expression, if the protein folding is not being damaged. In this study the aglycosylated yVP2 produced by cytosolic expression used as a vaccine in immunization of chickens (III.3). The expression level of IBD-VP2 in P. pasroris reached up to 76 mg/l culture after 4 days of induction (figure III.17). The standardized purification procedure enabled the recovery of about 50% of the produced VP2 at ~96.2% purity (figure III.21). SEC and TEM, respectively, showed that in contrast to the tobacco expression system, VP2 monomers assembled into higher assembled symmetrical SVPs in yeast (figure III.20). However, a smaller part of the VP2 fraction was not fully-assembled. While the VP2 content from the secreted version (VP2sec) accumulated in the medium was not present in the assembled form, the cell extract from the same culture apparently contained the SVPs, which was confirmed by SEC and TEM (figure III.20). Such SVPs had different configuration compared to SVPs present in VP2cyto producing yeast due to the glycosylation. This observation showed that only less assembled forms of VP2 and monomers pass through the cell wall pores and reach to the medium while fully assembled particles, due to their size, entrapped in extracellular space between the cell membrane and cell wall.

IV.3 Vaccine preparation Vaccination is an efficient and most cost-effective method for preventing infectious disease. Homogeneity to ensure the consistent dosing of subjects; efficacy to achieve protective immunity; stability to reduce the costs of storage and shipping and potency (or concentrated material) to provide easy delivery of desired dose are main concerns in any vaccination platform development. Recombinant vaccines have attracted the attention especially in last two decades due to their benefits and less concerns. However, so far, most developed vaccines of these types

Page 105: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

99

are delivered by injection (parenteral), which makes mass immunization costly, laborious and less safe especially in poor-developed regions of the world. In contrast, oral delivery of vaccine is safe, easy to apply and reduces the immunization costs. Other advantage of an oral vaccine is the possibility of mucosal immune response induction, in particular, secretory IgA (SIgA). SIgA inhibits pathogen from colonization in the gut and consequently disease. Although oral administration of vaccines is beneficial, in practice, it has been faced several obstacles such as harsh conditions exist in the digestive tract and targeting M cells in the gut, which have limited its success. A successful oral vaccine delivery comprises of different elements and stages; (1) the proper immunogen (vaccine) (2) it must enter via the oral cavity and passes through the gastrointestinal tract and reaches to mucosal inductive sites in the intestine, (3) it must be taken up by M cells to reach to gut-associated lymphoid tissue (GALT) and (4) it must induce protective immune response against the target pathogen. The neutralizing antibody response is required for protection against IBDV. VP2 has been identified as the major host protective immunogen against IBDV that carries neutralizing epitopes (Jianrong et al., 2004). The ability of VP2 to confer protection is dependent on its conformation and structure. VP2 produced in E. coli was unable to induce neutralizing antibodies (Jagadish et al., 1998). Although later Rong et al. (2005) showed that live E. coli producing VP2 could confer 90-100% protection. VP2-based DNA vaccines can prevent the clinical outbreaks and mortality but can not prevent bursal damage. That may be due to the inherent low immunogenicity of DNA vaccines or improper folding (Jianrong et al., 2004). In contrast, it was also shown that VP2 as a subunit vaccine is resistant to the proteolytic and acidic conditions of the gastrointestinal tract, is taken by M cells, and has the ability to induce a protective immune response in chickens (Li et al., 2004). VLPs and SVPs are multi-immunogenic forms of the antigen that have less available target sites for proteolytic enzymes compared to VP2 monomers and low levels of assembly or tubular aggregation (Martinez-Torrecuadrada et al., 2003). Therefore full-assembly of VP2 into either VLP or SVP improves the vaccine efficacy and decreases the required dosage. Protection of vaccines against the acidic and proteolytic conditions of the gastrointestinal tract has been achieved by co-administration of sodium bicarbonate as acid neutralizing agents in chicken (McCluskie et al., 2000; Hoshi et al., 1995). It was also shown that oral administration of Virginiamycin (antibiotic) and Cysteamine (Yang et al., 2007) had a positive effect on the stimulation of the mucosal immune system in chicken. Using a system consisting of non-ionic surfactant vesicles in companion with bile salts (bilosomes) to protect the vaccine against gastrointestinal digestion was also accomplished (Mann et al., 2009). In this study, the generation of IBD-SVPs via recombinant expression of IBD-VP2 was aimed. Expression of VP2 in tobacco did not lead to generation of SVP whereas expression in Pichia resulted in the production of SVP structures at high protein levels (76 mg/l). Cytosolic expression of VP2 not only prevented the glycosylation, which could induce a non-favorable immune response but also capsulated the produced SVPs inside the Pichia cells. These cells, after

Page 106: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

100

successful heat inactivation without any damage to the vaccine (figure III.25), were used as oral vaccine. The idea was that cell capsulation should protect the vaccine against the acidic and proteolytic conditions of the gut and consequently targets the vaccine more efficiently to the inductive sites. To standardize and concentrate the cell-capsulated vaccine, a simple food-processing technique was used in this study. Freeze-drying is a simple and inexpensive method that facilitates antigen stability at ambient temperatures for a long time without any loss of antigenic protein. In addition, freeze-dried materials can be formulated in combination with food and adjuvants. Besides, purification of the vaccine is carried out with a simple, fast and cost-effective method and yielded to highly pure protein suitable for immunization approach. It was shown that induction of immune tolerance in chickens in contrast to mammals is difficult to generate even with high doses of the antigen due to two main reasons: first, innocuous food antigens are ignored by the immune system. Second, the soluble antigens are immunogen rather than tolerogen (Klipper et al., 2000). Oral tolerance was induced by using encapsulated powdered antigens in chickens but not with the same antigen dissolved in water (Friedman et al., 1998). To address this issue, cell-capsulated or purified IBD-SVPcyto has been used in this thesis. However, the highest considered dosage (4 mg of antigen) was relatively high but there was no concern for induction of immune tolerance since previously 10 mg of IBD-VP2 produced in rice seeds was successfully tested as an oral vaccine (Wu et al., 2007). In separate previous studies, the positive affect of CpG ODN (Mallick et al., 2011; Zhang et al., 2008; McCluskie et al., 2001) and NaF (Hoshi et al., 1999) or their mixture (Zhang et al., 2007) as oral adjuvants for chicken immunization has been shown. For the experiments in this thesis the mixture of CpG ODN and NaF has been added to the antigen as described before (Zhang et al., 2007).

IV.4 Induction of immune response against IBDV by applying Pichia-produced IBD-SVPcyto administered either orally or parenterally

Attenuated vaccines are usually efficient in inducing long-lasting protective immune response including B and T cell responses upon just one-shot vaccination. But they carry the potential risk of reversion to virulence and transmission of the attenuated virus to susceptible individuals with immature immune system such as newborns. To provide more safety, inactivated and subunit vaccines have been utilized. But safety comes at the cost. Inactivated virus and subunit vaccines are less immunogenic than attenuated virus-based vaccines. To induce protective immunity, usually multiple boosts and adjuvants are required. Viruses are highly immunogenic due to their ability of proliferation inside the cells that leads to induction of cellular immune response and their ordered-repetitive antigen presentation. In case of IBDV, only mild and intermediate strains are used as attenuated virus vaccines (Carballeda et al., 2011). More pathogenic strains may cause bursal atrophy and thus immunosuppression as possible consequences (Müller et al., 2012). Due to the emergence of very viral IBDV strains, conventional vaccine does not confer full protection. Therefore the development of new protective vaccines is required. Another problem with applying full virus as a vaccine both in inactivated and attenuated forms is interfering of these sort of vaccine in differentiating infected from vaccinated animal (DIVA). Infection with the virus like vaccination induces humoral immune response that prevents the

Page 107: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

101

serological discrimination between infected and healthy vaccinated animals, hampering controlled eradication of the disease. Whereas recombinant VP2 vaccine makes DIVA feasible through the detection of antibody responses against other viral proteins such as VP3 that only exist in the full virus but not in the vaccine. On the other hand, subunit vaccines are safe because they are free of pathogen genetic materials compared to the risk of incomplete viral inactivation and reversion of virulence when using full virus vaccines. Also, recombinant vaccines eliminate the need to infect and sacrifice animals to propagate and extract adequate amount of virus for full virus vaccine production. As an alternative to cell culture that need sequential passage of the virus through the culture in order to attenuate the virus, subunit vaccine production is less laborious and more economical. Moreover subunit vaccines are the only option for the viral strains that can not be adapted to the cell culture. It was shown previously that Pichia cells are safe for chickens (Pitkovski et al., 2003). Hundred microgram of partially purified rVP2 produced in Pichia was administered intramuscularly to the chickens without showing any harm. The large-scalability of Pichia production has made it the most economical eukaryotic system for recombinant protein production. Induction of neutralizing antibodies is required to confer protection against IBDV. Therefore, VP2 carrying the neutralizing epitopes was used as antigen in this study. The neutralizing epitopes are conformational (Rong et al., 2005). Therefore, to maintain the antigen structure the full-length of mature IBD-VP2 was used for recombinant production of vaccine. Furthermore, IBD-VP2 subunits are capable of self-assembling to subviral particles (SVPs). VLPs and SVPs are genome-free viral particles that mimic the virus in stimulation of host immune response. Due to genome depletion, they are unable to induce innate immunity via pattern-recognition receptors (PRRs) such as Toll-like receptors (TLRs) and RIG-I-like receptors (RLRs). TLR-3, TLR-7, TLR-8 and TLR-9 are endosomal membrane proteins that recognize dsRNA, ssRNA, G-rich oligonucleotide and unmethylated CpG DNA, respectively (Kawai and Akira, 2008). In this study CpG ODN was added to the antigen to compensate the lack of unmethylated CpG DNA but its effect could not be statistically analyzed due to high variation in immune responses and low number of the chickens in each immunization group. But chickens of group 8 that received high doses of the purified IBD-SVPcyto in combination with adjuvant containing NaF and CpG ODN showed better immunity against IBDV; BF/BW ratios comparable with the oral application of attenuated virus and parenteral vaccines, full protection and moderate bursal damage (table III.5). Achieving 60% viral clearance indicates that the antigen with adjuvant mixture was able to induce CTL response. VLPs and SVPs are able to induce an innate immune response due to not clearly known procedure but perhaps by contaminating proteins from their expression system (Demi et al., 2005). Also their symmetrical structure and repetitive epitope presentation may serve to induce cellular as well as humoral immune response even in the lack of adjuvants (Bachmann et al., 1993). They are also ideal as a mucosal vaccine because their particulate nature and size facilitates better uptake by M cells and dendritic cells.

Page 108: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

102

Ten to fourteen day old birds are the most suitable source for starting the specific pathogen free (SPF) chicken immunization (Rong et al., 2007). The immune system of newborn chickens and their lymphoid organs develop slowly (Beirao et al., 2012). It has been shown that the proportion of immune cell subsets differ dramatically by age. This proportion correlates with susceptibility and resistance of chickens to pathogens (Beirao et al., 2012, Cheng et al., 2001a). For instance, the T helper cell population is significantly increasing in two phases; since the day of birth until the 5th week and since then to the 12th week of age (Beirao et al., 2012). It was previously shown that the pathogenesis of IBDV may differ depending on the age of the infected chickens due to unknown reasons. Chickens between 3-6 weeks after hatching are most susceptible for clinical signs, while younger chickens are devoid of clinical signs but severe immunosuppression (Rautenschlein et al., 2007). Pathogenesis variation of a specific pathogen such as IBDV in different ages can be explained by the maturity stage of immune system. These facts may explain why the antibody response in the chickens remains irresponsive during the early immunizations even until virus encounter (figure III.29). Previous studies and field observations has indicated that genetic variation between individuals, affect the induction of innate and acquired immune response when encountered to IBDV (Aricibasi et al., 2010). Therefore, it was suggested to analyze individuals rather than pooled samples from the immunized group (Douce et al., 1999). Accordingly, all the detailed observations for each chicken were listed into the table in appendix VII.6 and the result was also studied individually as well as statistically. To make the data more valuable, a large number of chickens in each experimental group have to be investigated to provide enough data for statistical evaluations. As mentioned before, neutralizing antibodies are essential to promote protection against IBDV. Therefore VP2 carrying the neutralizing epitopes was used as antigen in this study and the antibody response was investigated in the immunized and challenged chickens. In addition, the efficacy of the vaccine was evaluated by studying the potency of cytotoxic T cells to remove the virus. The contribution of humoral immunity in conferring the protection has been proved as demonstrated by transferring the maternal antibody. Evidence in finding the role of cell-mediated immunity is in progress (Eteradossi and Saif, 2008). There is a positive correlation between being IgY seropositive and survival (table III.4). This means that the chickens that had developed IgY immune response, survived after the viral challenge. However this correlation is not always present. For instance, in groups 4, 5 and 7 there are survivors that were not IgY seropositive but IgM seropositive (table III.4). Perfect compatibility between being seropositive in the IDEXX kit assay and survivance reveals that in those individuals IgM instead of IgY is responsible to develop protective immune response. Study the IgM response for some individuals in groups 1 that immunized with 250 mg Pichia yVP2cyto, group 4 that immunized with 25 mg Pichia yVP2cyto with adjuvant mixture, group 5 that immunized with 50 µg of purified IBD-SVPcyto, group 6 that immunized with 50 µg of purified IBD-SVPcyto with oral adjuvant mixture as well as mock-vaccinated group (group 9)

Page 109: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

103

showed that in spite of induction of IgM immune response, without IgY response, the chickens were unable to tolerate the viral challenge. This observation can be explained by the fact that IgM is the primary induced serum antibody in response to the pathogen. The immune system of these individuals is unprepared and those IgM molecules are immature to combat the virus following viral challenge. Consequently they start to produce IgM lately in response to the attacking virus when the disease has developed into a uncontrollable stage. Among the individuals in groups 7 and 8 that were immunized with 500 µg of purified IBD-SVPcyto with and without adjuvant, respectively, as well as mock-immunized chickens in group 9 no IgM response was detected while a protective IgY response was detectable (data not shown). As described above, IgM is the primary humoral immune response to the pathogen. But afterwards, the IgM response shifts to more stable antibody isotype, IgY, during a process named isotype switching. Therefore, in such individuals, obviously due to their genetic background, the immune system is well prepared to face with the virus and to prevent the disease. One of the major sites for detection of IgA response are bile samples but the disadvantage of measuring biliary IgA is that IgA measured in the bile may not be effective on other remote sites such as intestine (Mockett and Rose, 1986). This is due to selective localization of mucosal lymphocytes to specific tissue that called “homing”. Homing phenomena explains the intrinsic local mucosal immune response and determines the most efficient route of mucosal administration of vaccines. To provide more precise data, the intestinal fluid (data not shown) and feces samples were collected to study the IgA response in the intestine (figure III.28). Besides, the serum IgA was also analyzed (figure III.28). Although no significant amount of IgA was detected in the intestinal fluid and feces, IgA response is detectable in the serum of all chickens, which received the vaccine orally and even in the parenterally immunized chickens. But as explained in the result section, probably this response is against Pichia proteins orally administered and could not be evaluated as mucosal immune response against VP2. Overall, the IgY and IgM response was detectable in all the orally administrated groups upon vaccination and challenge. This observation indicates that the induction of the systemic immune response through oral feeding of Pichia-produced SVPs either in the purified form or cell-capsulated is possible. However, it seems that purified SVPs (III.2.3) showed an improved immunogenicity compared to cell-capsulated SVPs. The reason is that purified IBD-SVPcyto (III.2.3) are stable enough to pass harsh condition in the gastrointestinal tract and reach the M cells and become absorbed by them while perhaps due to incomplete digestion of the Pichia-cell wall, the chance to release from the cells and absorption is less for cell-capsulated vaccines. The lack of correlation between antibody titer and clinical signs indicates the critical role of cell-mediated immune response in controlling the disease. Previously, it was shown that cell-mediated immunity alone in the absence of antibody can be protective against IBDV (Yeh et al., 2002). Moreover, inherent resistance has been observed in some breeds of chickens (Hassan et al., 2002). The weight increase of the chickens during the experiment is a hint that the vaccines were not interfering in chickens’ growth rate. Monitoring the IBDV clinical signs following viral challenge (table VII.1) indicated that the clinical signs initially appeared in the groups that mock-

Page 110: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

104

immunized (group 9) and challenge control (group 11) as well as those received 50 µg of purified IBD-SVPcyto (group 5 and 6). Then the slight clinical signs appeared in all the orally vaccinated groups including those that received Pichia-capsulated and purified antigen (groups 1-8). In the following days moderate to severe clinical signs observed in challenge control (group 11) and mock-immunized (group 9) groups, the intensity of clinical signs and mortality increased in those groups as well as the groups that received 50 µg of purified IBD-SVPcyto (group 5 and 6). While, parenterally-vaccinated as well as the group that received the live-attenuated virus as a vaccine showed no clinical signs. Within orally vaccinated groups, chickens in the groups 7 and 8 which received 500 µg purified IBD-SVPcyto without and with adjuvant showed less clinical signs and just in slight form whereas chickens that received 4 mg of vaccine in cell-capsulated form were not protected to that level. This result is in agreement to antibody titers (figure III.29 and table III.4) and revealed that the cells need to be disrupted before administration as vaccine in order to improve the efficiency of vaccine presentation and absorption by the M cells. The challenge virus (MB3) is a classic strain and differs from the IBD-SVP vaccine strain (IR01) that belongs to very virulent strains (I.4.1). It was shown that protection against IBDV was highly dependent on the antigenic correlation between the vaccine and the circulating virus strains (Shaw and Davison, 2000). On the other hand, IBD-VP2 amino acid sequences of all known strains described all over the world are more than 93% identical. This homogeneity between Asian and European strains is more than 98%. IR01 is an Asian strain that used to generate the subunit vaccine and MB3 belongs to European strain obtained from recent IBDV outbreak in Germany. The variations between the strains locate in major hydrophilic region where the virus neutralizing epitopes exist (I.4.6; figure I.4). Nevertheless, it was demonstrated that vaccination with classical strains confers partial protection against variant strains whereas vaccination with variant strains confers protection against homologous and heterologous variant and classical IBDV strains (Ismail et al., 1991). Therefore, vaccination with higher pathogenic strains confers protection against less pathogenic challenge strains. However, the dose and strain of both vaccine and challenge virus affects the degree of protection (Jackwood et al., 2001; Ismail et al., 1991). Cross protection has been observed in IBDV vaccines. Since the protective immune mechanism to IBDV is mostly humoral, the serological cross reactivity existing among IBDV isolates is the reason of conferred protection. Therefore, it is expectable to see that chickens vaccinated with IBD-SVPcyto derived from IR01 strain show reduction in viral damage when challenged with the heterologous classical strain, MB3. Therefore, challenging with a heterologous strain may help to evaluate either the cross-protection or in contrast, the specificity of the vaccine. Cross protection is of interest for vaccine development but is not the case all the time. Conventional inactivated influenza vaccine does not provide cross-protection against unmatched strains (Schneider-Ohrum and Ross, 2012). As a result, vaccines should be generated based on seasonal or pandemic strains at each time and region. From the aspect of virus clearance, the injected and live-virus vaccinations were thoroughly successful in virus removal and confer full protection. Groups that received 500 µg of purified IBD-SVPcyto regardless of their partial virus removal confer full protection to their receivers.

Page 111: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

105

Although the rest have been protected from the disease but they may act as virus carrier to other healthy individuals. Generally, in this experiment, orally vaccinated groups (groups 1-8) were unable to remove the virus as efficient as both injected and attenuated virus vaccination. In addition, bursal damage scores as putative damage index of IBDV was used to analyze the efficiency of the vaccines. The data demonstrated that chickens, which received 50 µg of purified IBD-SVPcyto (groups 5 and 6) as well as chickens in mock-immunized and negative control group showed the maximum score. Other orally vaccinated groups (groups 1-4 and 7-8) showed moderate bursal damage. Injected and live-virus vaccinated groups were quite efficacious without any damage to bursa. Another index for evaluation the vaccine efficiency is the ratio of bursa weight in gram to the body weigh in kilogram (Bursa/BW ratio). The obtained data showed that chickens in challenge control, mock-immunized, and those that received 50 µg of purified IBD-SVPcyto in groups 5 and 6 have the minimum values or maximum bursal atrophy, respectively. Statistical categorization puts the group 8 that received 500 µg of IBD-SVPcyto in combination with adjuvant in the same group as attenuated virus vaccinated and intramuscularly vaccinated group. This observation indicated that among orally vaccinated groups only 500 µg purified IBD-SVPs dose in companion with adjuvant worked as efficacious as either attenuated virus or parenteral vaccine to confer protection. Although the chickens received live attenuated vaccine (group 12) showed more bursal atrophy (3.16) comparing to those that received the vaccine parenterally in group 10 (3.72), the difference is not statistically significant. While the resulting data do not show the advantage of oral over parenteral immunization in prevention of IBDV and conferring the protective immunity, there are indications that oral vaccination with 500 µg purified IBD-SVPcyto could potentially provide an alternative to vaccination through injection. The challenge trial manifested definitive evidence that this approach could protect the chickens against IBDV. In addition, oral vaccination provide a number of advantages over injection such as ease of administration, reducing the stress for chickens and cutting the risk of contamination due to needleless vaccination and decreasing the cost of vaccine due to omitting the need to medical facilities and treated personnels. The drawback of oral immunization that is low immunogenicity can be improved by optimization of the format, dose and co-administration with proper adjuvants.

IV.5 IBD-SVPcyto as a potential platform for nanobiotechnology Viral nanoparticles (VNPs) are suitable for a vast range of applications ranging from medical imaging and drug delivery to the design of novel materials and electronic devices. It is important as part of the development process to determine the versatility of new VNPs and their tolerance of different chemical environments, particularly the harsh environments necessary for modification and functionalization. Many bioconjugation reactions must be carried out in the presence of the organic solvent DMSO, whereas ethanol is required for mineralization and electronic devices must be heat resistant. The tolerances are often highly constrained and specific, e.g. mineralization generally requires a narrow pH and temperature range specific for each

Page 112: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

106

mineral (Douglas et al., 1999). Novel SVPs such as the particles based on IBDV described herein must therefore undergo tolerance tests to study their integrity across a range of temperatures, pH values and solvent concentrations, and the accessibility of addressable conjugation targets on the particle surface must also be determined. In this study IBD-SVPcyto with a T= 1 produced in P. pastoris was purified by pH shift and salt precipitation and polished by SEC (III.2.3). The spectrophotometry was found to be the most accurate method to determine particle concentration in solution. Because colorimetric methods cannot distinguish SVPs from stray peptides, whereas spectrophotometry generates separate peaks for peptides without aromatic side chains (190–220 nm) and proteins plus peptides with aromatic side chains (280 nm). The integrity of the particles has been investigated by Native agarose gel electrophoresis (NAGE), Dynamic light scattering (DLS) and Transmission electron microscopy (TEM). DMSO is a versatile solvent for bioconjugation but high concentrations can induce conformational changes in proteins by binding to hydrophobic and aromatic side chains that are normally shielded inside the structure, resulting in precipitation (Arakawa et al., 2007). Similarly, ethanol is required for mineralization reactions but high concentrations disrupt intermolecular hydrogen bonds and cause proteins to denature and precipitate (Falk et al., 2002). It was found that IBD-SVPcyto remained stable in solutions containing up to 20% (v/v) DMSO or ethanol, but that higher concentrations altered the electrophoretic mobility of the particles and caused them to precipitate. DLS showed that particles in 20% (v/v) ethanol and DMSO increased in size (the radius increased by 1.3 and 0.6 nm, respectively; figure III.30). In contrast, 50% (v/v) DMSO either stretched the particle into an elliptical conformation or shrank them to a smaller radius (peak at 10 nm) and faster running in electrophoretic mobility, which induced rapid precipitation. Previous studies have shown that IBDV is moderately stable in an acidic environment (pH 3), and also in the presence of certain disinfectants and when heated to 60°C for 30 min (MacLachlat and Stott, 2005). It was expected the corresponding SVP to have similar properties despite the additional rigidity of the parent virus conferred by RNA-protein and heteromeric protein-protein interactions, because the VP2 protein plays an important role in virion stability. It was found that the IBD-SVPcyto were thermostable at 60ºC for 1 h, retaining 90% of the solubility observed at room temperature. After incubation at 68ºC for 1 h, 70% of the particles remained stable and soluble. VLPs can display extraordinary thermostability, e.g. bacteriophage PP7 VLPs are stable at 90ºC for more than 2 min because of the disulfide bonds linking coat protein dimers (Caldeira and Peabody, 2007). IBD-SVPcyto do not form disulfide bonds but strong interactions between VP2 monomers help to preserve the integrity of the particles at temperatures between 60 and 70ºC, making their heat tolerance comparable to CPMV (Strable and Finn, 2009). The IBD-SVPcyto was also stable across a wide pH range. As previously reported (Reiss et al., 2004), TEM analysis revealed minor conformational (more condensed in pH 9.0) changes in alkaline solutions but this did not affect the hydrodynamic radius of the particles, and the slight change in electrophoretic mobility is likely to reflect the differing net charge of the particles in different pH environments. The IBD-SVPcyto therefore appears comparable to acidophilic viruses such as Sulfolobus islandicus rod shaped virus 2 (isolated from pH 2.5 acidic springs in Iceland) (Steinmetz et al., 2008).

Page 113: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

107

The ability to modify specific sites on the surface of VNPs allows them to be used as scaffolds that can be functionalized with diverse reagents including peptides, nucleic acids, metal ions, organic molecules and florescent dyes. Amine groups on lysine side chains are often used as attachment sites, e.g. for polymers (Comellas-Aragones et al., 2009), fluorescent dyes (Steinmetz et al., 2010; Barnhill et al., 2007b) and redox-active complexes (Steinmetz et al., 2006a). There are five accessible lysine side chains per VP2 monomer, and with each SVP comprising 60 VP2 subunits this equates to 300 accessible amine groups per particle. Carboxyl groups on the side chains of acidic residues can also be used as attachment sides following activation, if the conjugation target contains a primary amine group (Barnhill et al., 2007b; Steinmetz et al., 2006b; Schlick et al., 2005). Each VP2 monomer has four Glu residues and nine Asp residues with an accessible carboxyl group, making 13 accessible sites per monomer and potentially 780 per SVP. TEM analysis confirmed successful biotinylation; several antibodies were bound per IBD-SVP (figure III.35), indicating multivalent display of biotin moieties. The immuno-staining and TEM method, however, can not be regarded as a quantitative method; steric hindrance effects prevent large numbers of gold-labeled anti-biotin antibodies binding to the SVP surface (Steinmetz et al., 2008). To gain quantitative data on biotin labeling efficiency, the number of addressable amine sites per SVP was determined by the direct coupling of fluorescent (Alexa fluor 488) N-hydroxysuccinimide (NHS) esters. This showed that each SVP carried an average of 60 dye molecules, indicating that only one amine side chain can be occupied on each monomer, perhaps due to steric hindrance of the four remaining lysine residues once the first dye molecule is installed. A small amount of inter-particle aggregation observed during carboxylate conjugation, probably reflecting the formation of peptide bonds between the activated carboxyl groups on one particle and exposed amine groups on the lysine side chains of other particles (data not shown), but overall the integrity of the particles was maintained with both conjugation methods. It has previously been suggested that the sensitivity of IBD-SVP detection by spectrophotometry (A280) is too low for SEC (Cheng et al., 2001b). It is shown that A215 measurements are more useful for this purpose and show adequate sensitivity.

IV.6 Conclusion and future prospects Although the IBD-VP2 expression level in tobacco is high enough for application as oral vaccine, assembly into SVPs did not occur in the plant system. As an alternative, Pichia pastoris has many of the advantages of higher eukaryotic expression systems such as protein processing, protein folding, and posttranslational modification, while being as easy to manipulate as E. coli or Saccharomyces cerevisiae. It is faster, easier, and less expensive to use than other eukaryotic expression systems and generally gives higher expression levels (10- to 100-fold higher heterologous protein expression levels compared to S. cerevisiae). In addition, lower degree of immunogenic N-glycosylation compared to other yeast expression systems and very little O-linked glycosylation make Pichia very useful as a protein expression system especially for vaccine production. The high production levels of IBD-VP2 in Pichia cells as well as non-toxicity of the expression system and economical optimized protein purification make Pichia as profitable IBDV subunit vaccine production. The generated vaccine either in the form of cell-

Page 114: Expression and Characterization of Infectious Bursal

Chapter IV Discussion

108

capsulated or purified IBD-SVPcyto enhanced the IBDV-specific antibody response and provides protection against a heterologous classical IBDV strain. Further research is needed to optimize the vaccine dose in combination with a proper adjuvant for use against very virulent strains such as the homologous strain IR01. The Pichia-produced IBD-SVP were also shown to be stable in 20% (v/v) solutions of DMSO or ethanol and also resistant to heat and pH extremes. This means IBD-SVPcyto can be used under the harsh environmental conditions required for bioconjugation, mineralization and other modifications. It was confirmed that IBD-SVPcyto could be functionalized on exposed amine and carboxyl groups by decorating the particles with biotin. The combination of stability, accessibility and amenability for large-scale production in yeast makes IBD-SVPcyto an ideal platform for diverse applications in nanobiotechnology. The multivalent display of fluorescent molecules at discrete sites on the VNP surface provides a high density of such traceable molecules without fluorescent quenching, increasing the sensitivity of detection. Combined with the possibility of dual labeling, IBD-SVPcyto could also be developed as a multifunctional platform, e.g. carrying both targeting ligands and fluorescent tracers for bioimaging applications (Brunel et al., 2010; Lockney et al., 2011). IBD-SVPcyto could therefore be developed as a novel platform for medical imaging and/or targeted drug delivery. Although no in vivo studies have yet been performed, on the basis of the presented in vitro results it is speculated that IBD-SVPcyto could be recruited as a nanobiotechnology platform.

Page 115: Expression and Characterization of Infectious Bursal

Chapter V Summary

109

V Summary Infectious bursal disease virus is a causative agent of infectious bursal disease in young chickens causing significant losses to the poultry industry worldwide. In this study the gene encoding the viral capsid protein (VP2, IR01 strain) with self-assembly capability was cloned into a plant expression pTRAkc vector and expressed in tobacco. Recombinant protein levels in tobacco leaves reached to 260 µg/g and 160 µg/g fresh leaf weights (FLW) in apoplastic and cytosolic protein accumulation, respectively. These numbers were 20-30 µg/g and 10-20 µg/g FLW in stable transformation while the assembly level did not reach higher than trimers. Nonetheless the recombinant VP2 protein was highly immunogenic in mice. Alternatively, VP2 was cloned into the yeast expression vector, pPICZ_B, and produced in Pichia pastoris resulting in the formation of ~23 nm homomeric subviral particles (SVPs) composed of the VP2 protein. The recombinant protein levels reached 76 mg/l culture and the final yield after purification was 38 mg/l. Anti-IBDV antibodies were detected in chickens vaccinated by injection of purified IBD-SVPcyto or by feeding either purified IBD-SVPcyto or recombinant Pichia expressing IBD-VP2cyto. Challenge of vaccinated chickens with the heterologous IBDV strain (MB3) and subsequent analysis of antibody response, mortality, virus clearance, bursal damage score and bursa weight to body weight (BF/BW) ratio demonstrated that 20 µg of purified IBD-SVPcyto intramuscularly administered confers full protection, complete viral clearance and thoroughly prevents bursal damage and atrophy whereas the mortality rate in non-vaccinated and mock-immunized groups was 60% with a high bursal damage score and atrophy. Commercial vaccine also conferred full protection and viral clearance while partial bursal atrophy was inevitable. Orally administered purified 500 µg IBD-SVPcyto with and without adjuvant conferred 100% protection, however, only in companion with adjuvant showed 60% viral clearance and in the absence of the adjuvant showed no viral clearance. In both cases, moderate bursal damage was observed whereas in combination with adjuvant provided BF/BW ratio similar to live-attenuated vaccine and intramuscularly administered 20 µg of purified IBD-SVPcyto. Administration of 250 mg of Pichia-capsulated IBD-SVPcyto with and without adjuvant resulted in 100% and 60% protection, respectively. Moderate bursal damage and moderate atrophy was present in both groups. In the groups that received 25 mg of Pichia-capsulated IBD-SVPcyto and purified 50 µg IBD-SVPcyto, the protection rate was 90-100% and 40-60%, respectively. In the first group moderate bursal lesions and severe atrophy and in the latter groups severe bursal lesions and severe atrophy was observed. Monitoring the weight of chickens as a health index showed no interruption of vaccination in growth rate. This study demonstrates the ability of Pichia-produced IBD-SVPcyto to induce specific immune response against IBDV either in oral or parenteral administration. The ability of orally administered vaccine in induction of protective local as well as systemic immune response is demonstrated. In the second part of this study, the potential application of Pichia-produced IBD-SVPcyto as a novel nano-platform was investigated. The purified particles were able to tolerate organic solvents up to 20% concentration (ethanol or dimethylsulfoxide); they resisted temperatures up to

Page 116: Expression and Characterization of Infectious Bursal

Chapter V Summary

110

65 ˚C and remained stable over a wide pH range (2.5–9.0). Bioconjugation to the amine groups of lysine residues and to the carboxyl groups of aspartic and glutamic acid residues were achieved, allowing the functionalization of IBD-SVPcyto with biotin. The accessibility of surface amine groups was measured using Alexa Fluor 488 N-hydroxysuccinimide (NHS) ester, an amine-selective fluorescent dye, revealing that approximately 60 dye molecules were attached to the surface of each particle. IBD-SVPcyto can therefore be exploited as a robust and versatile nanoscaffold to display diverse functional ligands.

Page 117: Expression and Characterization of Infectious Bursal

Chapter VI Zusammenfassung

111

VI Zusammenfassung Das Infectious bursal disease virus (IBDV) ist der Auslöser der infektiösen Bursitis bei jungen Hühnern. IBDV führt weltweit zu beträchtlichen ökonomischen Schäden in der Geflügelindustrie. Im Rahmen dieser Arbeit wurde die cDNA für das selbst-assemblierende Capsid-Protein (VP2, IBDV Stamm IR01) in den Pflanzenexpressionsvektor pTRAkc kloniert und in Tabak exprimiert. Bei transienter Expression wurden für das rekombinante Protein eine Akkumulation von 260 µg/g Frischgewicht im Apoplasten und 160 µg/g Frischgewicht im Cytosol von Tabakblattgewebezellen beobachtet. Analog wurden in stabil transformierten Pflanzenlinien 20-30 µg/g bzw. 10-20 µg/g festgestellt. Das VP2 lag dabei in trimerisierter und nicht wie erhofft in größeren Aggregationsformen vor. Trotzdem war das rekombinante VP2 Protein hoch-immunogen in Mäusen. Alternativ wurde VP2 in den Hefeexpressionvektor pPICZ_B kloniert und in Pichia pastoris produziert. Bei Expression in Hefezellen konnte die Assemblierung von VP2 Monomeren in ~23 nm subvirale Partikel (SVPs) beobachtet werden. Dabei wurden für das rekombinante Protein eine Produktivität von 76 mg pro Liter Kultuvolumen erreicht und die finale Ausbeute nach Reinigung lag bei 38 mg/l. Nach parenteraler und oraler Vakzinierung von Hühnern mit gereinigtem oder Pichia-verkapseltem IBD-SVPcyto konnten IBDV-spezifische Antikörperantworten nachgewiesen werden. Die vakzinierten Tiere wurden mit dem heterologen IBDV-Stamm MB3 infiziert und beobachtet. Anschließend wurden Antikörperantworten, Mortalität, Virus Clearance, Bursale Läsionen und Atrophien sowie das Verhältniss Bursagewicht zu Körpergewicht gemessen und analysiert. Während für intramuskulär verabreichtes, gereinigtes IBD-SVPcyto (20 µg) vollständige Protektion und Virus Clearance sowie die komplette Abwesenheit von Bursaschäden beobachtet wurde, waren bei nicht- oder Mock-immunisierten Tieren eine hohe Mortalitäts-Rate von 60% sowie starke bursale Läsionen und Atrophien zu verzeichnen. Ein als Kontrolle verwendetes kommerzielles Vakzin erreichte ebenfalls vollständige Protektion und Virus Clearance. Auch hier wurden bursale Atrophien beobachtet. Orale Administration von gereinigtem IBD-SVPcyto (500 µg) mit und ohne Adjuvant induzierte eine 100%ige Protektion, wobei nur in Kombination mit dem Adjuvant eine Virus Clearance von 60% beobachtet werden konnte. Bursale Läsionen konnten in beiden Fällen beobachtet werden. Die Administration von 250 mg Pichia-verkapseltem IBD-SVPcyto mit und ohne Adjuvant resultierte in 100%- bzw. 60%iger Protektion, wobei in beiden Gruppen nur moderate bursale Läsionen oder Atrophien festgestellt wurden. In den Gruppen, denen entweder 25 mg Pichia-verkapseltes IBD-SVPcyto oder 50 µg gereinigtes IBD-SVPcyto verabreicht wurde, konnte ein Schutz von 90-100% sowie 40-60% beobachtet werden. Dabei waren in der ersten Gruppe moderate bursale Läsionen und starke Atrophie und in der zweiten Gruppe starke bursale Läsionen und Atrophien zu verzeichen. Über Monitoring und Vergleich der Versuchstier-Lebendgewichte über den Verlauf der Studie wurde außerdem demonstriert, dass es keinen Einfluss der Immunisierungen auf das Wachstum der Tiere gab.

Page 118: Expression and Characterization of Infectious Bursal

Chapter VI Zusammenfassung

112

In dieser Arbeit konnte erfolgreich gezeigt werden, dass in Pichia produzierte subvirale IBDV Partikel sowohl bei parenteraler als auch bei oraler Verabreichung protektive, lokale und systemische Immunantworten gegen IBDV auslösen können. Im zweiten Teil dieser Arbeit wurde die Eignung von Pichia-produzierten subviralen IBDV Partikeln (IBDV-SVPs) als neuartige Nano-Platform untersucht. Gereinigte IBDV-SVPs waren in bis zu 20%tigen Lösungen der organischen Lösungsmittel Ethanol und Dimethylsulfoxid stabil und wiesen eine hohen Temperatur- und pH-Stabilität (65˚C, pH 2.5-9.0) auf. Eine Funktionalisierung konnte am Beispiel der Biokonjugation von Biotin an auf der Oberfläche der IBDV-SVPs liegende Aminogruppen (Lysin) oder Carboxylgruppen (Aspartat und Glutamat) demonstriert werden. Über die Kopplung von Alexa Fluor 488 N-hydroxysuccinimid (NHS) Ester konnte die hohe Zahl von adressierbaren Aminogruppen (>60) pro IBDV-SVPs quantitativ bestätigt werden. Mit diesen Studien konnte gezeigt werden, dass IBDV-SVPs robuste und vielseitig funktionalisierbare Nanopartikel mit großem Entwicklungspotenzial darstellen.

Page 119: Expression and Characterization of Infectious Bursal

Chapter VII References

113

VII References Acosta-Rivero N, Rodriguez A, Musacchio A, Falcon V, Suarez VM, Martinez G, Guerra I, Paz-Lago D, Morera Y, de la Rosa MC, Morales-Grillo J, Duenas-Carrera S. In vitro assembly into virus-like particles is an intrinsic quality of Pichia pastoris derived HCV core protein. Biochem Biophys Res Commun 2004 Dec 3;325(1):68-74. Allnutt FC, Bowers RM, Rowe CG, Vakharia VN, LaPatra SE, Dhar AK. Antigenicity of infectious pancreatic necrosis virus VP2 subviral particles expressed in yeast. Vaccine 2007 Jun 21;25(26):4880-8. Altman E, Bundle DR. Polysaccharide affinity columns for purification of lipopolysaccharide-specific murine monoclonal antibodies. Methods Enzymol 1994;247:243-53. Apweiler R, Hermjakob H, Sharon N. On the frequency of protein glycosylation, as deduced from analysis of the SWISS-PROT database. Biochim Biophys Acta 1999 Dec 6;1473(1):4-8. Arakawa T, Kita Y, Timasheff SN. Protein precipitation and denaturation by dimethyl sulfoxide. Biophysical Chemistry 2007 Dec;131(1-3):62-70. Aricibasi M, Jung A, Heller ED, Rautenschlein S. Differences in genetic background influence the induction of innate and acquired immune responses in chickens depending on the virulence of the infecting infectious bursal disease virus (IBDV) strain. Vet Immunol Immunopathol 2010 May 15;135(1-2):79-92. Azad AA, Barrett SA, Fahey KJ. The characterization and molecular cloning of the double-stranded RNA genome of an Australian strain of infectious bursal disease virus. Virology 1985 May;143(1):35-44. Azad AA, Jagadish MN, Brown MA, Hudson PJ. Deletion mapping and expression in Escherichia coli of the large genomic segment of a birnavirus. Virology 1987 Nov;161(1):145-52. Azad AA, McKern NM, Macreadie IG, Failla P, Heine HG, Chapman A, Ward CW, Fahey KJ. Physicochemical and immunological characterization of recombinant host-protective antigen (VP2) of infectious bursal disease virus. Vaccine 1991 Oct;9(10):715-22. Aziz MA, Midha S, Waheed SM, Bhatnagar R. Oral vaccines: new needs, new possibilities. Bioessays 2007 Jun;29(6):591-604. Bachmann MF, Rohrer UH, Kundig TM, Burki K, Hengartner H, Zinkernagel RM. The influence of antigen organization on B cell responsiveness. Science 1993 262:1448-51. Bae JL, Lee JG, Kang TJ, Jang HS, Jang YS, Yang MS. Induction of antigen-specific systemic and mucosal immune responses by feeding animals transgenic plants expressing the antigen. Vaccine 2003 Sep 8;21(25-26):4052-8.

Page 120: Expression and Characterization of Infectious Bursal

Chapter VII References

114

Bahmaninejad MA, Hair-Bejo M, Omar AR, Aini I, Toroghi R. Characterization of three infectious bursal disease virus isolates obtained from layer chickens in Iran. Acta Virol 2008;52(3):167-74. Barnhill HN, Claudel-Gillet S, Ziessel R, Charbonniere LJ, Wang Q. Prototype protein assembly as scaffold for time-resolved fluoroimmuno assays. J Am Chem Soc 2007a Jun 27;129(25):7799-806. Barnhill HN, Reuther R, Ferguson PL, Dreher T, Wang Q. Turnip yellow mosaic virus as a chemoaddressable bionanoparticle. Bioconjug Chem 2007b May-Jun;18(3):852-9. Bayliss CD, Spies U, Shaw K, Peters RW, Papageorgiou A, Muller H, Boursnell ME. A comparison of the sequences of segment A of four infectious bursal disease virus strains and identification of a variable region in VP2. J Gen Virol 1990 Jun;71 ( Pt 6):1303-12. Becht H, Muller H, Muller HK. Comparative studies on structural and antigenic properties of two serotypes of infectious bursal disease virus. J Gen Virol 1988 Mar;69 ( Pt 3):631-40. Beirao BC, Favaro C, Jr., Nakao LS, Caron LF, Zanata SM, Mercadante AF. Flow cytometric immune profiling of specific-pathogen-free chickens before and after infectious challenges. Vet Immunol Immunopathol 2012 Jan 15;145(1-2):32-41. Bennion BJ, Daggett V. The molecular basis for the chemical denaturation of proteins by urea. Proc Natl Acad Sci U S A 2003 Apr 29;100(9):5142-7. Bentley WE, Wang MY, Vakharia V. Development of an efficient bioprocess for poultry vaccines using high-density insect cell culture. Ann N Y Acad Sci 1994 Nov 30;745:336-59. Biemelt S, Sonnewald U, Galmbacher P, Willmitzer L, Muller M. Production of human papillomavirus type 16 virus-like particles in transgenic plants. J Virol 2003 Sep;77(17):9211-20. Block H, Meyer-Block K, Rebeski DE, Scharr H, de Wit S, Rohn K, Rautenschlein S. A field study on the significance of vaccination against infectious bursal disease virus (IBDV) at the optimal time point in broiler flocks with maternally derived IBDV antibodies. Avian pathology : journal of the WVPA 2007 Oct;36(5):401-9. Bortesi L, Rossato M, Schuster F, Raven N, Stadlmann J, Avesani L, Falorni A, Bazzoni F, Bock R, Schillberg S, Pezzotti M. Viral and murine interleukin-10 are correctly processed and retain their biological activity when produced in tobacco. BMC Biotechnol 2009;9:22. Bosch D, Castilho A, Loos A, Schots A, Steinkellner H. N-Glycosylation of plant-produced recombinant proteins. Curr Pharm Des 2013 Feb 1. Bosch D, Schots A. Plant glycans: friend or foe in vaccine development? Expert Rev Vaccines 2010 Aug;9(8):835-42. Boyaka PN, Tafaro A, Fischer R, Fujihashi K, Jirillo E, McGhee JR. Therapeutic manipulation of the immune system: enhancement of innate and adaptive mucosal immunity. Curr Pharm Des 2003;9(24):1965-72.

Page 121: Expression and Characterization of Infectious Bursal

Chapter VII References

115

Brandtzaeg P, Kiyono H, Pabst R, Russell MW. Terminology: nomenclature of mucosa-associated lymphoid tissue. Mucosal Immunol 2008 Jan;1(1):31-7. Brennan FR, Jones TD, Hamilton WD. Cowpea mosaic virus as a vaccine carrier of heterologous antigens. Mol Biotechnol 2001 Jan;17(1):15-26. Bronstein LM. Virus-based nanoparticles with inorganic cargo: what does the future hold? Small 2011 Jun 20;7(12):1609-18. Brown MD, Skinner MA. Coding sequences of both genome segments of a European 'very virulent' infectious bursal disease virus. Virus Res 1996 Jan;40(1):1-15. Brown SD, Fiedler JD, Finn MG. Assembly of hybrid bacteriophage Qbeta virus-like particles. Biochemistry 2009 Dec 1;48(47):11155-7. Brumfield S, Willits D, Tang L, Johnson JE, Douglas T, Young M. Heterologous expression of the modified coat protein of Cowpea chlorotic mottle bromovirus results in the assembly of protein cages with altered architectures and function. J Gen Virol 2004 Apr;85(Pt 4):1049-53. Brun A, Barcena J, Blanco E, Borrego B, Dory D, Escribano JM, Le Gall-Recule G, Ortego J, Dixon LK. Current strategies for subunit and genetic viral veterinary vaccine development. Virus Res 2011 Apr;157(1):1-12. Brunel FM, Lewis JD, Destito G, Steinmetz NF, Manchester M, Stuhlmann H, Dawson PE. Hydrazone ligation strategy to assemble multifunctional viral nanoparticles for cell imaging and tumor targeting. Nano Lett 2010 Mar 10;10(3):1093-7. Caldeira JC, Peabody DS. Stability and assembly in vitro of bacteriophage PP7 virus-like particles. J Nanobiotechnology 2007;5:10. Carballeda JM, Zoth SC, Gomez E, Gravisaco MJ, Berinstein A. Activation of the immune response against Infectious Bursal Disease Virus after intramuscular inoculation of an intermediate strain. Immunobiology 2011 Sep;216(9):1028-33. Casanas A, Navarro A, Ferrer-Orta C, Gonzalez D, Rodriguez JF, Verdaguer N. Structural insights into the multifunctional protein VP3 of birnaviruses. Structure 2008 Jan;16(1):29-37. Casares S, Inaba K, Brumeanu TD, Steinman RM, Bona CA. Antigen presentation by dendritic cells after immunization with DNA encoding a major histocompatibility complex class II-restricted viral epitope. J Exp Med 1997 Nov 3;186(9):1481-6. Caston JR, Martinez-Torrecuadrada JL, Maraver A, Lombardo E, Rodriguez JF, Casal JI, Carrascosa JL. C terminus of infectious bursal disease virus major capsid protein VP2 is involved in definition of the T number for capsid assembly. J Virol 2001 Nov;75(22):10815-28. Chakma J, Masum H, Perampaladas K, Heys J, Singer PA. Indian vaccine innovation: the case of Shantha Biotechnics. Globalization and health 2011;7:9. Chandra A, D. Pental. Regeneration and genetic transformation of grain legumes: An overview. Current science 2003;84(3):381-7.

Page 122: Expression and Characterization of Infectious Bursal

Chapter VII References

116

Chang HC, Lin TL, Wu CC. DNA-mediated vaccination against infectious bursal disease in chickens. Vaccine 2001 Nov 12;20(3-4):328-35. Chebolu S, Daniell H. Chloroplast-derived vaccine antigens and biopharmaceuticals: expression, folding, assembly and functionality. Curr Top Microbiol Immunol 2009;332:33-54. Chen CS, Suen SY, Lai SY, Chang GR, Lu TC, Lee MS, Wang MY. Purification of capsid-like particles of infectious bursal disease virus (IBDV) VP2 expressed in E. coli with a metal-ion affinity membrane system. J Virol Methods 2005 Dec;130(1-2):51-8. Chen TH, Chen TH, Hu CC, Liao JT, Lee CW, Liao JW, Lin MY, Liu HJ, Wang MY, Lin NS, Hsu YH. Induction of protective immunity in chickens immunized with plant-made chimeric Bamboo mosaic virus particles expressing very virulent Infectious bursal disease virus antigen. Virus Res 2012 Mar 1. Cheng HW, Eicher SD, Chen Y, Singleton P, Muirt WM. Effect of genetic selection for group productivity and longevity on immunological and hematological parameters of chickens. Poultry science 2001a Aug;80(8):1079-86. Cheng YS, Lee MS, Lai SY, Doong SR, Wang MY. Separation of pure and immunoreactive virus-like particles using gel filtration chromatography following immobilized metal ion affinity chromatography. Biotechnol Prog 2001b Mar-Apr;17(2):318-25. Chevalier C, Lepault J, Da Costa B, Delmas B. The last C-terminal residue of VP3, glutamic acid 257, controls capsid assembly of infectious bursal disease virus. J Virol 2004 Apr;78(7):3296-303. Chevalier C, Lepault J, Erk I, Da Costa B, Delmas B. The maturation process of pVP2 requires assembly of infectious bursal disease virus capsids. J Virol 2002 Mar;76(5):2384-92. Chikwamba R, McMurray J, Shou H, Frame B, Pegg SE, Scott P, Mason H and Wang K. Expression of a synthetic E. coli heat labile enterotoxin B subunit (LT B) in maize. Molecular Breeding 2002 a;10(4):253-65. Comellas-Aragones M, de la Escosura A, Dirks AT, van der Ham A, Fuste-Cune A, Cornelissen JJ, Nolte RJ. Controlled integration of polymers into viral capsids. Biomacromolecules 2009 Nov 9;10(11):3141-7. Condon C, Watkins SC, Celluzzi CM, Thompson K, Falo LD, Jr. DNA-based immunization by in vivo transfection of dendritic cells. Nat Med 1996 Oct;2(10):1122-8. Copland MJ, Rades T, Davies NM, Baird MA. Lipid based particulate formulations for the delivery of antigen. Immunol Cell Biol 2005 Apr;83(2):97-105. Cosgrove AA. An apparently new disease of chicks- avian nephrosis. Avian Dis 1962; 6:385-551.

Coulibaly F, Chevalier C, Gutsche I, Pous J, Navaza J, Bressanelli S, Delmas B, Rey FA. The birnavirus crystal structure reveals structural relationships among icosahedral viruses. Cell 2005 Mar 25;120(6):761-72.

Page 123: Expression and Characterization of Infectious Bursal

Chapter VII References

117

Crisci E, Fraile L, Moreno N, Blanco E, Cabezon R, Costa C, Mussa T, Baratelli M, Martinez-Orellana P, Ganges L, Martinez J, Barcena J, Montoya M. Chimeric calicivirus-like particles elicit specific immune responses in pigs. Vaccine 2012 Feb 3. Cui X, Nagesha HS, Holmes IH. Identification of crucial residues of conformational epitopes on VP2 protein of infectious bursal disease virus by phage display. J Virol Methods 2003 Apr;109(1):75-83. Da Costa B, Chevalier C, Henry C, Huet JC, Petit S, Lepault J, Boot H, Delmas B. The capsid of infectious bursal disease virus contains several small peptides arising from the maturation process of pVP2. J Virol 2002 Mar;76(5):2393-402. Dar A, Potter A, Tikoo S, Gerdts V, Lai K, Babiuk LA, Mutwiri G. CpG oligodeoxynucleotides activate innate immune response that suppresses infectious bronchitis virus replication in chicken embryos. Avian Dis 2009 Jun;53(2):261-7. Davis HL, McCluskie MJ. DNA vaccines for viral diseases. Microbes Infect 1999 Jan;1(1):7-21. Davison F, Magor KE, Kaspers B. Structure and evolution of avian immunoglobulins. In F. Davison, B. Kaspers & K.A. Schat (Eds.). Avian Immunology 1st edn (pp. 107-128). London: Academic Press. 2008. Davoodi-Semiromi A, Samson N, Daniell H. The green vaccine: A global strategy to combat infectious and autoimmune diseases. Hum Vaccin 2009 Jul;5(7):488-93. Demento SL, Bonafe N, Cui W, Kaech SM, Caplan MJ, Fikrig E, Ledizet M, Fahmy TM. TLR9-targeted biodegradable nanoparticles as immunization vectors protect against West Nile encephalitis. J Immunol 2010 Sep 1;185(5):2989-97. Dertzbaugh MT. Genetically engineered vaccines: an overview. Plasmid 1998;39(2):100-13. Destito G, Yeh R, Rae CS, Finn MG, Manchester M. Folic acid-mediated targeting of cowpea mosaic virus particles to tumor cells. Chem Biol 2007 Oct;14(10):1152-62. Dey S, Upadhyay C, Madhan Mohan C, Kataria JM, Vakharia VN. Formation of subviral particles of the capsid protein VP2 of infectious bursal disease virus and its application in serological diagnosis. J Virol Methods 2009 Apr;157(1):84-9. Dhar AK, Bowers RM, Rowe CG, Allnutt FC. Expression of a foreign epitope on infectious pancreatic necrosis virus VP2 capsid protein subviral particle (SVP) and immunogenicity in rainbow trout. Antiviral Res 2010 Mar;85(3):525-31. Donnelly JJ, Ulmer JB, Shiver JW, Liu MA. DNA vaccines. Annu Rev Immunol 1997;15:617-48. Donnelly JJ, Wahren B, Liu MA. DNA vaccines: progress and challenges. J Immunol 2005 Jul 15;175(2):633-9.

Page 124: Expression and Characterization of Infectious Bursal

Chapter VII References

118

Doong SR, Chen YH, Lai SY, Lee CC, Lin YC, Wang MY. Strong and heterogeneous adsorption of infectious bursal disease VP2 subviral particle with immobilized metal ions dependent on two surface histidine residues. Analytical Chemistry 2007 Oct 15;79(20):7654-61. Dower WJ, Miller JF, Ragsdale CW. High efficiency transformation of E. coli by high voltage electroporation. Nucleic Acids Res 1988 Jul 11;16(13):6127-45. Dybing JK, Jackwood DJ. Expression of MD infectious bursal disease viral proteins in baculovirus. Avian Dis 1997 Jul-Sep;41(3):617-26. Eo SK, Lee S, Kumaraguru U, Rouse BT. Immunopotentiation of DNA vaccine against herpes simplex virus via co-delivery of plasmid DNA expressing CCR7 ligands. Vaccine 2001 Sep 14;19(32):4685-93. Epizooties OId. Infectious bursal disease virus. In Manual of Diagnostic Tests and Vaccines for Terrestrial Animals 2009(pp. 549-565):Paris: OIE.

Eteradossi N, Saif YM. Infectious Bursal Disease. In: Diseases of Poultry, Iowa State Press, Ames, Iowa. 2008 2008:pp185-209. Fahey KJ, Erny K, Crooks J. A conformational immunogen on VP-2 of infectious bursal disease virus that induces virus-neutralizing antibodies that passively protect chickens. J Gen Virol 1989 Jun;70 ( Pt 6):1473-81. Falk K, Lau JM, Santambrogio L, Esteban VM, Puentes F, Rotzschke O, Strominger JL. Ligand exchange of major histocompatibility complex class II proteins is triggered by H-bond donor groups of small molecules. J Biol Chem 2002 Jan 25;277(4):2709-15. Faye L, Boulaflous A, Benchabane M, Gomord V, Michaud D. Protein modifications in the plant secretory pathway: current status and practical implications in molecular pharming. Vaccine 2005 Mar 7;23(15):1770-8. Faye L, Gomord V, Fitchette-Laine AC, Chrispeels MJ. Affinity purification of antibodies specific for Asn-linked glycans containing alpha 1-->3 fucose or beta 1-->2 xylose. Anal Biochem 1993 Feb 15;209(1):104-8. Fernandez-Arias A, Risco C, Martinez S, Albar JP, Rodriguez JF. Expression of ORF A1 of infectious bursal disease virus results in the formation of virus-like particles. J Gen Virol 1998 May;79 ( Pt 5):1047-54. Fischer R, Stoger E, Schillberg S, Christou P, Twyman RM. Plant-based production of biopharmaceuticals. Curr Opin Plant Biol 2004 Apr;7(2):152-8. Fischer R, Schillberg S. Molecular farming. Plant-derived vaccines: progress and constraints. Wiley-VCH Verlag GmbH & Co kGaA, Weinheim, Germany 2004. Garg NK, Mangal S, Khambete H, Sharma PK, Tyagi RK. Mucosal delivery of vaccines: role of mucoadhesive/biodegradable polymers. Recent Pat Drug Deliv Formul 2010 Jun 1;4(2):114-28.

Page 125: Expression and Characterization of Infectious Bursal

Chapter VII References

119

Garriga D, Navarro A, Querol-Audi J, Abaitua F, Rodriguez JF, Verdaguer N. Activation mechanism of a noncanonical RNA-dependent RNA polymerase. Proc Natl Acad Sci U S A 2007 Dec 18;104(51):20540-5. Garriga D, Querol-Audi J, Abaitua F, Saugar I, Pous J, Verdaguer N, Caston JR, Rodriguez JF. The 2.6-Angstrom structure of infectious bursal disease virus-derived T=1 particles reveals new stabilizing elements of the virus capsid. J Virol 2006 Jul;80(14):6895-905. Gillitzer E, Willits D, Young M, Douglas T. Chemical modification of a viral cage for multivalent presentation. Chem Commun (Camb) 2002 Oct 21(20):2390-1. Grasso S, Santi L. Viral nanoparticles as macromolecular devices for new therapeutic and pharmaceutical approaches. Int J Physiol Pathophysiol Pharmacol 2010;2(2):161-78. Gravier R, Dory D, Laurentie M, Bougeard S, Cariolet R, Jestin A. In vivo tissue distribution and kinetics of a pseudorabies virus plasmid DNA vaccine after intramuscular injection in swine. Vaccine 2007 Sep 28;25(39-40):6930-8. Grgacic EV, Anderson DA. Virus-like particles: passport to immune recognition. Methods 2006 Sep;40(1):60-5. Guy-Grand D, Griscelli C, Vassalli P. The gut-associated lymphoid system: nature and properties of the large dividing cells. Eur J Immunol 1974 Jun;4(6):435-43. Hair-Bejo M, Ng MK, Ng HY. Day Old Vaccination Against Infectious Bursal Disease in Broiler Chickens. International J Poultry Science 2004;3 (2):124-8. Hassan MK, Afify M, Aly MM. Susceptibility of vaccinated and unvaccinated Egyptian chickens to very virulent infectious bursal disease virus. Avian pathology : Journal of the WVPA 2002 Apr;31(2):149-56. Heine HG, Boyle DB. Infectious bursal disease virus structural protein VP2 expressed by a fowlpox virus recombinant confers protection against disease in chickens. Arch Virol 1993;131(3-4):277-92. Heine HG, Haritou M, Failla P, Fahey K, Azad A. Sequence analysis and expression of the host-protective immunogen VP2 of a variant strain of infectious bursal disease virus which can circumvent vaccination with standard type I strains. J Gen Virol 1991 Aug;72 ( Pt 8):1835-43. Hellwig S, Robin F, Drossard J, Raven NP, Vaquero-Martin C, Shively JE, Fischer R. Production of carcinoembryonic antigen (CEA) N-A3 domain in Pichia pastoris by fermentation. Biotechnol Appl Biochem 1999 Dec;30 ( Pt 3):267-75. Hjalmarsson A, Carlemalm E, Everitt E. Infectious pancreatic necrosis virus: identification of a VP3-containing ribonucleoprotein core structure and evidence for O-linked glycosylation of the capsid protein VP2. J Virol 1999 Apr;73(4):3484-90. Holmgren J, Czerkinsky C. Mucosal immunity and vaccines. Nat Med 2005 Apr;11(4 Suppl):S45-53.

Page 126: Expression and Characterization of Infectious Bursal

Chapter VII References

120

Hood EE, Woodard SL, Horn ME. Monoclonal antibody manufacturing in transgenic plants--myths and realities. Curr Opin Biotechnol 2002 13, 630-635. Hopkins D, Gomathinayagam S, Rittenhour AM, Du M, Hoyt E, Karaveg K, Mitchell T, Nett JH, Sharkey NJ, Stadheim TA, Li H, Hamilton SR. Elimination of beta-mannose glycan structures in Pichia pastoris. Glycobiology 2011 Dec;21(12):1616-26. Hoque MM, Omar AR, Chong LK, Hair-Bejo M, Aini I. Pathogenicity of SspI-positive infectious bursal disease virus and molecular characterization of the VP2 hypervariable region. Avian pathology : journal of the WVPA 2001 Aug;30(4):369-80. Horton RM, Hunt HD, Ho SN, Pullen JK, Pease LR. Engineering hybrid genes without the use of restriction enzymes: gene splicing by overlap extension. Gene 1989 Apr 15;77(1):61-8. Hoshi S, Nakamura T, Nunoya T, Ueda S. Induction of protective immunity in chickens orally immunized with inactivated infectious bursal disease virus. Vaccine 1995 Feb;13(3):245-52. Hoshi S, Uchino A, Saito N, Kusanagi KI, Ihara T, Ueda S. Comparison of adjuvants with respect to serum IgG antibody response in orally immunized chickens. Comp Immunol Microbiol Infect Dis 1999 Jan;22(1):63-9. Hu YC, Bentley WE. A kinetic and statistical-thermodynamic model or baculovirus infection and virus-like particle assembly in suspended insect cells. Chemical Engineering Science 2000;55:3991-4008. Hu YC, Bentley WE, Edwards GH, Vakharia VN. Chimeric infectious bursal disease virus-like particles expressed in insect cells and purified by immobilized metal affinity chromatography. Biotechnol Bioeng 1999 Jun 20;63(6):721-9. Huang Z, Elankumaran S, Yunus AS, Samal SK. A recombinant Newcastle disease virus (NDV) expressing VP2 protein of infectious bursal disease virus (IBDV) protects against NDV and IBDV. J Virol 2004 Sep;78(18):10054-63. Huang Z, Elkin G, Maloney BJ, Beuhner N, Arntzen CJ, Thanavala Y, Mason HS. Virus-like particle expression and assembly in plants: hepatitis B and Norwalk viruses. Vaccine 2005 Mar 7;23(15):1851-8. Hudson PJ, McKern NM, Power BE, Azad AA. Genomic structure of the large RNA segment of infectious bursal disease virus. Nucleic Acids Res 1986 Jun 25;14(12):5001-12. Hwang KW. Haemophilus influenza type b (Hib) vaccine and its carrier proteins. Arch Pharm Res 2010 Jun;33(6):793-5. Ismail NM, Saif YM. Immunogenicity of infectious bursal disease viruses in chickens. Avian Dis 1991 Jul-Sep;35(3):460-9. Jackwood DJ, Jackwood RJ. Infectious bursal disease viruses: molecular differentiation of antigenic subtypes among serotype 1 viruses. Avian Dis 1994 Jul-Sep;38(3):531-7. Jackwood DJ, Sommer SE. Molecular studies on suspect very virulent infectious bursal disease virus genomic RNA samples. Avian Dis 2005 Jun;49(2):246-51.

Page 127: Expression and Characterization of Infectious Bursal

Chapter VII References

121

Jackwood DJ, Sommer SE, Knoblich HV. Amino acid comparison of infectious bursal disease viruses placed in the same or different molecular groups by RT/PCR-RFLP. Avian Dis 2001 Apr-Jun;45(2):330-9. Jagadish MN, Staton VJ, Hudson PJ, Azad AA. Birnavirus precursor polyprotein is processed in Escherichia coli by its own virus-encoded polypeptide. J Virol 1988 Mar;62(3):1084-7. Jagadish MN, Vaughan PR, Irving RA, Azad AA, Macreadie IG. Expression and characterization of infectious bursal disease virus polyprotein in yeast. Gene 1990 Nov 15;95(2):179-86. Jennings GT, Bachmann MF. The coming of age of virus-like particle vaccines. Biological chemistry 2008 May;389(5):521-36. Jesnowski R, Naehring J, Wolf K. A rapid and reliable method for PCR-based amplification of chromosomal and mitochondrial DNA from intact yeast cells. Curr Genet 1995 Mar;27(4):318-9. Jha RK, Xu ZR, Bai SJ, Sun JY, Li WF, Shen J. Protection of Procambarus clarkii against white spot syndrome virus using recombinant oral vaccine expressed in Pichia pastoris. Fish Shellfish Immunol 2007 22, 295-307. Jiang Z, Tong G, Cai B, Xu Y, Lou J. Purification and immunogenicity study of human papillomavirus 58 virus-like particles expressed in Pichia pastoris. Protein Expr Purif 2011 Dec;80(2):203-10. Johansen FE, Natvig Norderhaug I, Roe M, Sandlie I, Brandtzaeg P. Recombinant expression of polymeric IgA: incorporation of J chain and secretory component of human origin. Eur J Immunol 1999a May;29(5):1701-8. Johansen FE, Pekna M, Norderhaug IN, Haneberg B, Hietala MA, Krajci P, Betsholtz C, Brandtzaeg P. Absence of epithelial immunoglobulin A transport, with increased mucosal leakiness, in polymeric immunoglobulin receptor/secretory component-deficient mice. J Exp Med 1999b Oct 4;190(7):915-22. Juneja SS, Ramneek, Deka D, Oberoi MS, Singh A. Molecular characterization of field isolates and vaccine strains of infectious bursal disease virus. Comp Immunol Microbiol Infect Dis 2008 Jan;31(1):11-23. Kaiser P. The avian immune genome-a glass half-full or half-empty? Cytogenetic and genome research 2007;117(1-4):221-30. Kaltgrad E, O'Reilly MK, Liao L, Han S, Paulson JC, Finn MG. On-virus construction of polyvalent glycan ligands for cell-surface receptors. J Am Chem Soc 2008 Apr 9;130(14):4578-9. Kapila J, De Rycke R, van Montagu M, Agenon G. An Agrobacterium mediated transient gene expression system for intact leaves. Plant Sci 1997;122:101-8. Kasanga CJ, Yamaguchi T, Wambura PN, Maeda-Machang'u AD, Ohya K, Fukushi H. Molecular characterization of infectious bursal disease virus (IBDV): diversity of very virulent IBDV in Tanzania. Arch Virol 2007;152(4):783-90.

Page 128: Expression and Characterization of Infectious Bursal

Chapter VII References

122

Kawai T, Akira S. Toll-like receptor and RIG-I-like receptor signaling. Ann N Y Acad Sci 2008 Nov;1143:1-20. Kelsall BL, Strober W. Distinct populations of dendritic cells are present in the subepithelial dome and T cell regions of the murine Peyer's patch. J Exp Med 1996 Jan 1;183(1):237-47. Kerneis S, Bogdanova A, Kraehenbuhl JP, Pringault E. Conversion by Peyer's patch lymphocytes of human enterocytes into M cells that transport bacteria. Science 1997 Aug 15;277(5328):949-52. Kibenge FS, Qian B, Nagy E, Cleghorn JR, Wadowska D. Formation of virus-like particles when the polyprotein gene (segment A) of infectious bursal disease virus is expressed in insect cells. Can J Vet Res 1999 Jan;63(1):49-55. Kim R, Yokota H, Kim SH. Electrophoresis of proteins and protein-protein complexes in a native agarose gel. Anal Biochem 2000 Jun 15;282(1):147-9. Klikova M, Rhee SS, Hunter E, Ruml T. Efficient in vivo and in vitro assembly of retroviral capsids from Gag precursor proteins expressed in bacteria. J Virol 1995 Feb;69(2):1093-8. Klipper E, Sklan D, Friedman A. Immune responses of chickens to dietary protein antigens. I. Induction of systemic and intestinal immune responses following oral administration of soluble proteins in the absence of adjuvant. Vet Immunol Immunopathol 2000 May 23;74(3-4):209-23. Kochan G, Gonzalez D, Rodriguez JF. Characterization of the RNA-binding activity of VP3, a major structural protein of Infectious bursal disease virus. Arch Virol 2003 Apr;148(4):723-44. Kolarich D, Weber A, Turecek PL, Schwarz HP, Altmann F. Comprehensive glyco-proteomic analysis of human alpha1-antitrypsin and its charge isoforms. Proteomics 2006 Jun;6(11):3369-80. Kong LL, Omar AR, Hair Bejo M, Ideris A, Tan SW. Development of SYBR green I based one-step real-time RT-PCR assay for the detection and differentiation of very virulent and classical strains of infectious bursal disease virus. J Virol Methods 2009 Jul 8. Kusnadi AR, Hood EE, Witcher DR, Howard JA, Nikolov ZL. Production and purification of two recombinant proteins from transgenic corn. Biotechnol Prog 1998 Jan-Feb;14(1):149-55. Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 1970 Aug 15;227(5259):680-5. Lai SY, Lee MS, Chen HC, Shen PC, Jinn TR, Kao SS, Wang MY. Production and purification of immunogenic virus-like particles formed by the chimeric infectious bursal disease virus structural protein, rVP2H, in insect larvae. Process Biochemistry 2004;39:571-7. Lee CC, Ko TP, Chou CC, Yoshimura M, Doong SR, Wang MY, Wang AH. Crystal structure of infectious bursal disease virus VP2 subviral particle at 2.6A resolution: implications in virion assembly and immunogenicity. J Struct Biol 2006a Jul;155(1):74-86.

Page 129: Expression and Characterization of Infectious Bursal

Chapter VII References

123

Lee LA, Niu Z, Wang Q. Viruses and virus-like protein assemblies—Chemically programmable nanoscale building blocks. Nano Res 2009;2(5):349-64. Lee LH, Yu SL, Shieh HK. Detection of infectious bursal disease virus infection using the polymerase chain reaction. J Virol Methods 1992 Dec 1;40(3):243-53. Lee MS, Doong SR, Lai SY, Ho JY, Wang MY. Processing of infectious bursal disease virus (IBDV) polyprotein and self-assembly of IBDV-like particles in Hi-5 cells. Biotechnol Prog 2006b May-Jun;22(3):763-9. Lerouge P, Cabanes-Macheteau M, Rayon C, Fischette-Laine AC, Gomord V, Faye L. N-glycoprotein biosynthesis in plants: recent developments and future trends. Plant Mol Biol 1998 Sep;38(1-2):31-48. Lessard IA, Domingo GJ, Borges A, Perham RN. Expression of genes encoding the E2 and E3 components of the Bacillus stearothermophilus pyruvate dehydrogenase complex and the stoichiometry of subunit interaction in assembly in vitro. Eur J Biochem 1998 Dec 1;258(2):491-501. Letzel T, Coulibaly F, Rey FA, Delmas B, Jagt E, van Loon AA, Mundt E. Molecular and structural bases for the antigenicity of VP2 of infectious bursal disease virus. J Virol 2007 Dec;81(23):12827-35. Levin B. R. LM, Bonhoeffer S. Population biology, evolution, and infectious disease: convergence and synthesis. Science 1999;283:806–9. Li J, Liang X, Huang Y, Meng S, Xie R, Deng R, Yu L. Enhancement of the immunogenicity of DNA vaccine against infectious bursal disease virus by co-delivery with plasmid encoding chicken interleukin 2. Virology 2004 Nov 10;329(1):89-100. Li Y, Wu S, Ouyang J, Mao L, Li W, Lin H. Expression of insulin-like growth factor-1 of orange-spotted grouper (Epinephelus coioides) in yeast Pichia pastoris. Protein Expr Purif 2012 84, 80-85. Li Y, Wu T, Cheng X, Zhang C. Molecular characteristic of VP2 gene of infectious bursal disease viruses isolated from a farm in two decades. Virus Genes 2009 Jun;38(3):408-13. Lillard JW, Jr., Boyaka PN, Taub DD, McGhee JR. RANTES potentiates antigen-specific mucosal immune responses. J Immunol 2001 Jan 1;166(1):162-9. Lim BL, Cao Y, Yu T, Mo CW. Adaptation of very virulent infectious bursal disease virus to chicken embryonic fibroblasts by site-directed mutagenesis of residues 279 and 284 of viral coat protein VP2. J Virol 1999 Apr;73(4):2854-62. Lin GJ, Deng MC, Chen ZW, Liu TY, Wu CW, Cheng CY, Chien MS, Huang C. Yeast expressed classical swine fever E2 subunit vaccine candidate provides complete protection against lethal challenge infection and prevents horizontal virus transmission. Vaccine 2012 Mar 16;30(13):2336-41.

Page 130: Expression and Characterization of Infectious Bursal

Chapter VII References

124

Lin TW, Lo CW, Lai SY, Fan RJ, Lo CJ, Chou YM, Thiruvengadam R, Wang AH, Wang MY. Chicken heat shock protein 90 is a component of the putative cellular receptor complex of infectious bursal disease virus. J Virol 2007 Aug;81(16):8730-41. Lipin ID, Chuan YP, Neibert M, Fan YY, Middelberg A. Processing and in vitro Assembly of Virus Like Particle Nanostructures. IEEExplore 2006. Littel-van den Hurk SV, Babiuk SL, Babiuk LA. Strategies for improved formulation and delivery of DNA vaccines to veterinary target species. Immunological Reviews 2004 Jun;199(1):113-25. Liu J, Zhou J, Kwang J. Antigenic and molecular characterization of recent infectious bursal disease virus isolates in China. Virus Genes 2002 Mar;24(2):135-47. Lockney DM, Guenther RN, Loo L, Overton W, Antonelli R, Clark J, Hu M, Luft C, Lommel SA, Franzen S. The Red clover necrotic mosaic virus capsid as a multifunctional cell targeting plant viral nanoparticle. Bioconjug Chem 2011 Jan 19;22(1):67-73. Luque D, Saugar I, Rodriguez JF, Verdaguer N, Garriga D, Martin CS, Velazquez-Muriel JA, Trus BL, Carrascosa JL, Caston JR. Infectious bursal disease virus capsid assembly and maturation by structural rearrangements of a transient molecular switch. J Virol 2007 Jul;81(13):6869-78. MacLachlat N. J. and Stott JL. Birnaviridae. In: Hirsh DC, MacLachlan N. J., and Walker, R. L., editor. Veterinary microbiology 2nd edition. Ames, Iowa: Blackwell Publishing, 2005: 407-9, Chapter 64. Macreadie IG, Vaughan PR, Chapman AJ, McKern NM, Jagadish MN, Heine HG, Ward CW, Fahey KJ, Azad AA. Passive protection against infectious bursal disease virus by viral VP2 expressed in yeast. Vaccine 1990 Dec;8(6):549-52. Mahgoub HA. An overview of infectious bursal disease. Arch Virol 2012 Jun 17. Malabadi RB, Ganguly A, Silva JA, Parashar A, Suresh MR, Sunwoo H. Overview of plant-derived vaccine antigens: Dengue virus. Journal of pharmacy & pharmaceutical sciences : a publication of the Canadian Society for Pharmaceutical Sciences, Societe canadienne des sciences pharmaceutiques 2011;14(3):400-13. Mallick AI, Parvizi P, Read LR, Nagy E, Behboudi S, Sharif S. Enhancement of immunogenicity of a virosome-based avian influenza vaccine in chickens by incorporating CpG-ODN. Vaccine 2011 Feb 11;29(8):1657-65. Mann JF, Shakir E, Carter KC, Mullen AB, Alexander J, Ferro VA. Lipid vesicle size of an oral influenza vaccine delivery vehicle influences the Th1/Th2 bias in the immune response and protection against infection. Vaccine 2009 Jun 2;27(27):3643-9. Manzenrieder F, Luxenhofer R, Retzlaff M, Jordan R, Finn MG. Stabilization of virus-like particles with poly(2-oxazoline)s. Angew Chem Int Ed Engl 2011 Mar 7;50(11):2601-5.

Page 131: Expression and Characterization of Infectious Bursal

Chapter VII References

125

Maraver A, Clemente R, Rodriguez JF, Lombardo E. Identification and molecular characterization of the RNA polymerase-binding motif of infectious bursal disease virus inner capsid protein VP3. J Virol 2003 Feb;77(4):2459-68. Martinez-Torrecuadrada JL, Caston JR, Castro M, Carrascosa JL, Rodriguez JF, Casal JI. Different architectures in the assembly of infectious bursal disease virus capsid proteins expressed in insect cells. Virology 2000 Dec 20;278(2):322-31. Martinez-Torrecuadrada JL, Saubi N, Pages-Mante A, Caston JR, Espuna E, Casal JI. Structure-dependent efficacy of infectious bursal disease virus (IBDV) recombinant vaccines. Vaccine 2003 Jul 4;21(23):3342-50. Mason HS, Ball JM, Shi JJ, Jiang X, Estes MK, Arntzen CJ. Expression of Norwalk virus capsid protein in transgenic tobacco and potato and its oral immunogenicity in mice. Proc Natl Acad Sci U S A 1996 May 28;93(11):5335-40. Mason HS, Lam DM, Arntzen CJ. Expression of hepatitis B surface antigen in transgenic plants. Proc Natl Acad Sci U S A 1992 Dec 15;89(24):11745-9. Mattanovich D, Branduardi P, Dato L, Gasser B, Sauer M, Porro D. Recombinant protein production in yeasts. Methods Mol Biol 2012;824:329-58. Mayer L, Shao L. Therapeutic potential of oral tolerance. Nature reviews Immunology 2004 Jun;4(6):407-19. McCluskie MJ, Davis HL. Mucosal immunization with DNA vaccines. Microbes Infect 1999 Jul;1(9):685-98. McCluskie MJ, Weeratna RD, Krieg AM, Davis HL. CpG DNA is an effective oral adjuvant to protein antigens in mice. Vaccine 2000 Nov 22;19(7-8):950-7. McConkey SJ, Reece WH, Moorthy VS, Webster D, Dunachie S, Butcher G, Vuola JM, Blanchard TJ, Gothard P, Watkins K, Hannan CM, Everaere S, Brown K, Kester KE, Cummings J, Williams J, Heppner DG, Pathan A, Flanagan K, Arulanantham N, Roberts MT, Roy M, Smith GL, Schneider J, Peto T, Sinden RE, Gilbert SC, Hill AV. Enhanced T-cell immunogenicity of plasmid DNA vaccines boosted by recombinant modified vaccinia virus Ankara in humans. Nat Med 2003 Jun;9(6):729-35. Miermont A, Barnhill H, Strable E, Lu X, Wall KA, Wang Q, Finn MG, Huang X. Cowpea mosaic virus capsid: a promising carrier for the development of carbohydrate based antitumor vaccines. Chemistry 2008;14(16):4939-47. Mishra N, Goyal AK, Tiwari S, Paliwal R, Paliwal SR, Vaidya B, Mangal S, Gupta M, Dube D, Mehta A, Vyas SP. Recent advances in mucosal delivery of vaccines: role of mucoadhesive/biodegradable polymeric carriers. Expert Opin Ther Pat 2010 May;20(5):661-79. Mockett AP, Rose ME. Immune responses to eimeria: quantification of antibody isotypes to Eimeria tenella in chicken serum and bile by means of the ELISA. Parasite Immunol 1986 Sep;8(5):481-9.

Page 132: Expression and Characterization of Infectious Bursal

Chapter VII References

126

Muller H, Mundt E, Eterradossi N, Islam MR. Current status of vaccines against infectious bursal disease. Avian pathology : Journal of the WVPA 2012 Apr;41(2):133-9. Muller H, Scholtissek C, Becht H. The genome of infectious bursal disease virus consists of two segments of double-stranded RNA. J Virol 1979 Sep;31(3):584-9. Mundt E, Kollner B, Kretzschmar D. VP5 of infectious bursal disease virus is not essential for viral replication in cell culture. J Virol 1997 Jul;71(7):5647-51. Neutra MR, Frey A, Kraehenbuhl JP. Epithelial M cells: gateways for mucosal infection and immunization. Cell 1996 Aug 9;86(3):345-8. Neutra MR, Kozlowski PA. Mucosal vaccines: the promise and the challenge. Nature Reviews Immunology 2006 Feb;6(2):148-58. Nichols ME, Stanislaus T, Keshavarz-Moore E, Young HA. Disruption of leaves and initial extraction of wildtype CPMV virus as a basis for producing vaccines from plants. J Biotechnol 2002 Jan 18;92(3):229-35. O'Hagan DT. Recent advances in immunological adjuvants: the development of particulate antigen delivery systems. Expert Opin Investig Drugs 1998 Mar;7(3):349-59. Ona A, Luque D, Abaitua F, Maraver A, Caston JR, Rodriguez JF. The C-terminal domain of the pVP2 precursor is essential for the interaction between VP2 and VP3, the capsid polypeptides of infectious bursal disease virus. Virology 2004 Apr 25;322(1):135-42. Parede LH, Sapats S, Gould G, Rudd M, Lowther S, Ignjatovic J. Characterization of infectious bursal disease virus isolates from Indonesia indicates the existence of very virulent strains with unique genetic changes. Avian pathology : journal of the WVPA 2003 Oct;32(5):511-8. Paul M, Ma JK. Plant-made immunogens and effective delivery strategies. Expert Rev Vaccines 2010 Aug;9(8):821-33. Peabody DS. A Viral Platform for Chemical Modification and Multivalent Display. J Nanobiotechnology 2003 Jul 15;1(1):5. Petrescu AJ, Milac AL, Petrescu SM, Dwek RA, Wormald MR. Statistical analysis of the protein environment of N-glycosylation sites: implications for occupancy, structure, and folding. Glycobiology 2004 Feb;14(2):103-14. Phenix KV, Wark K, Luke CJ, Skinner MA, Smyth JA, Mawhinney KA, Todd D. Recombinant Semliki Forest virus vector exhibits potential for avian virus vaccine development. Vaccine 2001 Apr 30;19(23-24):3116-23. Pitcovski J, Gutter B, Gallili G, Goldway M, Perelman B, Gross G, Krispel S, Barbakov M, Michael A. Development and large-scale use of recombinant VP2 vaccine for the prevention of infectious bursal disease of chickens. Vaccine 2003 Dec 1;21(32):4736-43.

Page 133: Expression and Characterization of Infectious Bursal

Chapter VII References

127

Pous J, Chevalier C, Ouldali M, Navaza J, Delmas B, Lepault J. Structure of birnavirus-like particles determined by combined electron cryomicroscopy and X-ray crystallography. J Gen Virol 2005 Aug;86(Pt 8):2339-46. Promdonkoy P, Tang K, Sornlake W, Harnpicharnchai P, Kobayashi RS, Ruanglek V, Upathanpreecha T, Vesaratchavest M, Eurwilaichitr L, Tanapongpipat S. Expression and characterization of Aspergillus thermostable phytases in Pichia pastoris. FEMS Microbiol Lett 2009 Jan;290(1):18-24. Provencher S. CONTIN: A general purpose constrained regularization program for inverting noisy linear algebraic and integral equations. Comput Phys Commun 1982a;27:229-42. Provencher SW. A constrained regularization method for inverting data represented by linear algebraic or integral equations. Comp Phys Commun 1982b;27:213–27. Rae CS, Khor IW, Wang Q, Destito G, Gonzalez MJ, Singh P, Thomas DM, Estrada MN, Powell E, Finn MG, Manchester M. Systemic trafficking of plant virus nanoparticles in mice via the oral route. Virology 2005 Dec 20;343(2):224-35. Raja KS, Wang Q, Finn MG. Icosahedral virus particles as polyvalent carbohydrate display platforms. Chembiochem 2003 Dec 5;4(12):1348-51. Rautenschlein S, von Samson-Himmelstjerna G, Haase C. A comparison of immune responses to infection with virulent infectious bursal disease virus (IBDV) between specific-pathogen-free chickens infected at 12 and 28 days of age. Vet Immunol Immunopathol 2007 Feb 15;115(3-4):251-60. Reddy SK, Silim A, Ratcliffe MJ. Biological roles of the major capsid proteins and relationships between the two existing serotypes of infectious bursal disease virus. Arch Virol 1992;127(1-4):209-22. Reiss BD, Mao, C., Solis, D. J., Ryan, K. S., Thomson T. and Belcher, A. M. Biological routes to metal alloy ferromagnetic nanostructures. Nano Lett 2004;4:1127-32. Remond M, Da Costa B, Riffault S, Parida S, Breard E, Lebreton F, Zientara S, Delmas B. Infectious bursal disease subviral particles displaying the foot-and-mouth disease virus major antigenic site. Vaccine 2009 Jan 1;27(1):93-8. Ribot WJ, Powell BS, Ivins BE, Little SF, Johnson WM, Hoover TA, Norris SL, Adamovicz JJ, Friedlander AM, Andrews GP. Comparative vaccine efficacy of different isoforms of recombinant protective antigen against Bacillus anthracis spore challenge in rabbits. Vaccine 2006 Apr 24;24(17):3469-76. Rieger J, Freichels H, Imberty A, Putaux JL, Delair T, Jerome C, Auzely-Velty R. Polyester nanoparticles presenting mannose residues: toward the development of new vaccine delivery systems combining biodegradability and targeting properties. Biomacromolecules 2009 Mar 9;10(3):651-7. Rodgers RE, Chang D, Cai X, Consigli RA. Purification of recombinant budgerigar fledgling disease virus VP1 capsid protein and its ability for in vitro capsid assembly. J Virol 1994 May;68(5):3386-90.

Page 134: Expression and Characterization of Infectious Bursal

Chapter VII References

128

Rodriguez-Lecompte JC, Nino-Fong R, Lopez A, Frederick Markham RJ, Kibenge FS. Infectious bursal disease virus (IBDV) induces apoptosis in chicken B cells. Comp Immunol Microbiol Infect Dis 2005 Jul;28(4):321-37. Rogel A, Benvenisti L, Sela I, Edelbaum O, Tanne E, Shachar Y, Zanberg Y, Gontmakher T, Khayat E, Stram Y. Vaccination with E. coli recombinant empty viral particles of infectious bursal disease virus (IBDV) confer protection. Virus Genes 2003 Oct;27(2):169-75. Rong J, Jiang T, Cheng T, Shen M, Du Y, Li S, Wang S, Xu B, Fan G. Large-scale manufacture and use of recombinant VP2 vaccine against infectious bursal disease in chickens. Vaccine 2007 Nov 14;25(46):7900-8. Roy P, Noad R. Virus-like particles as a vaccine delivery system: myths and facts. Hum Vaccin 2008 Jan-Feb;4(1):5-12. Ruiz-Feria CA, Abdukalykova ST. Arginine and vitamin E improve the antibody responses to infectious bursal disease virus (IBDV) and sheep red blood cells in broiler chickens. Br Poult Sci 2009 May;50(3):291-7. Ruth C, Zuellig T, Mellitzer A, Weis R, Looser V, Kovar K, Glieder A. Variable production windows for porcine trypsinogen employing synthetic inducible promoter variants in Pichia pastoris. Systems and synthetic biology 2010 Sep;4(3):181-91. Sack M, Paetz A, Kunert R, Bomble M, Hesse F, Stiegler G, Fischer R, Katinger H, Stoeger E, Rademacher T. Functional analysis of the broadly neutralizing human anti-HIV-1 antibody 2F5 produced in transgenic BY-2 suspension cultures. FASEB J 2007 Jun;21(8):1655-64. Salunke DM, Caspar DL, Garcea RL. Self-assembly of purified polyomavirus capsid protein VP1. Cell 1986 Sep 12;46(6):895-904. Salyaev RK, Rigano MM, Rekoslavskaya NI. Development of plant-based mucosal vaccines against widespread infectious diseases. Expert Rev Vaccines 2010 Aug;9(8):937-46. Sambrook J, Russell, D. W. Molecular Cloning: A Laboratory Manual (3rd ed.). Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York. 2001. Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci U S A 1977 Dec;74(12):5463-7. Sapsford KE, Soto CM, Blum AS, Chatterji A, Lin T, Johnson JE, Ligler FS, Ratna BR. A cowpea mosaic virus nanoscaffold for multiplexed antibody conjugation: application as an immunoassay tracer. Biosens Bioelectron 2006 Feb 15;21(8):1668-73. Saugar I, Irigoyen N, Luque D, Carrascosa JL, Rodriguez JF, Caston JR. Electrostatic interactions between capsid and scaffolding proteins mediate the structural polymorphism of a double-stranded RNA virus. J Biol Chem 2010 Feb 5;285(6):3643-50. Saugar I, Luque D, Ona A, Rodriguez JF, Carrascosa JL, Trus BL, Caston JR. Structural polymorphism of the major capsid protein of a double-stranded RNA virus: an amphipathic alpha helix as a molecular switch. Structure 2005 Jul;13(7):1007-17.

Page 135: Expression and Characterization of Infectious Bursal

Chapter VII References

129

Schlick TL, Ding Z, Kovacs EW, Francis MB. Dual-surface modification of the tobacco mosaic virus. J Am Chem Soc 2005 Mar 23;127(11):3718-23. Schneider-Ohrum K, Ross TM. Virus-like particles for antigen delivery at mucosal surfaces. Curr Top Microbiol Immunol 2012;354:53-73. Schoen C, Stritzker J, Goebel W, Pilgrim S. Bacteria as DNA vaccine carriers for genetic immunization. Int J Med Microbiol 2004 Oct;294(5):319-35. Sedighzadeh SS, Shamsara M, Shahpiri A. Fusion Protein Strategy to Increase Expression and Solubility of Hypervariable Region of VP2 Protein of Infectious Bursal Disease Virus in Escherichia coli. The Protein J 2012 Oct;31(7):580-4. Shah SN, Steinmetz NF, Aljabali AA, Lomonossoff GP, Evans DJ. Environmentally benign synthesis of virus-templated, monodisperse, iron-platinum nanoparticles. Dalton transactions 2009 Oct 28(40):8479-80. Shaw I, Davison TF. Protection from IBDV-induced bursal damage by a recombinant fowlpox vaccine, fpIBD1, is dependent on the titre of challenge virus and chicken genotype. Vaccine 2000 Aug 1;18(28):3230-41. Singh P, Prasuhn D, Yeh RM, Destito G, Rae CS, Osborn K, Finn MG, Manchester M. Bio-distribution, toxicity and pathology of cowpea mosaic virus nanoparticles in vivo. J Control Release 2007 Jul 16;120(1-2):41-50. Snyder DB, Lana DP, Savage PK, Yancey FS, Mengel SA, Marquardt WW. Differentiation of infectious bursal disease viruses directly from infected tissues with neutralizing monoclonal antibodies: evidence of a major antigenic shift in recent field isolates. Avian Dis 1988 Jul-Sep;32(3):535-9. Soares-Costa A, Silveira RS, Novo MT, Alves MF, Carmona AK, Belasque J, Jr., Henrique-Silva F. Recombinant expression and characterization of a cysteine peptidase from Xanthomonas citri subsp citri. Genet Mol Res 2012 11, 4043-4057. Staats HF, Bradney CP, Gwinn WM, Jackson SS, Sempowski GD, Liao HX, Letvin NL, Haynes BF. Cytokine requirements for induction of systemic and mucosal CTL after nasal immunization. J Immunol 2001 Nov 1;167(9):5386-94. Stadlmann J, Pabst M, Kolarich D, Kunert R, Altmann F. Analysis of immunoglobulin glycosylation by LC-ESI-MS of glycopeptides and oligosaccharides. Proteomics 2008 Jul;8(14):2858-71. Steinmetz NF, Bize A, Findlay KC, Lomonossoff GP, Manchester M, Evans DJ and Prangishvili D. Site-specific and spatially controlled addressability of a new viral nanobuilding block: Sulfolobus islandicus rod-shaped virus 2. Adv Funct Mater 2008;18:3478-86. Steinmetz NF, Evans DJ. Utilisation of plant viruses in bionanotechnology. Org Biomol Chem 2007 Sep 21;5(18):2891-902.

Page 136: Expression and Characterization of Infectious Bursal

Chapter VII References

130

Steinmetz NF, Evans DJ, Lomonossoff GP. Chemical introduction of reactive thiols into a viral nanoscaffold: a method that avoids virus aggregation. Chembiochem 2007 Jul 9;8(10):1131-6. Steinmetz NF, Lomonossoff GP, Evans DJ. Decoration of cowpea mosaic virus with multiple, redox-active, organometallic complexes. Small 2006 Apr;2(4):530-3. Steinmetz NF, Lomonossoff GP, Evans DJ. Cowpea mosaic virus for material fabrication: addressable carboxylate groups on a programmable nanoscaffold. Langmuir 2006 Apr 11;22(8):3488-90. Steinmetz NF, Mertens ME, Taurog RE, Johnson JE, Commandeur U, Fischer R, Manchester M. Potato virus X as a novel platform for potential biomedical applications. Nano Lett 2010 Jan;10(1):305-12. Steinmetz NF, Shah SN, Barclay JE, Rallapalli G, Lomonossoff GP, Evans DJ. Virus-templated silica nanoparticles. Small 2009 Apr;5(7):813-6. Stoger E, Rademacher T, Sack M, Arcalis E, Stadlmann J, Balzer S, Altmann F, Quendler H, Stiegler G, Kunert R, Fischer R. Recombinant antibody 2G12 produced in maize endosperm efficiently neutralizes HIV-1 and contains predominantly single-GlcNAc N-glycans. Plant Biotechnology Journal 2008 Feb;6(2):189-201. Stoute ST, Jackwood DJ, Sommer-Wagner SE, Cooper GL, Anderson ML, Woolcock PR, Bickford AA, Senties-Cue CG, Charlton BR. The diagnosis of very virulent infectious bursal disease in California pullets. Avian Dis 2009 Jun;53(2):321-6. Strable E, Finn MG. Chemical modification of viruses and virus-like particles. Curr Top Microbiol Immunol 2009;327:1-21. Strable E, Prasuhn DE, Jr., Udit AK, Brown S, Link AJ, Ngo JT, Lander G, Quispe J, Potter CS, Carragher B, Tirrell DA, Finn MG. Unnatural amino acid incorporation into virus-like particles. Bioconjug Chem 2008 Apr;19(4):866-75. Streatfield SJ. Mucosal immunization using recombinant plant-based oral vaccines. Methods 2006 Feb;38(2):150-7. Tacken MG, Peeters BP, Thomas AA, Rottier PJ, Boot HJ. Infectious bursal disease virus capsid protein VP3 interacts both with VP1, the RNA-dependent RNA polymerase, and with viral double-stranded RNA. J Virol 2002 Nov;76(22):11301-11. Tacket CO. Plant-based vaccines against diarrheal diseases. Transactions of the American Clinical and Climatological Association 2007;118:79-87. Taghavian O., Mandal MK, Steinmetz NF, Rasche S, Spiegel H, Fischer R, Schillberg S. A potential nanobiotechnology platform based on infectious bursal disease subviral particles. Rsc Adv. 2012;2:1970-1978. Tao Q, Wang X, Bao H, Wu J, Shi L, Li Y, Qiao C, Yakovlevich SA, Mikhaylovna PN, Chen H. Detection and differentiation of four poultry diseases using asymmetric reverse transcription polymerase chain reaction in combination with oligonucleotide microarrays. J Vet Diagn Invest 2009 Sep;21(5):623-32.

Page 137: Expression and Characterization of Infectious Bursal

Chapter VII References

131

Thanavala Y, Mahoney M, Pal S, Scott A, Richter L, Natarajan N, Goodwin P, Arntzen CJ, Mason HS. Immunogenicity in humans of an edible vaccine for hepatitis B. Proc Natl Acad Sci U S A 2005 Mar 1;102(9):3378-82. Thomas TC, McNamee MG. Purification of membrane proteins. Methods Enzymol 1990;182:499-520. Tiwari S, Verma PC, Singh PK, Tuli R. Plants as bioreactors for the production of vaccine antigens. Biotechnology advances 2009 Jul-Aug;27(4):449-67. Tizard I. The avian antibody response. Semin Avian Exot PetMed 2002;11:2-14. Trovato M, Krebs SJ, Haigwood NL, De Berardinis P. Delivery strategies for novel vaccine formulations. World J of virol 2012;1(1):4-10. Tsoka S, Ciniawskyj OC, Thomas OR, Titchener-Hooker NJ, Hoare M. Selective flocculation and precipitation for the improvement of virus-like particle recovery from yeast homogenate. Biotechnol Prog 2000 Jul-Aug;16(4):661-7. Tsukamoto K, Kojima C, Komori Y, Tanimura N, Mase M, Yamaguchi S. Protection of chickens against very virulent infectious bursal disease virus (IBDV) and Marek's disease virus (MDV) with a recombinant MDV expressing IBDV VP2. Virology 1999 May 10;257(2):352-62. Tsukamoto K, Matsumura T, Mase M, Imai K. A highly sensitive, broad-spectrum infectivity assay for infectious bursal disease virus. Avian Dis 1995 Jul-Sep;39(3):575-86. Tsukamoto K, Sato T, Saito S, Tanimura N, Hamazaki N, Mase M, Yamaguchi S. Dual-viral vector approach induced strong and long-lasting protective immunity against very virulent infectious bursal disease virus. Virology 2000 Apr 10;269(2):257-67. Ulmer JB, Valley U, Rappuoli R. Vaccine manufacturing: challenges and solutions. Nat Biotechnol 2006 Nov;24(11):1377-83. Vajdy M, Srivastava I, Polo J, Donnelly J, O'Hagan D, Singh M. Mucosal adjuvants and delivery systems for protein-, DNA- and RNA-based vaccines. Immunol Cell Biol 2004 Dec;82(6):617-27. Vakharia VN, Snyder DB, He J, Edwards GH, Savage PK, Mengel-Whereat SA. Infectious bursal disease virus structural proteins expressed in a baculovirus recombinant confer protection in chickens. J Gen Virol 1993 Jun;74 ( Pt 6):1201-6. van Loon AA, de Haas N, Zeyda I, Mundt E. Alteration of amino acids in VP2 of very virulent infectious bursal disease virus results in tissue culture adaptation and attenuation in chickens. J Gen Virol 2002 Jan;83(Pt 1):121-9. Vaquero C, Sack M, Chandler J, Drossard J, Schuster F, Monecke M, Schillberg S, Fischer R. Transient expression of a tumor-specific single-chain fragment and a chimeric antibody in tobacco leaves. Proc Natl Acad Sci U S A 1999 Sep 28;96(20):11128-33.

Page 138: Expression and Characterization of Infectious Bursal

Chapter VII References

132

Varsani A, Williamson AL, Rose RC, Jaffer M, Rybicki EP. Expression of Human papillomavirus type 16 major capsid protein in transgenic Nicotiana tabacum cv. Xanthi. Arch Virol 2003 Sep;148(9):1771-86. Varsani A, Williamson AL, Stewart D, Rybicki EP. Transient expression of Human papillomavirus type 16 L1 protein in Nicotiana benthamiana using an infectious tobamovirus vector. Virus Res 2006 Sep;120(1-2):91-6. Veremeiko TA, Lebedev LR, Chikaev NA, Il'ichev AA, Karpenko LI. [Humoral immune response of BALB/c mice immunized with chimer HBcAg proteins carrying the epitopes of surface hepatic B virus protein]. Vopr Virusol 2007 Jan-Feb;52(1):40-5. Wang MY, Kuo YY, Lee MS, Doong SR, Ho JY, Lee LH. Self-assembly of the infectious bursal disease virus capsid protein, rVP2, expressed in insect cells and purification of immunogenic chimeric rVP2H particles by immobilized metal-ion affinity chromatography. Biotechnol Bioeng 2000 Jan 5;67(1):104-11. Wang Q, Kaltgrad E, Lin T, Johnson JE, Finn MG. Natural supramolecular building blocks. Wild-type cowpea mosaic virus. Chem Biol 2002 Jul;9(7):805-11. Wang Q, Lin T, Johnson JE, Finn MG. Natural supramolecular building blocks. Cysteine-added mutants of cowpea mosaic virus. Chem Biol 2002 Jul;9(7):813-9. Wang Q, Lin T, Tang L, Johnson JE, Finn MG. Icosahedral virus particles as addressable nanoscale building blocks. Angew Chem Int Ed Engl 2002 Feb 1;41(3):459-62. Wang Q, Raja KS, Janda KD, Lin T, Finn MG. Blue fluorescent antibodies as reporters of steric accessibility in virus conjugates. Bioconjug Chem 2003 Jan-Feb;14(1):38-43. Wang YS, Ouyang W, Liu XJ, He KW, Yu SQ, Zhang HB, Fan HJ, Lu CP. Virus-like particles of hepatitis B virus core protein containing five mimotopes of infectious bursal disease virus (IBDV) protect chickens against IBDV. Vaccine 2012 Mar 9;30(12):2125-30. Werten MW, van den Bosch TJ, Wind RD, Mooibroek H, de Wolf FA. High-yield secretion of recombinant gelatins by Pichia pastoris. Yeast 1999 Aug;15(11):1087-96. Wong C, Sridhara S, Bardwell JC, Jakob U. Heating greatly speeds Coomassie blue staining and destaining. Biotechniques 2000 Mar;28(3):426-8, 30, 32. Wongwisansri S, Promdonkoy P, Matetaviparee P, Roongsawang N, Eurwilaichitr L, Tanapongpipat S. High-level production of thermotolerant beta-xylosidase of Aspergillus sp. BCC125 in Pichia pastoris: Characterization and its application in ethanol production. Bioresour Technol 2012. Wu DY, Ugozzoli L, Pal BK, Qian J, Wallace RB. The effect of temperature and oligonucleotide primer length on the specificity and efficiency of amplification by the polymerase chain reaction. DNA Cell Biol 1991 Apr;10(3):233-8. Wu H, Singh NK, Locy RD, Scissum-Gunn K, Giambrone JJ. Expression of immunogenic VP2 protein of infectious bursal disease virus in Arabidopsis thaliana. Biotechnol Lett 2004 May;26(10):787-92.

Page 139: Expression and Characterization of Infectious Bursal

Chapter VII References

133

Wu J, Yu L, Li L, Hu J, Zhou J, Zhou X. Oral immunization with transgenic rice seeds expressing VP2 protein of infectious bursal disease virus induces protective immune responses in chickens. Plant Biotechnol J 2007 Sep;5(5):570-8. Wu PC, Su HY, Lee LH, Lin DT, Yen PC, Liu HJ. Secreted expression of the VP2 protein of very virulent infectious bursal disease virus in the methylotrophic yeast Pichia pastoris. J Virol Methods 2005 Feb;123(2):221-5. Wu S, Letchworth GJ. High efficiency transformation by electroporation of Pichia pastoris pretreated with lithium acetate and dithiothreitol. Biotechniques 2004 Jan;36(1):152-4. Wu Y, Hong L, Ye J, Huang Z, Zhou J. The VP5 protein of infectious bursal disease virus promotes virion release from infected cells and is not involved in cell death. Arch Virol 2009 Oct 20. Wyeth PJ, Chettle NJ, Mohepat AR. Use of an inactivated infectious bursal disease oil emulsion vaccine in commercial layer chicks. Vet Rec 1992 Jan 11;130(2):30-2. Wyeth PJ, Cullen GA. The use of an inactivated infectious bursal disease oil emulsion vaccine in commercial broiler parent chickens. Vet Rec 1979 Mar 3;104(9):188-93. Xu J, Zhang Z, Yin Y, Cui S, Xu S, Guo Y, Li J, Wang J, Liu X, Han L. Development of reverse-transcription loop-mediated isothermal amplification for the detection of infectious bursal disease virus. J Virol Methods 2009 Jul 28. Yamada T, Iwasaki Y, Tada H, Iwabuki H, Chuah MK, VandenDriessche T, Fukuda H, Kondo A, Ueda M, Seno M, Tanizawa K, Kuroda S. Nanoparticles for the delivery of genes and drugs to human hepatocytes. Nat Biotechnol 2003 Aug;21(8):885-90. Yamaguchi T, Iwata K, Kobayashi M, Ogawa M, Fukushi H, Hirai K. Epitope mapping of capsid proteins VP2 and VP3 of infectious bursal disease virus. Arch Virol 1996;141(8):1493-507. Yang ZQ, Liu QQ, Pan ZM, Yu HX, Jiao XA. Expression of the fusion glycoprotein of Newcastle disease virus in transgenic rice and its immunogenicity in mice. Vaccine 2007 Jan 8;25(4):591-8. Yeh HY, Rautenschlein S, Sharma JM. Protective immunity against infectious bursal disease virus in chickens in the absence of virus-specific antibodies. Vet Immunol Immunopathol 2002 Oct 28;89(3-4):149-58. Yin Y, Xu J, Chen W. [Hepatitis B virus core protein as an epitope vaccine carrier: a review]. Sheng Wu Gong Cheng Xue Bao 2010 Apr;26(4):431-8. Yonekura-Sakakibara K, Saito K. Review: Genetically modified plants for the promotion of human health. Biotechnol Lett 2006 Dec;28(24):1983-91. Yu J, Langridge WH. A plant-based multicomponent vaccine protects mice from enteric diseases. Nat Biotechnol 2001 Jun;19(6):548-52.

Page 140: Expression and Characterization of Infectious Bursal

Chapter VII References

134

Yusibov V, Rabindran S. Recent progress in the development of plant derived vaccines. Expert Rev Vaccines 2008 Oct;7(8):1173-83. Zhang W, Carmichael J, Ferguson J, Inglis S, Ashrafian H, Stanley M. Expression of human papillomavirus type 16 L1 protein in Escherichia coli: denaturation, renaturation, and self-assembly of virus-like particles in vitro. Virology 1998 Apr 10;243(2):423-31. Zhang L, Zhang M, Li J, Cao T, Tian X, Zhou F. Enhancement of mucosal immune responses by intranasal co-delivery of Newcastle disease vaccine plus CpG oligonucleotide in SPF chickens in vivo. Res Vet Sci 2008 Dec;85(3):495-502. Zhang X, Buehner NA, Hutson AM, Estes MK, Mason HS. Tomato is a highly effective vehicle for expression and oral immunization with Norwalk virus capsid protein. Plant Biotechnol J 2006 Jul;4(4):419-32. Zhang X, Zhang X, Yang Q. Effect of compound mucosal immune adjuvant on mucosal and systemic immune responses in chicken orally vaccinated with attenuated Newcastle-disease vaccine. Vaccine 2007 Apr 30;25(17):3254-62. Zhao Q, Chen W, Chen Y, Zhang L, Zhang J, Zhang Z. Self-assembled virus-like particles from rotavirus structural protein VP6 for targeted drug delivery. Bioconjug Chem 2011 Mar 16;22(3):346-52.

Page 141: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

135

VIII Appendices VIII.1 Abbreviations 2, 4-D 2, 4-Dichlorophenoxyacetic acid

Ab Antibody

ABTS 2,2’-Azino-di-(3-ethylbenzthiazoline sulphonate)

Amp Ampicillin

AP Alkaline phosphatase

BCIP 5-bromo-4-chloro-3-indolyl phosphate

bp Baise pair

BSA Bovine serum albumin

CaCl2 Calcium chloride

CaMV Cauliflower mosaic virus

Carb Carbenicillin

cDNA Complementary DNA

cfu Colony-forming units

CTAB Cetyltrimethylammonium bromide

Cys Cysteine

cv. Cultivar

DCs DEPC

Dendritic cells Diethylpyrocarbonate

DLS DMSO

Dynamic light scattering Dimethyl sulfoxide

DNA Deoxyribonucleic acid

dNTP Deoxyribonucleoside triphosphate

DTT 1,4-dithiothreitol

E. coli Escherichia coli EDTA Ethylenediamine tetraacetic acid ELISA Enzyme-linked immunosorbent assay

ER Endoplasmatic reticulum

EtOH Ethanol

Fab Fragment antigen binding

FAE Fc

follicle-associated epithelium Fragment crystalline

GALT gut-associated lymphoid tissue

Page 142: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

136

GMO g

Genetically modified organism Relative centrifugal force (RCF)

GSM (Agrobacteria) glycerol stock media

h Hour(s)

HCl His (H) H6 Ig IMAC kbp kDa KH2PO4

Km LiAc M MES min MgSO4

MnCl2

mRNA ms NaAc NaCl NAGE NaH2PO4

Na2HPO4

NHS OD o/n ORF Ori PBS PBST PCR pH pIgR PVDF Rif RIG-I RNA rpm

Hydrochloride Histidine His-tag Immunoglobulin Immunized metal ion affinity chromatography Kilobase pair Kilodalton Potassium dihydrogen phosphate Kanamycin Lithium acetate Molarity 2-(N-morpholino)-ethanesulphonic acid Minute(s) Magnesium sulfate Manganese(II) chloride Messenger RNA Millisecond Sodium acetate Sodium chloride Native gel electrophoresis Sodium dihydrogen phosphate Disodium monohydrogen phosphate N-hydroxysuccinimide Optical density Overnight Open reading frame Origin of replication Phosphate buffered saline 0.05% (v/v) Tween-20 in PBS Polymerase chain reaction A negative logarithmic measure of hydrogen ion concentration Polymeric Ig receptor Polyvinylidene fluoride Rifampicin retinoic acid-inducible gene I Ribonucleic acid Revolutions per minute

Page 143: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

137

RT SDS-PAGE Sh ble SVP TBE TEM TEMED TGF-β TLR UTR V v/v VLP w/v w/w YNB

Room temperature Sodium dodecyl sulphate-polyacrylamide gel electrophoresis Streptoalloteichus hindustanus bleomycin resistance gene Subviral particle Tris- buffered saline electrophoresis buffer Transmission electron microscopy N,N,N′, N′-tetramethylene-ethylenediamine Transforming growth factor-beta Toll-like receptor untranslated region Volt Volume per volume Virus-like particle Weight per volume Weight per volume Yeast nitrogen base

Page 144: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

138

VIII.2 Schematic presentation of PCR and cloning procedure for generation of tobacco expression vectors

Page 145: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

139

Page 146: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

140

Figure VII.1 Schematic diagrams describing the cloning strategy for IBD-VP2 constructs and the final generated tobacco expression vectors used in this thesis (not drawn to scale). Cloning strategy description: The cDNA of the mature IBD-VP2 sequence (strain IR01, GenBank accession number: AY704912) sequence fused to C-terminal His-tag were amplified using the primers VP2-forward and VP2-reverse-withHis6 containing BspHI and XbaI restriction recognition sites, respectively. Following direct cloning into NcoI and XbaI restriction sites of (A) the vector pTRAkc-GFPapo containing the sequence for the leader sequence LPH of the murine antibody 24 heavy chain (Vaquero et al., 1999) or into (B) the vector pTRAkc-rbcs-cTP containing the Rbcs encoding for the chloroplast targeting signal peptide of Ribulose-1,5-bisphosphate carboxylase oxygenase (RuBisCO) small subunit from Solanum resulting in generation of the final expression vectors pTRAkc-VP2apo and pTRAkc-VP2chlo, respectively. (C) SOE (splicing by overlap extension) PCR was exploited using the CHS-elongation5’ and VP2-reverse-withHis6 primers for elongation and Solulin-for5’UT and VP2-reverse-withHis6 for amplification of the chalcone synthase 5’ UT from Petroselinum fused to the VP2 encoding sequence. The 1.4 kb resultant construct (CHS-VP2) was double digested with EcoRI and XbaI and cloned into the corresponding sites in the pTRAkc-GFPapo vector to generate pTRAkc-VP2cyto. (D) The His-tag was fused to the N-terminal end of VP2 using SOE-PCR using IBD-VP2 cDNA as template and CHS-elongation5’-withHis6 and VP2-reverse for elongation, and Solulin-for5’UT and VP2-reverse for amplification. The cytochrome b5 membrane anchor containing fragment (CytB5-MA) was amplified by PCR using CytB5 cDNA as template and CytB5memAnch.-for and vIL-10_XbaI-bw primer sets. Eventually, the two generated fragments were fused together using the generated overlapping region in a SOE-PCR and the joint sequence was amplified using the primers Solulin-for5’UT and vIL-10_XbaI-bw. Finally, the generated construct was double digested with EcoRI and XbaI and subsequently cloned into the corresponding sites of the pTRAkc-GFPapo vector. The final construct was named pTRAkc-VP2mem. Vector maps description: LB and RB are left and right border of the T-DNA; Pnos and pAnos: promoter and terminator of the nopaline synthase gene, respectively; nptII: coding sequence of the neomycin phosphotransferase gene; bla: coding sequence of the beta-lactamase gene; SAR: scaffold attachment region; P35S and pA35S: 35S promoter with doubled enhancer and terminator of the Cauliflower mosaic virus (CaMV) 35S gene, respectively; CHS5’UT: chalcone synthase 5’ untranslated region from Petroselinum; RK2 ori and ColE1 ori: plasmid origin of replication for Agrobacterium and E. coli respectively; LPH; plant codon optimized leader sequence derived from the murine heavy chain of the TMV-specific mAb24; rbcs-cTP: Rubisco small subunit chloroplast targeting signal; CytB5: cytochrome b5 membrane anchor; IBD-VP2: Codon sequence of the Infectious bursal disease virus VP2 corresponding to amino acids 1-441.

Page 147: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

141

VIII.3 Schematic presentation of PCR and cloning procedure for generation of Pichia expression vectors

Page 148: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

142

Figure VII.2 Schematic diagrams describing the cloning strategy for IBD-VP2 constructs and the final generated Pichia expression vectors used in this thesis (not drawn to scale). Cloning strategy description: IBD-VP2 encoding fragments were excised from the plant expression vectors pTRAkc-VP2apo and pTRAkc-VP2cyto using EcoRI and XbaI and cloned into the same restriction digestion sites of pPICZ_B, Pichia expression vector, resulting in the generation of pPICZ_B-VP2sec and pPICZ_B-VP2cyto, respectively. As a result, VP2 fused to the C-terminal His-tag, was expressed under the control of the methanol-inducible alcohol oxidase 1 (AOX1) promoter and terminator, downstream of the CHS5’UT sequence with (pPICZ_B-VP2sec) and without (pPICZ_B-VP2cyto) the leader sequence LPH of the murine antibody 24 heavy chain (Vaquero et al., 1999). Vector maps description: 5’ AOX1 and AOX TT: alcohol oxidase 1 promoter und terminator, respectively; CHS5’UT: chalcone synthase 5’ untranslated region from Petroselinum; LPH; leader peptide of heavy chain (Murin originated); IBD-VP2: Codon sequence of Infectious bursal disease virus VP2 corresponding to amino acids 1-441 (mature VP2). PTEF1: transcription elongation factor 1 gene promoter from S. cerevisiae that drives expression of the Sh ble gene in Pichia, conferring Zeocin resistance; PEM7: synthetic synthetic prokaryotic promoter that drives expression of Sh ble in E. coli, conferring Zeocin resistance; Sh ble: zeocin gene resistance; Cyc1 TT: cyc1 transcription termination; pUC origin: plasmid origin of replication for E. coli.

Page 149: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

143

VIII.4 Protein sequence alignment of IBD-VP2 IBD-VP2_AF109154 MTNLTDQTQQIVPFIRSLLMPTTGPASIPDDTLEKHTLRSETSTYNLTVGDTGSGLIVFF 60 IBD-VP2_AY704912 MTNLQDQTQQIVPFIRSLLMPTTGPASIPDDTLEKHTLRSETSTYNLTVGDTGSGLIVFF 60 **** ******************************************************* IBD-VP2_AF109154 PGFPGSIVGAHYTLQSNGNYKFDQMLLTAQNLPASYNYCRLVSRSLTVRSSTLPGGVYAL 120 IBD-VP2_AY704912 PGFPGSIVGAHYTLQSNGNYKFDQMLLTAQNLPASYNYCRLVSRSLTVRSSTLPGGVYAL 120 ************************************************************ IBD-VP2_AF109154 NGTINAVTFQGSLSDLTDVSYNGLMSATANINDKIGNVLVGEGVTVLSLPTSYDLGYVRL 180 IBD-VP2_AY704912 NGTINAVTFQGSLSELTDVSYNGLMSATANINDKIGNVLVGEGVTVLSLPTSYDLGYVRL 180 ************** ********************************************* IBD-VP2_AF109154 GDPIPAIGLDPKMVATCDSSDRPRVYTITAADDYQFSSQYQSGGVTITLFSANIDAITSL 240 IBD-VP2_AY704912 GDPIPAIGLDPKMVATCDSSDRPRVYTITAADDYQFSSQYQAGGVTITLFSANIDAITSL 240 ***************************************** ****************** IBD-VP2_AF109154 SIGGELVFHTSVHGLALDATIYLIGFDGTTVITRAVASDNGLTTGIDNLMPFNLVIPTNE 300 IBD-VP2_AY704912 SIGGELVFQTSVQGLILGATIYLIGFDGTAVITRAVAADNGLTAGTDNLMPFNIVVPTSE 300 ******** *** ** * *********** ******* ***** * ******* * ** * IBD-VP2_AF109154 ITQPITSIKLEIVTSKSGGQAGDQMSWSAMWSLAVTIHGGNYPGALRPVTLVAYERVATG 360 IBD-VP2_AY704912 ITQPITSIKLEIVTSKSGGQAGDQMSWSASGSLAVTIHGGNYPGALRPVTLVAYERVAKG 360 ***************************** *************************** * IBD-VP2_AF109154 SVVTVAGVSNFELIPNPELAKNLVTEYGRFDPGAMNYTKLILSERERLGIKTVWPTREYT 420 IBD-VP2_AY704912 SVVTVAGVSNFELIPNPELAKNLVTEYGRFDPGAMNYTKLILSERDRLGIKTVWPTREYT 420 ********************************************* ************** IBD-VP2_AF109154 DFREYFMEVADLNSPLKIAGA 441 IBD-VP2_AY704912 DFREYFMEVADLNSPLKIAGA 441 *********************

Figure VII.3 Protein sequence alignment of IBD-VP2 sequence used in this thesis (AY704912) and the one used by Doong et al. (2007) (AF109154). The alignment was performed using Clustal 2.1 algorithm. “*” = the residues in that column are identical in two sequences in the alignment. His249 and His253 (in red) which were replaced by Q in AY704912, facilitate purification by IMAC for generated IBD-SVP.

Page 150: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

144

VIII.5 Monitoring of IBDV clinical signs among the vaccinated chickens after viral challenge.

Table VII.1 Observations and the intensity of the clinical signs of infectious bursal disease were recorded during the time-course before euthanization. In case of no specific observation, the box was left empty.

Group Vaccine Observation at the first dpc 1 250 mg Pichia yVP2cyto 2 250 mg Pichia yVP2cyto+ Oral adjuvant

3 25 mg Pichia yVP2cyto 4 25 mg Pichia yVP2cyto+ Oral adjuvant

5 50 µg IBD-SVPcyto few chickens with greyish-greenish but well formed feces

6 50 µg IBD-SVPcyto+ Oral adjuvant few chickens with greyish-greenish but well formed feces

7 500 µg IBD-SVPcyto 8 500 µg IBD-SVPcyto+ Oral adjuvant 9 250 mg lyophilized inactivated wild type Pichia 10 20 µg of Purified IBD-SVPcyto+ Adjuvant 100 (i.m) 11 Challenge control 12 AviPro Gumboro vac. (Oral control) 13 Vaccine control (without challenge)

Group Observations at the second dpc

1 many chickens with slightly reduced clinical signs 2 many chickens with slightly reduced clinical signs 3 many chickens with slightly reduced clinical signs 4 many chickens with slightly reduced clinical signs 5 many chickens with slightly reduced clinical signs 6 many chickens with slightly reduced clinical signs 7 many chickens with slightly reduced clinical signs 8 many chickens with slightly reduced clinical signs

9 many chickens with moderately reduced clinical signs, 1 chicken with severely reduced clinical signs

10 11 many chickens with moderately reduced clinical signs 12

13

Group Observations at the third dpc 1 all chickens with slight to moderate reduced clinical signs 2 all chickens with slight to moderate reduced clinical signs 3 all chickens with slight to moderate reduced clinical signs 4 all chickens with slight to moderate reduced clinical signs 5 2 chickens died, some chickens with severely reduced clinical signs

Page 151: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

145

6 1 chicken died, some chickens with severely reduced clinical signs 7

8

9 3 chickens died, 1 chicken with moderate to severe and 1 chicken with slightly reduced clinical condition

10

11 1 chicken died, 2 chickens with moderately to severely and 1 chicken with slightly reduced clinical condition

12 13

Group Observations at the forth dpc

1 2 chickens died 2 1 chicken with slightly reduced clinical condition 3

4 1 chicken died 5 1 chicken with slightly and 1 chicken with severely reduced clinical condition 6 2 chickens died 7 1 chicken with slightly reduced clinical condition 8 1 chicken with slightly reduced clinical condition 9 1 chicken with slightly reduced clinical condition 10

11

1 chicken died, 1 chicken with moderately and 1 chicken with slightly reduced clinical condition

12 13

Group Observations at the fifth dpc Observations at the sixth dpc

1 2

3

1 chicken with reduced clinical condition 4

1 chicken with reduced clinical condition

5

1 chicken with reduced clinical condition 6

1 chicken with reduced clinical condition

7 8 9 10 11 1 chicken died , 1 chicken with slightly reduced clinical condition

1 chicken with slightly reduced clinical condition

12 13

Page 152: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

146

VIII.6 Serological VP2 antibody titers

Table VII.2 Serological VP2 antibody titer in immunized chickens after challenge with IBDV measured by the IDEXX IBD-XR kit, protection status, virus detection in the bursa, histopathological damage evaluation and the morphological observation of the bursa fabricious damage score extracted from chickens 7 dpc. Chickens with IDEXX index value over 396 are resistant to IBD (Rong et al., 2007). D= detected, ND= not detected. (-): no specific observation.

Gro

up

Bird

No.

anti-VP2 titer

measured by

IDEXX IBD-XR Ab test

kit

Protection

Detection of IBDV

in the bursa

Bursa lesion score

Bursa damage

1 1 8488 D 1 - 1 2 10132

D 2 -

1 3 18 died D 2 mildly swollen, edema around bursa, hemorrhages in bursa

1 4 2400

ND 1 - 1 5 0 died D 2 mildly swollen 2 6 2079

D 1 -

2 7 5866

D 1 - 2 8 8782

D 1 -

2 9 2166

D 1 - 2 10 2327

D 1 -

3 11 11480

D 1 - 3 12 2327

D 1 -

3 13 9669

D 2 accumulated pus 3 14 5408

D 1 -

3 15 13078

D 2 accumulated pus, hemorrhages 4 16 6954

D 1 -

4 17 201 died D 2 mildly swollen 4 18 10147

D 1 -

4 19 4495

ND 1 - 4 20 11034

D 1 -

5 21 57 died D 3 severely swollen, edema around bursa, cherry red

5 22 448

ND 0 - 5 23 7985

D 3 -

5 24 11226

D 2 - 5 25 127 died D 3 severely swollen, cherry red 6 26 13905

D 2 -

6 27 252 died D 2 moderately swollen, edema around bursa, hemorrhages in bursa

6 28 0 died D 3 moderately swollen, cherry red 6 29 8342

D 1 -

6 30 0 died D 3 moderately swollen, hemorrhages in bursa 7 31 7434

D 1 -

Page 153: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

147

Gro

up

Bird

No.

anti-VP2 titer

measured by

IDEXX IBD-XR Ab test

kit

Protection

Detection of IBDV

in the bursa

Bursa lesion score

Bursa damage

7 32 10704

D 2 - 7 33 7400

D 2 accumulated pus

7 34 7842

D 1 - 7 35 4105

D 1 -

8 36 8953

D 1 - 8 37 1549

ND 0 -

8 38 9298

D 3 accumulated pus, hemorrhages 8 39 6827

ND 1 -

8 40 6657

ND 0 - 9 41 253 died D 0 edema around bursa 9 42 0 died D 3 moderately swollen, cherry red 9 43 6776

ND 1 -

9 44 13204

D 1 - 9 45 0 died D 3 severely swollen, cherry red

10 46 2246

ND 0 - 10 47 1875

ND 0 -

10 48 1533

ND 0 - 10 49 681

ND 0 -

10 50 4895

ND 0 - 11 51 172 died D 3 - 11 52 9914

ND 1 -

11 53 0 died D 3 mildly swollen 11 54 66 died D 3 mildly swollen, hemorrhages 11 55 11250

D 1 -

12 56 4522

ND 0 - 12 57 1207

ND 0 -

12 58 4883

ND 0 - 12 59 2587

ND 0 -

12 60 10657

ND 0 - 13 61

ND 0 -

13 62

ND 0 - 13 63

ND 0 -

13 64

ND 0 - 13 65 ND 0 -

Page 154: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

148

VIII.7 Figure legends Figure I.1 The schematic feature of mucosal immune system in the gut.

Figure I.2 Schematic configuration of IBDV and its genome.

Figure I.3 Three VP2 proteins assemble together and form a trimer, the structural unit of the virus.

Figure I.4 Hydrophobicity plot of IBD-VP2 provided by CLC Main workbench 6.5 using Kyte-Doolittle hydrophobicity scale.

Figure I.5 (A) Protein structure of VP2; (B) The trimeric VP2 structure from the top view

Figure I.6 Schematic overview of the work presented in this dissertation.

Figure II.1 Weight development in specific pathogen free (SPF) chickens.

Figure III.1 Schematic drawing of the generated pTRAkc plant expression vectors for IBD-VP2 expression in tobacco.

Figure III.2 Comparison of VP2 accumulation in various cell compartments using immunoblot.

Figure III.3 Determination of VP2 content accumulated in different cell compartments of transiently transformed N. tabacum cv. Petite Havana SR1 leaves.

Figure III.4 SDS-PAGE analysis of VP2 purified by IMAC form transiently transformed tobacco leaves using pTRAkc-VP2apo and pTRAkc-VP2cyto.

Figure III.5 SDS-PAGE analysis to check the VP2 purity.

Figure III.6 Prediction of potential N-glycosylation sites of IBD-VP2 protein predicted by the NetNGlyc 1.0 server (http://www.cbs.dtu.dk/services/NetNGlyc/).

Figure III.7 SDS-PAGE for glycosylation analysis of the tobacco-produced VP2 followed by (A) Coomassie brilliant blue staining and (B) glycan detection by immunoblot.

Figure III.8 SDS-PAGE analysis of the deglycosylated tobacco-produced affinity-purified VP2apo using peptide N-Glycosidase F (PGNase F).

Figure III.9 Liquid chromatography-mass spectrometry (LC-MS) of the three N-glycosylation sites of plant-produced and affinity-purified VP2apo.

Figure III.10 Chromatographic profile obtained by separation of purified (III.1.4) VP2cyto (red) and VP2apo (blue) in two separate runs using a Superdex 200 column.

Figure III.11 ELISA analysis for the VP2 content of the tobacco-produced VP2cyto separated by sucrose gradient ultracentrifuge (10-60%).

Figure III.12 Chromatographic profile obtained by separation of VP2cyto before (blue) and after incubation with urea (brown) in two separate runs using a Superdex 200 column.

Figure III.13 Schematic diagram shows mice immunization and blood sampling plan.

Figure III.14 Determination of polyclonal IgG antibody titers in the sera of five mice immunized with (A) VP2apo or (B) VP2cyto, respectively, by indirect ELISA.

Figure III.15 Schematic presentation of the final yeast pPICZ_B based expression vectors. Figure III.16 Dotblot analyses for screening of yVP2sec or yVP2cyto in producing P. pastoris cultures. Figure III.17 Time-course study for production level evaluation of Pichia-produced (A) yVP2sec and

(B) yVP2cyto accumulated in the medium and inside the cell, respectively.

Figure III.18 SDS-PAGE analysis of yVP2sec and yVP2cyto obtained from selected Pichia colonies (III.2.2) after 96 h expression induction, followed by (A and C) Coomassie brilliant blue staining and (B and D) immunoblot.

Figure III.19 First step purification of yVP2cyto using pH-shift from 6 to 4.

Page 155: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

149

Figure III.20 Separation of Pichia-produced IBD-SVPcyto from not-fully assembled VP2 using size exclusion chromatography and further electron microscopy studies.

Figure VIII.21 Analyses the purity of the purified yVP2. Figure III.22 Lectin blot analysis of yVP2 produced in Pichia. Figure III.23 Analysis of deglycosylated VP2 using peptide N-glycosidase F (PGNase F) and lection

blot.

Figure III.24 Liquid chromatography-mass spectrometry (LC-MS) of the three N-glycosylation sites of Pichia-produced and affinity-purified yVP2sec-m.

Figure III.25 Analysis of stability and integrity for IBD-SVPcyto after Pichia cell inactivation using SDS-PAGE and SEC.

Figure III.26 Immunization of the chickens randomly allocated between 13 immunization groups.

Figure III.27 Weight of the chickens was monitored as a health index during immunization.

Figure III.28 Serum IgA and intestinal IgA response in chickens immunized with freeze-dried inactivated cell-capsulated and purified IBD-SVPcyto after viral challenge.

Figure III.29 Immunization of chickens with freeze-dried inactivated cell-capsulated and purified IBD-SVPcyto

Figure III.30 Stability of IBD-SVPcyto in ethanol–water and DMSO–water mixtures determined by NAGE (A), A280 spectroscopy (B), DLS (C) and TEM (D).

Figure III.31 Stability of IBD-SVPcyto at different temperatures determined by NAGE (A), A280 spectroscopy (B), DLS (C) and TEM (D).

Figure III.32 Stability of IBD-SVPcyto in different pH environments determined by NAGE (A), A280 spectroscopy (B), DLS (C) and TEM (D).

Figure III.33 Structural analysis of IBD-SVPcyto. Figure III.34 Labeling of amine groups on the surface of IBD-SVPcyto using biotinamidohexanoic acid

N-hydroxysuccinimide ester. Figure III.35 Biochemical analysis of biotinylated IBD-SVPcyto by western blot (A), SEC (B), TEM

after amine conjugation (C) and TEM after carboxyl conjugation (D). Figure III.36 Labeling carboxyl groups on the surface of IBD-SVP.

Figure VII.1 Schematic diagrams describing the cloning strategy for IBD-VP2 constructs and the final generated tobacco expression vectors used in this thesis.

Figure VII.2 Schematic diagrams describing the cloning strategy for IBD-VP2 constructs and the final generated Pichia expression vectors used in this thesis.

Figure VII.3 Protein sequence alignment of IBD-VP2 sequence used in this thesis (AY704912) and the one used by Doong et al. (2007) (AF109154).

Page 156: Expression and Characterization of Infectious Bursal

Chapter VII Appendices

150

VIII.8 Tables Table I.1 Recombinant expression systems that have been used for production of subunit vaccines

for chickens against IBDV by expression of IBD-VP2. Table VIII.1 Primers used for generation of DNA fragments by PCR. Table VIII.2 SOE-PCR primers for introducing extra DNA fragments into constructs or joining two

separate constructs. Table VIII.3 Primers used for DNA sequencing. Table II.4 Phosphorothioate CpG ODN used as adjuvant in oral immunization of chickens. Table VIII.5 Names, suppliers and genotypes of E. coli strains used throughout the work. Table VIII.6 PCR master mix. Table II.7 Experimental design for immunization of chickens. Table III.1 Glycan compositions and their ratio in the three N-glycan sites (1, 2 and 3) of plant-

produced and affinity-purified VP2 by liquid chromatography-mass spectrometry (LC-MS).

Table III.2 Glycan composition and their ratio at the three N-glycan sites (1, 2 and 3) of purified yVP2sec using IMAC form the medium (III.2.3) studied by liquid chromatography-mass spectrometry (LC-MS).

Table III.4 The frequency of IgM, IgY and IDEXX IBD seropositive chickens before and after viral challenge and the protection rate.

Table III.5 The virus clearance ability by the immune system of the immunized birds was evaluated 7 dpc.

Table VII.1 Observation and the intensity of the clinical signs of infectious bursal disease were recorded during the time-course before euthanization.

Table V VIII.2 Serological VP2 antibody titer in immunized chickens after challenge with IBDV measured by the IDEXX IBD-XR kit, protection status, virus detection in the bursa, histopathological damage evaluation and the morphological observation of the bursa fabricious damage score extracted from chickens 7 dpc.

Page 157: Expression and Characterization of Infectious Bursal

151

Acknowledgement It is my great pleasure to thank to the people who contributed directly and indirectly to the completion of this dissertation. First I would like to express my sincere gratitude towards Prof. Dr. Rainer Fischer for providing the opportunity to start and complete my thesis at the Institute for Molecular Biotechnology at the RWTH Aachen University. It has been an honor to be his PhD student. I am also thankful to him for revising the thesis and as my first examiner. Besides, I would like to thank the rest of my thesis committee: Prof. Dr. Stefan Schillberg, Head of the Plant Biotechnology Department, Fraunhofer Institute for Molecular Biology and Applied Ecology in Aachen and Dr. Nikolaus Schleich, Botanical Institute, RWTH Aachen University for reviewing the thesis and agreeing to be the co-examiner. I would like to acknowledge the German academic exchange service (DAAD) for their financial and spiritual support from the moment I arrived to Germany to the end of the project. I am extremely grateful to Prof. Dr. Stefan Schillberg, for supporting me to complete this thesis. I am also thankful for his precise project management, constructive discussions and correction of this thesis and my manuscripts. I am also very thankful to Holger Spiegel for his supervision, valuable information, advice, ideas and constant support throughout my entire doctoral work. I am also thankful to Dr. Grit Hehmann for her supervision and nice consults. I am thankful to Dr. Rüdiger Hauck from institute of Poultry Diseases, faculty of Veterinary Medicine, Freie Universität Berlin for his help and support in chicken immunization studies. I am also thankful to Dr. Nicole Steinmetz, department of Biomedical Engineering, School of Medicine, Case Western Reserve University, USA for her help and support in bioconjugation studies. Many thanks to Dr. Flora Schuster and Dr. Thomas Rademacher for their great care on generation and maintenance of stably-transformed transgenic plants. I also thank Yvonne Olbrich and Katrin Hauten for their help in establishment and maintenance of hybridoma cultures. I am also grateful to Dr. Manoj K. Mandal, Dr. Stefan Rasche, Alexander Boes and Dr. Rajan Sriraman for their discussions and consults. Thank to Dr. Nicole Raven, Dr. Greta Nölke, Dr. Nikolay Vasilev, Dr. Helga Schinkel and Dr. Andreas Schiermeyer for their guidance in lab work. I am especially thankful to my very close friend, Dara Forouzan for his admirable friendship as well as his nice and useful discussions. During the project I had a chance to experience team work in international atmosphere. I found many friends and had a great time with them. I would like to give my deep devotion to them in Fraunhofer IME and bio7 and especially to Dr. Manoj K. Mandal, Houtan Ahvari and Dr. Reza Safarnejad and wish them all the best for their scientific and private life. To all members of the plant biotechnology group and the Department of bio7, I am very grateful for their friendship, for their guidance, kindness and cooperation. This dissertation is dedicated to my adorable parents, Sousan Sadeghzadeh and Rahim Taghavian, my wonderful wife, Dr. Farzaneh Taslimi and to my lovely brothers, Afshin, Armin and Aidin, for their endless love, support and encouragements.

Page 158: Expression and Characterization of Infectious Bursal

152

CV

Personal data Name

Omid Taghavian

Date of Birth 27th Aug, 1981 Nationality Iranian Birth place Tehran, Iran Sex Male Marital Status Married Address Department of Plant Biotechnology Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074 Aachen, Germany E-Mail [email protected] [email protected]

Research experience 02/2011-Now Position: Research fellow Institute: Fraunhofer IME, Aachen, Germany. Supervisor: Prof. Dr. Rainer Fischer and Prof. Dr. Stefan Schillberg Project: Development of Subunit Vaccine against Infectious Salmon Anemia Virus (ISAV) 10/2006 – 10/2010 Position: Graduate (PhD) student (DAAD scholarship holder) Institute: Fraunhofer IME, Aachen, Germany Supervisor: Prof. Dr. Rainer Fischer and Prof. Dr. Stefan Schillberg Project: Expression and characterization of Infectious bursal disease virus

(IBDV) Protein in transgenic Plant and Yeast for poultry vaccine Development and Application in Nanotechnology

09/2003 – 09/2005 Position: Master student

Institute: National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran

Supervisor: Dr. Amir Mousavi and Dr. A. A. Sh. Boushehri Project: Assessment of the Function of Plastid Transit Peptides in Expression of Reporter Genes in Transgenic Potato Plants

09/2003 – 09/1999 Position: Undergraduate (Bachelor) student of Agricultural Engineering- Agronomy and Plant Breeding Institute: Faculty of Agriculture; University of Tehran, Iran Supervisor: Prof. Dr. Mansour Omidi Project: Cell Differentiation

Page 159: Expression and Characterization of Infectious Bursal

153

Educational background 2003 – 2005 Institute: Faculty of Agriculture; University of Tehran, Iran Degree: Master of Science, GPA: 17.66/20 Subject: Agricultural Biotechnology Supervisor: Dr. Amir Mousavi and Dr. A. A. Sh. Boushehri 1999 – 2003 Institute: Faculty of Agriculture; University of Tehran, Iran Degree: Bachelor, GPA: 15.66/20 Subject: Agricultural Engineering-Agronomy and Plant Breeding Supervisor: Prof. Dr. Mansour Omidi 1998 – 1999 Institute: Soroush Pre-University School Degree: Pre-University Degree (Life Science) 1996 – 1998 Institute: Soroush High School Degree: Diplom (Life Science), Grade: 18.33/20. Publication list • Omid Taghavian, Holger Spiegel, Rüdiger Hauck, Hafez M. Hafez, Rainer Fischer, Stefan

Schillberg; Protective oral vaccination against Infectious Bursal Disease Virus using the major viral antigenic Protein produced in Pichia pastoris (accepted to be published in PLoS ONE).

• Javier Alcaíno, Omid Taghavian, Manoj K. Mandal, Holger Spiegel, Rainer Fischer, Wolfgang Schuch, Martin Hevia, Stefan Schillberg, Mauricio Rios-Momberg; Nicotiana tabacum cv. Bright Yellow-2 (BY-2) and Pichia pastoris are safe platforms for production of oral vaccine for Atlantic Salmon (Salmo salar L.) (submitted to Aquaculture research).

• Omid Taghavian, Amir Mousavi, Haleh Hashemi Sohi, Esmat Jourabchi, Kasra Esfahani; Functional assessment of plastid signal peptide sequences LIM14 and AtCPrecA in localization of green fluorescent protein (GFP) and Beta-glucuronidase (GUS) reporter proteins in transgenic potato plants (in Farsi), IJPB, Vol. 5, No. 17 (article in press).

• Omid Taghavian, Manoj K. Mandal, Nicole F. Steinmetz, Stefan Rasche, Holger Spiegel,

Rainer Fischer, Stefan Schillberg; (2012), A potential nanobiotechnology platform based on infectious bursal disease subviral particles, RSC advances, DOI: 10.1039/c2ra00857b.

• Mansour Omidi, Reza Maali Amiri, Omid Taghavian, Masoud Akhshik; (2011), Cytology (in Farsi). University of Tehran Press. Tehran. Iran.

• Omid Taghavian; (2005), Yeast two hybrid system: An interacting protein entrapment technique, Modern Genetics (in Farsi), No.2, spring 2005

Page 160: Expression and Characterization of Infectious Bursal

154

Academic honors and awards

2006 – DAAD scholarship for full PhD course (2006-2010), Germany. Presentations

• Omid Taghavian, Holger Spiegel, Rainer Fischer, Stefan Schillberg; Infectious bursal

disease subviral particles as an efficient vaccine for chickens; Poster presentation in Virus-like particle and Nano-particle vaccines meeting; Cannes, France; 27-30 November, 2012

• Omid Taghavian, Manoj K. Mandal, Rainer Fischer, Stefan Schillberg; IBDV sub-viral particles as a platform for nano-biomedical applications; Poster presentation in Biomedica conference; Aachen, Germany; 17-18 March 2010.

• Omid Taghavian; Expression of Infectious bursal disease virus capsid protein in tobacco leaves in order to produce an edible vaccine for chickens; Oral presentation; First biology conference for Iranian scholars’ in Europe; Hamburg, Germany; 6-7th December, 2008.

• Omid Taghavian, Amir Mousavi, Hale Hashemi Sohi, Esmat Jourabchi; Assessment of the Function of Two Plastid Signal Peptides in Localization of Reporter Proteins in Transgenic Potato Plants; International Conference on Plant Transformation Technologies; Vienna – Austria; February 4-7, 2007.

• Dara Forouzan Boroojeni, Haleh Hashemi Sohi, Mansoor Omidi, Esmat Jourabchi, Neda

Naghibzadeh Tabatabaee, Omid Taghavian; Expression of human growth hormone in potato; Oral Presentation; 9th National Iranian Genetics Congress; Tehran, Iran; 20 – 22 May 2006.

• Omid Taghavian; Amir Mousavi; Haleh Hashemi Sohi; Esmat Jourabchi; Dara Forouzan

Boroojeni; Ali Akbar Boushehri; Functional Assessment of Plastid Signal Peptides in Expression of Reporter Genes in Transgenic Potato Plants; Oral Presentation; Proceeding of 4th Iranian National Biotechnology Congress; Kerman, Iran; 15 – 17th August 2005.