PhD Fusarium

217
SELECTION, CHARACTERIZATION, AND APPLICATION OF A MYCOTOXIN-SPECIFIC SINGLE DOMAIN ANTIBODY FRAGMENT. A Thesis Presented to The Faculty of Graduate Studies of The University of Guelph by PATRICK JOHN DOYLE In partial fulfillment of the requirements for the degree of Doctor of Philosophy December, 2009 © Patrick Doyle, 2009

description

Fusarium

Transcript of PhD Fusarium

Page 1: PhD Fusarium

i

SELECTION, CHARACTERIZATION, AND APPLICATION OF A

MYCOTOXIN-SPECIFIC SINGLE DOMAIN ANTIBODY FRAGMENT.

A Thesis

Presented to

The Faculty of Graduate Studies

of

The University of Guelph

by

PATRICK JOHN DOYLE

In partial fulfillment of the requirements

for the degree of

Doctor of Philosophy

December, 2009

© Patrick Doyle, 2009

Page 2: PhD Fusarium

i

ABSTRACT

SELECTION, CHARACTERIZATION, AND APPLICATION OF A

MYCOTOXIN-SPECIFIC SINGLE DOMAIN ANTIBODY FRAGMENT.

Patrick John Doyle Advisor:

University of Guelph, 2009 Professor J.C. Hall

15-Acetyl-deoxynivalenol (15-AcDON) is a low molecular weight (M.W. = 338

Da) sesquiterpenoid trichothecene mycotoxin associated with Fusarium Head Blight

(FHB), a global disease of small grain cereals. Estimated economic losses from the most

recent Canadian and US FHB epidemic during the mid 1990s range from $1.3 to $3.0

billion USD, depending on valuations assigned to reduced crop yield and mycotoxin

contamination of food and feed. The accumulation of DON and related trichothecene

mycotoxins within harvested grain is subject to stringent regulation as both toxins pose

dietary health risks to humans and animals. A property of 15-AcDON, and structurally-

similar trichothecene mycotoxins that are intimately involved in FHB pathogenesis, is the

propensity to inhibit peptidyl transferase activity and thus inhibit protein synthesis within

infected host-plant cells. This effect, coupled with secondary phytotoxic effects, act as

mechanisms of virulence for pathogenic Fusarium species.

A single-domain variable heavy chain (VHH) recombinant antibody (rAb) fragment

specific to 15-AcDON was isolated from a hyper-immunized phagemid library. The

dominant clone (NAT-267) was expressed and purified as a VHH monomer with a

competitive binding affinity of 1.24 µM. The gene encoding the NAT-267 camelid

single-domain antibody fragment (VHH) was expressed within the methylotropic yeast

Page 3: PhD Fusarium

ii

Pichia pastoris. Mycotoxin-mediated cytotoxicity was assessed by continuous

measurement of cellular growth over time. At equivalent doses, 15-AcDON was

significantly more toxic to wild-type P. pastoris than was DON, which, in turn, was more

toxic than 3-AcDON. Intracellular expression of this toxin-specific VHH „intrabody‟

within P. pastoris conveyed significant (p = 0.01) resistance to 15-AcDON cytotoxicity at

doses ranging from 20 to 100 µg·mL-1

, making this the first report of VHH-based

sequestration of a haptenic mycotoxin. Furthermore, a biochemical transformation of

DON to 15-AcDON was documented, which explained significant attenuation of the

efficacy of DON at 100 and 200 µg·mL-1

. This “proof of concept” model suggests that in

planta VHH expression may lead to enhanced tolerance to mycotoxins and thereby serve

as a novel tool to help limit Fusarium infection of commercial agricultural crops such as

wheat and barley that are engineered to express this mycotoxin-specific VHH.

Page 4: PhD Fusarium

i

PREFACE

This dissertation contains three manuscripts prepared for publication within a peer-

reviewed text book and two scientific journals. Each manuscript is listed below with the

respective publication status.

CHAPTER TWO

Doyle, P.J., Arbabi-Ghahroudi, M., Sheedy, C., Yau, K., Hall, J.C. 2008. Antibody

Engineering in Agricultural Biotechnology. Chapter 3. In: Immunoassays in

Agricultural Biotechnology. G. Shan (ed.). John Wiley & Sons, New York, NY. (In

Press).

CHAPTER FOUR

Doyle, P.J., Arbabi-Ghahroudi, M., Gaudette, N., Furzer, G., Savard, M.E., Gleddie, S.,

McLean, M.D., MacKenzie, C.R., Hall, J.C. 2008. Cloning, Expression, and

Characterization of a Single Domain Antibody Fragment with Affinity for 15-Acetyl-

Deoxynivalenol. Molec. Imm. 45: 3703 - 3713.

CHAPTER FIVE

Doyle, P.J., Saeed, H., Hermans, A., Gleddie, S.C., Hussack, G., Arbabi-Ghahroudi, M.,

Seguin, C., Savard, M.E., MacKenzie, C.R., Hall, J.C. 2009. Intracellular Expression

of a Single-Domain Antibody Reduces Cytotoxicity of 15-Acetyl-Deoxynivalenol in

Yeast. J. Biol. Chem. 284: 35029-35039. Manuscript M109.045047.

http://www.jbc.org/content/early/2009/09/25/jbc.M109.045047

Page 5: PhD Fusarium

ii

ACKNOWLEDGEMENTS

I would like to thank my graduate committee for their trust, wisdom, and patience in

helping to guide me through this degree. Chris, Marc, Roger, and Steve… it has been a

privilege to learn from you and work in your labs. Thank-you for your friendship and

help along the journey! Also, in alphabetical order… special regards and sincere thanks

to: Anne Hermans, Barb Blackwell, Charles Seguin, Christian Luebbert, Claudia Sheedy,

Garth Gourlay, Ginette Dubuc, Gord Furzer, Greg Hussack, Hanaa Saeed, Hong Tong-

Sevinc, Jamshid Tanha, Joy Roberts, Karen Gourlay, Keith Solomon, Larry Erickson,

Linda Veldhuis, Mehdi Arbabi-Ghahroudi, Mike McLean, Nathalie Gaudette, Nina

Weisser, “Osama-Bin Llama”, Shannon Ryan, Tomoko Hirama, and Yonghong Guan.

Sincere gratitude and thanks to my friends at Syngenta Crop Protection Canada Inc.

for the tremendous support and encouragement. Special thanks to Marian Stypa. You

believed in me and were always there for me (and my family) right from the start! To

Norm Dreger, thanks for adopting me into the “Business Development” family and for

the huge… patience, support, and of course for the valuable insights that you have taught

me about people. I would also like to thank Ms. Laurie Vescio and Mr. Jay Bradshaw for

the great support and words of kind encouragement over these six + years.

Finally, to my family. Thank-you so much… Nadine, you held our family together

all those times that I was away. Even today, I never worry about the most important stuff

because I always know that you have it covered. Mom and Dad… thanks for all the

advice and insights provided during all those late-night phone calls to Calgary. Finally, to

my kids… Liam and Danielle, I have watched you grow alongside this PhD project. I am

very sad that doing this took a lot of my time away from you. Danielle and Liam, you

always were, and will always be in my heart. All those times that I was working

on”school stuff”, you were always the light and my joy that kept me going forward. I

love you both with all my heart...!

Page 6: PhD Fusarium

iii

PICTURE

Freelton, Ontario. April 17, 2005.

Left to Right: “Osama-Bin” Llama; Danielle (hiding), Liam, and Nadine

Page 7: PhD Fusarium

iv

NOTE ABOUT WHY

In the preface of: Fusarium Head Blight of Wheat and Barley, the editors (Kurt J.

Leonard and William R. Bushnell) make the following dedication.

“This book is dedicated to those scientists who labor to solve the Fusarium head

blight problem, despite knowing full well that easier paths may be taken in the pursuit

of peer recognition and scientific acclaim.”

I do not believe that this dedication was a challenge (or „snub‟) to the challenges,

relevance, or prestige of the vast number of other disciplines and technical pursuits within

modern science. To me, Drs. Leonard and Bushnell were simply trying to acknowledge

what “bad actors” FHB and its toxin problem(s): i.e., DON... + acetylated “brother‟s”,

15-AcDON and 3AcDON can be.

For the record, my interest in FHB goes back to the epidemic in 1993/1994/1995

(esp. 1994!) when I ran a small research farm in southern Manitoba. FHB may be a

cyclical disease that comes along only once in a long while, but when it gets going, it is a

huge problem that affects many people, especially farmers. Like Leonard and Bushnell, I

take my hat off to, and admire, those scientists who contribute to this field.

Page 8: PhD Fusarium

v

LIST OF ABBREVIATIONS

15-AcDON 15-Acetyl-Deoxynivalenol

15-DON-BSA Bovine Serum Albumin Conjugated to C-15 of DON

15-DON-HRP Horse Radish Peroxidase Conjugated to C-15 of DON

15-DON-OVA Ovalbumin Conjugated to C-15 of DON

3-AcDON 3-Acetyl-Deoxynivalenol

AAFC Agriculture and AgriFood Canada

Ab Antibody

Ag Antigen

AOX Alcohol Oxidase Promoter

AP Alkaline Phosphatase

ARM Antibody-Ribosome-mRNA Complex

BCIP 5-Bromo-4-Chloro-3-Indolyl Phosphate

BSA Bovine Serum Albumin

CD-ELISA Competitive Direct ELISA

cDNA Complementary DNA

CDR Complementarity Determining Region

CH Constant Domain of the Heavy Chain

cIgG Conventional Immunoglobulin Gamma (γ) Isotype Abs

CL Constant Domain of the Light Chain

Da Dalton

DAI Days After Inoculation

DAPI 4′,6-diamidino-2-phenylindole dihydro-chloride

Page 9: PhD Fusarium

vi

DAS Diacetoxyscirpenol

DMAP Dimethylaminopyridine

DMSO Dimethyl sulfoxide

DNA Deoxyribonucleic Acid

DON Deoxynivalenol

dsFv Disulfide-Stabilized Fv

ECORC Eastern Cereal and Oilseed Research Center

EDTA Ethylenediaminetetraacetic acid

ELISA Enzyme-Linked Immunosorbent Assay

Fab Fragment, Antigen Binding Region of an Antibody

FDK Fusarium Damaged Kernels

Ff Filamentous Bacteriophage

FHB Fusarium Head Blight

FITC Fluorescein Isothiocyanate

FP Fluorescence Polarization

FPA Fluorescence Polarization Assay

FR Framework Region

Fv Variable Fragment of an Antibody

GC/MS Gas Chromatogram / Mass Spectrometry

HCAb Heavy Chain Antibody

HEPES N-(2-hydroxyethyl)-piperazine-N-2-ethanesulfonic acid

HPI Hours Post Infection

HPLC High Pressure Liquid Chromatography

Page 10: PhD Fusarium

vii

HRP Horseradish Peroxidase

IC50 Half Maximal Inhibitory Concentration

IgA Immunoglobulin Alpha (α) Isotype Antibody

IgM Immunoglobulin Mu (μ) Isotype Antibody

IMAC Immobilized Metal Affinity Chromatography

IPTG isopropyl β-D-1-thiogalactopyranoside

JECFA Joint Expert Committee on Food Additives

KD Equilibrium Dissociation Constant

kDa kilodalton

LD50 Median Lethal Dose

LSD Least Significant Difference

mAb Monoclonal Antibody

MCS Multiple Cloning Site

MGYH Minimum Glycerol Medium with Histidine

MMH Minimal Methanol Medium

mRNA Messenger RNA

MW Molecular Weight

NBT Nitro Blue Tetrazolium Chloride

NEO Neosolaniol

NMR Nuclear Magnetic Resonance

NRCC National Research Council of Canada

NSERC National Sciences and Engineering Research Council

OAc Acetyl Ester (-OCH3)

Page 11: PhD Fusarium

viii

OD620 Optical Density at 620nm

OIsoval Isovalerate Ester (-OCOCH2CH(CH3)2)

OMAFRA Ontario Ministry of Agriculture, Food and Rural Affairs

OVA Ovalbumin

PAGE Polyacrylamide Gel Electrophoresis

PBL Peripheral Blood Lymphocytes

PBL Peripheral Blood Lymphocytes

PBS Phosphate Buffered Saline

PBST Phosphate Buffered Saline Tween

PCR Polymerase Chain Reaction

PEG Polyethylene Glycol

pIII Minor Coat Protein of Filamentous Phage

PMSF Phenylmethylsulphonyl Fluoride

PMTDI Provisional Maximum Tolerable Daily Intake

PVDF Polyvinylidene Fluoride

pVIII Major Coat Protein of Filamentous Phage

rAb Recombinant Antibody

RNA Ribonucleic Acid

RPL3 Ribosomal Protein L3

SAR Systemic Acquired Resistance

scFv Single Chain Fv

sdAb Single Domain Ab

SDS Sodium Dodecyl Sulfate

Page 12: PhD Fusarium

ix

SPR Surface Plasmon Resonance

ssDNA Single-Stranded DNA

StEP Staggered Extension Process

TBST Tween Buffered Saline

TEA Triethylamine

TFAA Trifluoroacetic anhydride

TLC Thin Layer Chromatography

UGT UDP Glycosyltransferase

USD United States Dollars

VH Variable Domain of the Heavy Chain

VHH Variable Region on Single-Domain Heavy Chain Ab

VL Variable Domain of the Light Chain

YPD Yeast Potato Dextrose media

YPDS Yeast Extract Peptone Dextrose Sorbitol Media

ΔAUC Differential Area Under Curve

Page 13: PhD Fusarium

x

TABLE OF CONTENTS

ABSTRACT ........................................................................................................................ i

PREFACE .......................................................................................................................... ii

ACKNOWLEDGEMENTS ............................................................................................. ii

PICTURE .......................................................................................................................... iii

NOTE ABOUT WHY ...................................................................................................... iv

LIST OF ABBREVIATIONS ...........................................................................................v

TABLE OF CONTENTS ..................................................................................................x

LIST OF TABLES ........................................................................................................ xvii

LIST OF FIGURES ..................................................................................................... xviii

1 INTRODUCTION, OBJECTIVES, AND HYPOTHESIS ......................................1

2 LITERATURE REVIEW 1: FUSARIUM HEAD BLIGHT AND

TRICHOTHECENE MYCOTOXINS: NOVEL TARGETS FOR

RECOMBINANT ANTIBODY TECHNOLOGY ....................................................6

2.1 Introduction ...........................................................................................................6

2.2 Fusarium Head Blight ...........................................................................................8

2.2.1 Disease Cycle ...........................................................................................8

2.2.1.1 Inoculum ...................................................................................10

2.2.1.2 Environment ..............................................................................11

2.2.1.3 Host Crop ..................................................................................11

2.2.2 Pathways of FHB Pathogenesis ..............................................................13

2.2.2.1 Fungal Colonization ..................................................................13

2.2.2.2 Systemic Infection .....................................................................15

Page 14: PhD Fusarium

xi

2.2.3 FHB Control Strategies ..........................................................................16

2.2.3.1 Agronomic and Cultural Practices ............................................16

2.2.3.2 Fungicides .................................................................................17

2.2.3.3 Biological Control Agents ........................................................17

2.2.3.4 Crop Breeding ...........................................................................18

2.2.4 Classifications of FHB Resistance .........................................................19

2.3 Trichothecene Mycotoxins ..................................................................................20

2.3.1 Mode of Action ......................................................................................21

2.3.2 Mechanisms of Fungal Protection ..........................................................22

2.3.3 Effects on Plants .....................................................................................23

2.4 DON, 3-AcDON, and 15-AcDON ......................................................................24

2.4.1 Contamination of Food and Feed ...........................................................25

2.4.2 Mechanism of Virulence for FHB ..........................................................26

2.5 Transgenic Approaches to Limit Trichothecene Toxicity ...................................28

2.5.1 Inhibition of Trichothecene Biosynthesis ...............................................29

2.5.2 Alteration of Target Protein ....................................................................30

2.5.3 Metabolic Trichothecene Detoxification ................................................31

2.5.4 Reduction of Intracellular Concentrations ..............................................33

2.5.5 Plantibodies and Mimotopes ..................................................................33

3 LITERATURE REVIEW 2: ANTIBODY ENGINEERING IN

AGRICULTURAL BIOTECHNOLOGY ...............................................................36

3.1 Introduction .........................................................................................................36

3.2 Recombinant Antibody Fragments .....................................................................37

Page 15: PhD Fusarium

xii

3.2.1 Fab Fragment ..........................................................................................38

3.2.2 scFv Fragment ........................................................................................39

3.2.3 VHH Fragment ........................................................................................42

3.3 Development and Types of Antibody Libraries ..................................................44

3.3.1 Immunized Libraries...............................................................................44

3.3.2 Naïve Libraries .......................................................................................45

3.3.3 Synthetic / Semi-Synthetic Libraries ......................................................46

3.4 Display Systems ..................................................................................................47

3.4.1 Phage Display .........................................................................................47

3.4.1.1 Filamentous Bacteriophage .......................................................48

3.4.1.2 Phage vs. Phagemid Vectors .....................................................49

3.4.1.3 Panning .....................................................................................52

3.4.2 Bacteria Display .....................................................................................54

3.4.3 Yeast Display ..........................................................................................54

3.4.4 Ribosome Display ..................................................................................55

3.5 Optimization of Recombinant Antibodies ..........................................................57

3.5.1 Optimized Panning Procedures ..............................................................57

3.5.2 In vitro Affinity Maturation ....................................................................58

3.5.2.1 Random Mutagenesis ................................................................59

3.5.2.1.1 Error-Prone PCR ...................................................59

3.5.2.1.2 Bacterial Mutator Strains .......................................59

3.5.2.2 Focused Mutagenesis ................................................................60

3.5.2.2.1 Site-Directed Mutagenesis .....................................60

Page 16: PhD Fusarium

xiii

3.5.2.2.2 Mutational Hot Spots.............................................61

3.5.2.2.3 Parsimonious Mutagenesis ....................................62

3.5.3 Optimized Panning Procedures ..............................................................63

3.5.3.1 Chain Shuffling .........................................................................63

3.5.3.2 DNA Shuffling by Random Fragmentation and

Reassembly .......................................................................................64

3.5.3.3 Staggered Extension Process (StEP) .........................................65

3.6 Expression and Purification ................................................................................65

3.6.1 Prokaryotic Systems ...............................................................................66

3.6.1.1 E. coli Cytoplasmic Expression ................................................66

3.6.1.2 E. coli Periplasmic Expression .................................................67

3.6.2 Eukaryotic Systems ................................................................................68

3.6.2.1 Yeast Expression .......................................................................68

3.6.2.2 Insect Cell Expression ...............................................................71

3.6.2.3 In planta Expression .................................................................73

3.7 Application to Immunoassays and Agricultural Biotechnology .........................77

3.7.1 Immunoassays ........................................................................................77

3.7.2 Research Applications to Agricultural Biotechnology ...........................80

4 RESEARCH CHAPTER 1: CLONING, EXPRESSION, AND

CHARACTERIZATION OF A SINGLE DOMAIN ANTIBODY FRAGMENT

WITH AFFNIITY FOR 15-ACETYL-DEOXYNIVALENOL..............................82

4.1 Abstract ...............................................................................................................82

4.2 Introduction .........................................................................................................83

4.3 Materials and Methods ........................................................................................86

Page 17: PhD Fusarium

xiv

4.3.1 Reagents and Solutions ..........................................................................86

4.3.2 Immunogen and Llama Immunization....................................................87

4.3.3 Fractionation of Polyclonal Serum .........................................................88

4.3.4 Confirmation of Titer .............................................................................89

4.3.5 Library Construction ...............................................................................90

4.3.6 VHH Library Panning .............................................................................92

4.3.7 Phage ELISA ..........................................................................................93

4.3.8 Colony PCR and Sequencing .................................................................94

4.3.9 Monomer Expression and Purification ...................................................94

4.3.10 Pentamerization of VHH Monomer .......................................................95

4.3.11 Pentamer Expression and Purification ..................................................96

4.3.12 Determination of Binding Affinity ........................................................96

4.3.12.1 Fluorescence Polarization Assay..............................................96

4.3.12.2 Surface Plasmon Resonance ....................................................98

4.3.12.3 Competitive Inhibition ELISA .................................................99

4.4 Results ...............................................................................................................100

4.4.1 Polyclonal Response ..............................................................................100

4.4.1.1 Direct ELISA ..........................................................................100

4.4.1.2 Fluorescence Polarization .......................................................101

4.4.2 HCAb and cIgG Response .....................................................................102

4.4.2.1 Protein A and G Chromatography...........................................102

4.4.2.2 ELISA on Polyclonal Serum Fractions ...................................102

4.4.3 Library Construction ..............................................................................103

Page 18: PhD Fusarium

xv

4.4.4 Panning and Phage ELISA ....................................................................103

4.4.5 VHH Sequence Results ..........................................................................104

4.4.6 Expression of Soluble VHH Protein ......................................................105

4.4.7 VHH Affinity Measurements .................................................................105

4.4.7.1 Fluorescence Polarization .......................................................105

4.4.7.2 Surface Plasmon Resonance ...................................................106

4.4.7.3 CI ELISA ................................................................................110

4.5 Discussion .........................................................................................................110

4.6 Conclusions .......................................................................................................118

4.7 Acknowledgements ...........................................................................................119

5 RESEARCH CHAPTER 2: INTRACELLULAR EXPRESSION OF A

SINGLE-DOMAIN ANTIBODY REDUCES CYTOTOXICITY OF 15-

ACETYL-DEOXYNIVALENOL IN YEAST. ......................................................120

5.1 Abstract .............................................................................................................120

5.2 Introduction .......................................................................................................121

5.3 Experimental Procedures ..................................................................................124

5.3.1 VHH Genes ............................................................................................124

5.3.2 Transformation ......................................................................................125

5.3.3 Induction of VHH Expression ................................................................126

5.3.4 Preparation of Soluble VHH Extracts and Western Blot Analysis ........126

5.3.5 Cytotoxicity Assays ...............................................................................127

5.3.6 Data Analysis .........................................................................................128

5.3.7 Quantitative Western Blot Analysis ......................................................129

5.3.8 VHH Immunolocalization ......................................................................131

Page 19: PhD Fusarium

xvi

5.3.9 Mycotoxin Biotransformation Assays ...................................................132

5.4 Results ...............................................................................................................134

5.4.1 Sensitivity to Ribotoxin Treatments ......................................................134

5.4.2 Transformation and VHH Intrabody Expression ....................................135

5.4.3 Quantitative Western Blot Assay ..........................................................135

5.4.4 Cytotoxicity Assays ...............................................................................137

5.4.4.1 15-AcDON ..............................................................................137

5.4.4.2 DON, 3-AcDON, Cycloheximide ...........................................139

5.4.4.3 B-24 VHH Control ...................................................................141

5.4.5 VHH Immunolocalization ......................................................................141

5.4.6 Mycotoxin Biotransformation Assays ...................................................141

5.5 Discussion .........................................................................................................145

5.6 Acknowledgements ...........................................................................................153

5.7 Supplemental.....................................................................................................154

6 CONCLUSIONS ......................................................................................................159

7 REFERENCES .........................................................................................................162

Page 20: PhD Fusarium

xvii

LIST OF TABLES

Table 4.1 Nucleotide sequences of primers for PCR amplification of HCAb VHH

genes……………………………………………………………………...91

Table 4.2 HCAb and cIgG fractions of llama serum. Volumes of pre-immune, 42-

day and 70-day samples = 12.1, 12.5 and 15.5 mL, respectively............. 102

Table 5.1 Sensitivity of P. pastoris (KM71H) pPICZB transformants to various

ribotoxin treatments tested. Standard deviations (σ) for each mean value

shown in brackets..................................................................................... 134

Table 5.2 Mean VHH intrabody concentration (t = 0 h) within P. pastoris

transformants used in cytotoxicity assays.................................................136

Table 5.S1 Raw data and calculations of VHH intrabody expression in Pichia pastoris

transformants derived from quantitative Western blot analysis........155/156

Table 5.S2 HPLC retention times of trichothecene metabolites evaluated after 24 h of

growth of P. pastoris transformants cultured in YPD media supplemented

with 100 µg·mL-1

of highly-purified DON...............................................158

Page 21: PhD Fusarium

xviii

LIST OF FIGURES

Figure 1.1 Structure, composition, and molecular weight of trichothecene mycotoxins

used within this study................................................................................... 2

Figure 2.1 Photos of wheat showing progressive signs of FDK (Fusarium damaged

kernels) in wheat.......................................................................................... 6

Figure 2.2 Disease Cycle of FHB infection of small grain cereals................................9

Figure 2.3 The grass floret. A: Partly dissected at anthesis. The ovary and two

stigmas comprise the pistil. B: Transverse diagram showing overlap of

lemma and palea.........................................................................................12

Figure 2.4 Scanning electron micrographs of F. culmorum colonization of wheat (cv.

Agent).........................................................................................................13

Figure 2.5 Penetration of lemma of wheat (cv. Agent) by F. culmorum.....................14

Figure 2.6 Schematic diagram of the first two florets of a wheat spikelet, showing the

relationship among the ovary (O), lemma (L), glume (G), palea (P),

rachilla (RL), Rachis Node (RN), and Rachis (R) in terms of initial

Fusarium infection sites (red arrowheads) and systemic infection pathways

(blue arrows) of F. culmorum.................................................................... 15

Figure 2.7 Diagram of the general structure, numbering system of the tetracyclic

trichothecene nucleus, and specific side-chain groups of selected naturally-

occurring trichothecnes of the genus Fusarium..........................................20

Figure 2.8 Structure of DON, 15-AcDON, and 3-AcDON......................................... 25

Figure 2.9 Immunogold localization of Fusarium toxins in infected host tissue (cv.

Agent) by F. culmorum.............................................................................. 27

Figure 3.1 Schematic representation (left) and ribbon diagram (right) of recombinant

antibody fragment (Fab)............................................................................. 39

Page 22: PhD Fusarium

xix

Figure 3.2 Schematic representation (left) and ribbon diagram (right) of recombinant

single chain variable fragment (scFv) antibody.........................................40

Figure 3.3 Schematic representation (left) and ribbon diagram (right) of recombinant

HCAb variable domain (VHH) of antibody “AMD9”................................ 42

Figure 3.4 Schematic representation of a filamentous bacteriophage and its protein

coats...........................................................................................................50

Figure 3.5 Phage (left) and phagemid (right) vectors to construct rAb phage display

libraries through the minor coat protein pIII..............................................51

Figure 3.6 Phage display panning cycle.......................................................................53

Figure 3.7 Ribosome display panning cycle................................................................ 56

Figure 4.1 ELISA of polyclonal llama serum response to 250 μg 15-DON-BSA

conjugate...................................................................................................101

Figure 4.2 Amino acid sequence of NAT-267 VHH from fourth round of panning..104

Figure 4.3 FP inhibition of mNAT-267 and pNAT-267 VHH binding and 56-day

polyclonal llama serum binding to DON-fluorescein tracer with DON

toxin (A); 3-AcDON (B); and 15-AcDON (C) from 5 μg∙mL-1

to 9.8

ng∙mL-1

..................................................................................................... 107

Figure 4.4 Overlay sensorgrams of mNAT-267 VHH steady state (Req) binding to 15-

DON-HRP conjugate................................................................................108

Figure 4.5 (A) Steady state (Req) affinity binding (RU) mNAT-267 VHH to 15-DON-

HRP conjugate for ten monomer concentrations, as shown in Figure 4.4.

(B) Scatchard plot of steady state „Req‟ binding (RU) relative to Req / C

(mNAT-267 VHH concentration) (RU/μM)............................................. 109

Figure 4.6 Structure, composition, and molecular weight of trichothecene mycotoxins

used in this study...................................................................................... 114

Page 23: PhD Fusarium

xx

Figure 5.1 Structure, composition, and molecular weight of trichothecene mycotoxins

used in this study...................................................................................... 122

Figure 5.2 Diagram of pPICZB expression vector (3745 bp) with VHH gene (NAT-

267 = 387 bp, B-24 = 390 bp)................................................................. 125

Figure 5.3 Quantitative Western blot analysis of VHH intrabody expression within P.

pastoris cytotoxicity assays...................................................................... 135

Figure 5.4 Cytotoxicity assay of P. pastoris pPICZB and NAT-267 VHH transformant

growth measured by optical density at 620 nm (OD620) to 15-AcDON at

doses of 20 to 200 µg·mL-1

.......................................................................138

Figure 5.5 Cytotoxicity assay of P. pastoris pPICZB and NAT-267 VHH transformant

growth measured by optical density at 620 nm (OD620) to DON and

cycloheximide at doses of 50 to 200 µg·mL-1

.......................................... 140

Figure 5.6 Cytotoxicity assays showing the effect of various concentrations (50 to

200 µg·mL-1

) of 15-AcDON (A) and cycloheximide (B) on the growth of

P. pastoris pPICZB and non-specific B-24 VHH transformants as

measured by optical density at 620 nm (OD620)....................................... 142

Figure 5.7 Representative confocal microscopy photographs of P. pastoris (KM71H)

cells immuno-probed with anti-HA epitope mAb-FITC conjugate and

DAPI stain. Transformants expressing pPICZB empty vector (control) (A)

and NAT-267 VHH intrabody (B).............................................................143

Figure 5.8 Representative 500 mHz 1H NMR spectrum of A = 15-AcDON Standard.

B = HPLC fraction of P. pastoris cell lysate sample (24 hrs).................. 144

Figure 5.S1 Homology chart of DNA and amino acid sequence of codon-optimized

NAT-267 and control B-24 VHH fragments.............................................154

Page 24: PhD Fusarium

1

1 INTRODUCTION, OBJECTIVES, AND HYPOTHESIS

Fusarium head blight (FHB) of cereals is caused by morphologically similar species

(F. graminearum, F. culmorum, etc.) common throughout global agricultural regions.

With few exceptions, Fusarium epidemics are characterized by cyclical and highly

aggressive infection of cereal crops with economic impacts on food and feed industries

that are immediate and far reaching (Demcey Johnson et al., 2003; McMullen, 2003;

Miller, 2008).

Trichothecene mycotoxins are a toxin class commonly found within agricultural

commodities infected by Fusarium. Trichothecene toxins represent a large group of >180

sesquiterpenoids that have molecular weights typically ranging from 200-500 Da and are

characterized by a tricyclic ring structure containing a double bond at C-9,10 and an

epoxide group at C-13 (reviewed in Desjardins, 2006; Foroud and Eudes, 2009; Mirocha

et al., 2003). Regardless of size and structural composition, trichothecenes are potent

inhibitors of eukaryotic protein synthesis with specific activity on ribosomal protein L3

(RPL3) within the 60S subunit resulting in inhibition of peptidyl transferase activity

(Carter and Cannon, 1977; Rocha et al., 2005). Although the capacity to inhibit protein

synthesis is regarded as central to trichothecene cytotoxicity (Ueno, 1984; Rotter et al.,

1996), adverse effects on eukaryotic cells may actually be attributed to dysregulation of

cellular signaling and alterations in downstream gene expression (Pestka, 2008).

Deoxynivalenol (3,7,15-trihydroxy, 12,13-epoxy-trichothec-9-en-8-one), or DON, is

one of many members of the trichothecene group of mycotoxins produced by Fusarium

species. DON is a low molecular weight hapten (MW = 296 Da) containing one primary

and two secondary hydroxyl groups conferring limited solubility in water and high

Page 25: PhD Fusarium

2

solubility in various polar solvents such as methanol and acetonitrile (reviewed in

Desjardins, 2006; Mirocha et al., 2003). Of the trichothecene mycotoxins that

accumulate within cereal grain, DON and its closely related-acetylated analogues (i.e.,

15-Acetyl-Deoxynivalenol (15-AcDON) and 3-Acetyl-Deoxynivalenol (3-AcDON))

(Figure 1.1) are considered among the most common and widely distributed food and

feed contaminants (Abramson et al., 2001; D‟Mello et al., 1999; Greenhalgh et al.,

1986; Snijders, 1990a).

Trichothecene MW

a

(Da)

Oxygenation and Esterification at Position

C-3 C-15

3-Acetyl-DON 338 O-CO-CH3 OH

15-Acetyl-DON 338 OH O-CO-CH3

Deoxynivalenol 296 OH OH a MW = Molecular Weight

Figure 1.1 Structure, composition, and molecular weight of DON, 15-AcDON, and 3-

AcDON (Desjardins, 2006).

There is a demonstrated correlation between in planta DON accumulation and

Fusarium virulence in susceptible cultivars of wheat (Mesterházy, 2002) and maize

(Harris et al., 1999a). Based on these findings, mechanisms which convey innate and

Page 26: PhD Fusarium

3

acquired host plant resistance to DON and other trichothecene toxins have received

considerable attention. To date, in planta trichothecene resistance has been achieved

through mechanisms that alter targeted proteins within host cell ribosomes (e.g., Harris

and Gleddie, 2001; Mitterbauer et al., 2004), promote metabolic transformation to less

toxic forms: e.g., DON-glucosyl conjugate (Poppenberger et al., 2003) or to 3-AcDON

(Alexander, 2008), and/or reduce intracellular concentrations to effectively limit

mycotoxin exposure to sensitive cellular targets. Collectively, such research can be

applied to assess and impart novel mechanisms of trichothecene resistance within

eukaryotic test systems such as yeast or Arabidopsis (Mitterbauer and Adam, 2002) for

eventual application in higher order plants such as cereals and maize.

Current applications of antibody (Ab)-based technology involving FHB and

trichothecene toxins such as DON, 3-AcDON, and 15-AcDON are generally limited to

immunoassays based on polyclonal and monoclonal antibodies (mAb) to detect

mycotoxins within grain, food, feed, and animal serum (e.g., Maragos and McCormick,

2000; Nicol et al., 1993; Sinha et al., 1995). Although these tests provide robust and

precise results, mAb development requires highly specialized equipment and labor-

intensive procedures to select, culture, and maintain optimal hybridoma cell lines.

Because of these limitations, recombinant Ab and phage display technologies are often

used to generate target-specific recombinant antibody (rAb) fragments from Ab cell lines.

In this thesis, these two technologies were used to develop and quantify the efficacy of a

single-domain heavy chain antibody fragment (i.e., VHH) with affinity for 15-AcDON.

Page 27: PhD Fusarium

4

VHH fragments are derived from the camelidae heavy chain IgG subfamily and are

among the smallest functional rAb fragments known. VHH fragments exhibit the same

exquisite specificity as larger immunoglobulins, and confer added biochemical

advantages of high solubility, stability, and robust expression in various recombinant

systems (Holliger and Hudson, 2005; Muyldermans, 2001). VHH fragments have been

generated against a wide variety of epitopes and antigen (Ag) types including low

molecular weight ligands (haptens) (Nguyen et al., 2000; Sheedy et al., 2006; van der

Linden et al., 2000) and toxins (e.g., Doyle et al., 2008).

The advantages associated with ease of expression, stability, and solubility of VHH

fragments can be attributed to key differences when compared to standard rAb fragments

(e.g., scFv, Fv, Fab, etc.). VHH fragments are small (~ 14 – 15 kDa), typically have

hydrophilic amino acid substitutions within the second framework region (FR-2), and by

their single-domain nature, do not require the linker associated with the standard scFv

format which can lead to problems associated with aggregation, proteolytic degradation,

and variable expression within recombinant hosts (Arbabi-Ghahroudi et al., 2005).

Improved functionality over various temperature and solvent regimes (Goldman et al.,

2006; van der Linden et al., 1999) coupled with high levels of recombinant expression

combine to make VHH fragments useful reagents within next-generation immunoassays

(Goldman et al., 2006, Ladenson et al., 2006). When expressed as intrabody fragments

within recombinant hosts, VHH fragments can be used as Ag-specific reagents to

elucidate potential in vivo mechanisms of immunomodulation with applications ranging

Page 28: PhD Fusarium

5

from pharmacology (Holliger and Hudson, 2005; Saerens et al., 2008) to plant science

(Jobling et al., 2003).

The research summarized within this Ph.D. project was based on a two related

objectives. Initial experiments were established to identify, characterize, and assess

binding affinity of a trichothecene-specific recombinant VHH fragment that was isolated

from a hyper-immunized phage-display library. The second group of experiments were

focused on an in vivo assessment of a VHH intrabody immunomodulation of the cytotoxic

effects of the target mycotoxin within a model eukaryotic system based on Pichia

pastoris. Collectively, this research was established as a „proof of concept‟ model to

demonstrate that intrabody expression, i.e. expression of recombinant VHH fragments,

could impart a novel means of mycotoxin-specific resistance in susceptible eukaryotic

organisms.

Hypothesis

Expression of a mycotoxin-specific VHH antibody fragment within a model

eukaryotic test system will limit mycotoxin-specific cytotoxicity and inherently

reduce disease symptoms associated with FHB pathogenesis.

Page 29: PhD Fusarium

6

2 LITERATURE REVIEW 1: FUSARIUM HEAD BLIGHT AND

TRICHOTHECENE MYCOTOXINS: NOVEL TARGETS FOR

RECOMBINANT ANTIBODY TECHNOLOGY

2.1 Introduction

Fusarium head blight (FHB) of small grain cereals is a fungal disease caused by a

range of Fusarium species common throughout the world‟s agricultural regions.

Epidemics are characterized by highly cyclical and aggressive plant pathogenesis with

few known mechanisms of control. Despite causing significant yield loss, the principal

economic impact of a FHB epidemic usually results in harvested grain with severely

reduced quality since Fusarium-damaged kernels (FDK) (Figure 2.1) are often shrunken

and shriveled with little economic value. Reduced seed quality and value is exacerbated

by the fact that infected grain is often contaminated with toxic fungal metabolites, or

mycotoxins, which affect food and feed quality (Bai and Shaner, 1994; Dickson, 1942;

Foroud and Eudes, 2009; McMullen, 2003; Miller, 2008; Parry et al., 1995; etc.).

Figure 2.1 Photos of wheat showing progressive signs of FDK (Fusarium damaged

kernels) in wheat (Harvest samples from Syngenta Field Trials, 2004).

The two major classes of mycotoxins common to Fusarium infected small grain

cereals are trichothecenes and zearalenone. Trichothecenes are tricyclic sesquiterpenes

that contain a 12,13-epoxide ring and a double bond between carbons 9 and 10.

Page 30: PhD Fusarium

7

Zearalenones are an unrelated group of Fusarium mycotoxins derived by different

cyclizations and modifications of nonaketide precursors. Like trichothecenes,

zearalenones are low molecular weight, heat-stable mycotoxins commonly found within

Fusarium-infected cereals and maize (Desjardins, 2006).

Unlike zearalenones, which are not generally considered to be phytotoxic

compounds, trichothecenes can produce a wide array of phytotoxic effects within plant

tissue. Some of the first evidence to link trichothecenes as a mechanism of virulence for

Fusarium infection of plant tissue was provided by gene disruption experiments. Various

researchers showed that loss-of-function mutations in the Tri5 gene (key enzyme within

the trichothecene biosynthesis pathway) within virulent Fusarium spp. resulted in reduced

fungal pathogenesis and colonization of plant tissue (Desjardins et al., 1996; Eudes et al.,

2001; Proctor et al., 1995). Subsequent research, as outlined within this chapter,

demonstrated that various other techniques to limit trichothecene (particularly

deoxynivalenol) phytotoxicity could limit Fusarium infection of small grain cereals and

maize. In addition, research has shown that the production of DON within plant tissue is

commonly associated with the necrotrophic stage of Fusarium pathogenesis during which

host-plant tissue dies as the fungus advances and proliferates within infected cereal tissue

(Mesterházy, 2002; Muehlbauer and Bushnell, 2003; Proctor et al., 2002).

The following chapter provides an overview of the general aspects related to the

occurrence of, and control strategies associated with, FHB infection and trichothecene

contamination of agricultural crops. Given the vast array and enormous scope of

publications covering this subject matter, this introductory chapter is intended to be a

Page 31: PhD Fusarium

8

concise and general overview to support, and align with subsequent research chapters

within this thesis.

2.2 Fusarium Head Blight

FHB has the most dramatic and widespread effect on cereal production during

major epidemics; however once established within an area, even at baseline infestation

levels, FHB can be a major limitation to cereal production (Diaz de Ackermann and

Kohli, 1997a; Mesterházy, 1997; Sutton, 1982; Wang, 1997). Arguably, the best

documented, and perhaps the most costly FHB epidemic in North America occurred in

the upper Great Plains of the USA and within the provinces of Manitoba, Ontario, and

Quebec, Canada between 1993 and 1998 (Gilbert et al., 1994; McMullen, 2003;

McMullen et al., 1997; Windels, 2000; etc.). Estimated economic losses associated with

lost crop yield and reduced harvest quality over the course of the epidemic range from

$1.3 to nearly $3.0 billion USD (Demcey Johnson et al., 2003; Windels, 2000).

2.2.1 Disease Cycle

The critical factors or conditions which must exist for FHB to occur are described

by the classic plant pathologist „Disease Triangle‟ denoted by: 1.) inoculum, 2.)

susceptible host, and 3.) the right mix of environmental conditions (Shaner, 2003). More

specifically, the establishment of FHB can be attributed to the release of inoculum from

partially-degraded crop residues throughout periods of warm, windy, humid weather

during anthesis (i.e. flowering) of susceptible host crops (Figure 2.2).

FHB is a monocyclic disease that is initiated by inoculum carried within air currents

or splashing water (Sutton, 1982). The quantity of primary inoculum therefore directly

Page 32: PhD Fusarium

9

influences the intensity and extent of disease development during infection (Bai and

Shaner, 1994; Khonga and Sutton, 1986, 1988). Although secondary infections are

theoretically possible, the monocyclic nature of FHB can be attributed to the relatively

short period of crop susceptibility and limited window of favorable environmental

conditions which favor FHB establishment (Fernando et al., 1997; Shaner, 2003).

Figure 2.2 Disease Cycle of FHB infection of small grain cereals (from Bergstrom,

1993).

Fusarium seedling blight, or Fusarium crown and foot rot, is a related seed-borne

disease of cereals (Figure 2.2). Affected seedlings emerge stunted and yellow with the

crown, roots and/or lower stem having a brown to red-brown appearance (Sutton, 1992).

Although FHB is not regarded as a systemic disease based on an infection that develops

from Fusarium infected seed, seedling blight may serve as a source of secondary

inoculum within the canopy of the growing crop (Parry et al., 1995).

Conidia

(asexual)

Ascospores

(sexual)

Moist, Warm Conditions at

Anthesis

Infected Seed

Fusarium

Seedling Blight

Reduced

tillage

Crop debris (wheat, maize, barley)

bearing perithecia and conidia

Fusarium

Head

Blight

Page 33: PhD Fusarium

10

2.2.1.1 Inoculum

While several species of Fusarium cause FHB around the world, including F.

graminearum Schw. (Teleomorph – Gibberella zeae Schw. Petch), F. culmorum (Wm. G.

Smith) Sacc., and F. avenaceum (Corda ex Fr.) Sacc. (Teleomorph - G. avenacea

(Cook)), etc., F. graminearum is the most common within North America, Europe and

Asia (McMullen et al., 1997; Parry et al., 1995; Sutton, 1982; Tekauz et al., 2000;

Wilcoxson et al., 1988).

Infected crop residue (i.e., debris of previously-harvested wheat, barley, maize) is

the principle source of primary F. graminearum inoculum for FHB infestations (Figure

2.2). Therefore, inoculum levels are related to the amount of crop residue and the degree

to which crop debris is infected on the surface of soil (Khonga and Sutton, 1988: Sutton,

1982). Challenges associated with inoculum build-up are exacerbated by reduced tillage

practices, which result in larger amounts of, and longer degradation profiles for, crop

residues within the top layer of soil (Shaner, 2003). Primary types of inoculum of

virulent Fusarium species include ascospores (F. graminearum), as well as conidia,

hyphal fragments, and chlamydospores (F. culmorum, F. avenaceum, etc.). Environment

has a direct impact on inoculum production and release since warm, moist, and windy

weather disseminate Fusarium inocula by rain-splash and air currents to enable direct

contact of sexual and asexual innoculum with susceptible tissue of host plants (Menzies

and Gilbert, 2003) (Figure 2.2).

Page 34: PhD Fusarium

11

2.2.1.2 Environment

The role of environment in FHB epidemiology is a complex subject that has been

extensively reviewed (Andersen, 1948; Sutton, 1982; Parry et al., 1995; Shaner, 2003).

The inherent complexity of the disease is associated primarily with temperature and

duration of continuous moisture, which are both tied to rainfall and prevailing humidity

levels and are inherently variable factors that affect both inoculum production and

dispersal as well as the duration of host crop susceptibility to Fusarium infection.

Therefore, the establishment of the FHB disease cycle (Figure 2.2) is dependent upon

prolonged precipitation and/or periods of high humidity (e.g., 92 - 94% R.H.) in

conjunction with warm temperatures during anthesis of host crops (Sutton, 1982).

Prevailing moisture has a greater role than temperature in terms of inoculum production,

distribution, and colonization and pathogen development within infected heads because

favorable temperatures for FHB establishment are generally common when cereal crops

are flowering (Shaner, 2003; Sutton, 1982). However, it is also well established that

longer periods of prevailing moisture are required for FHB infection when temperatures

are lower or higher than 25°C (Andersen, 1948).

2.2.1.3 Host Crop

The initial point of Fusarium infection associated with FHB of cereals is fungal

colonization of floret and caryopsis tissue. Cereal florets are composed of an inner palea

(Figure 2.3A) and an outer lemma (Figure 2.3B), which overlap and partially enclose the

edge of the palea. Together, the lemma and palea enclose the pistil, stamens, lodicules,

and an ovary to form the caryopsis (Figure 2.3) (Bushnell et al., 2003).

Page 35: PhD Fusarium

12

Floret and caryopsis host tissues are most susceptible to Fusarium infection within a

3 - 5 day period corresponding to flowering or anthesis (i.e., BBCH = 61 - 69). However,

depending on prevailing environmental conditions and innoculum levels, colonization

can occur from mid inflorescence (i.e., BBCH = 55) to the soft dough growth stage (i.e.,

BBCH = 85) (Paulitz et al., 1997). After fungal colonization, disease severity progresses

rapidly to a necrotrophic systemic infection which results in widespread Fusarium

infection throughout the cereal spike (Shaner, 2003).

Figure 2.3 The grass floret/caryopsis. A: Partly dissected at anthesis. The ovary and

two stigmas comprise the pistil. B: Transverse diagram showing overlap

of lemma and palea (adapted from Bushnell et al., 2003).

lemma

lodicule

palea

anther

ovary

lodicule

Figure 2.3B

Figure 2.3A

ovary rachilla

stigma

filament

anther

ventral

crease

Page 36: PhD Fusarium

13

2.2.2 Pathways of FHB Pathogenesis

2.2.2.1 Fungal Colonization

Upon initial germination of ascospores (i.e., first ~ six hours), Fusarium hyphae

rapidly elongate over the abaxial (exterior) surfaces of glumes and florets to form a

complex mycelial network across the surface of colonized host tissue (Figure 2.4A).

Figure 2.4 Scanning electron micrographs of F. culmorum colonization of wheat (cv.

Agent). A, Hyphal network between pollen grains (PG) on brush hairs at

the top of the ovary (48 hours post infection (HPI)). B, Hyphae (H) within

digested host cuticular layer on the inner surface of palea next to plant hair

structure (48 HPI). C, Hyphae (H) growing upward over the edge of the

glume from germinating macroconidia (M). (Kang and Buchenauer, 2000).

From initial stages of infection, hyphae initiate penetration of host tissue through

enzymatic degradation of abaxial surfaces of the caryopsis (Baláza and Bagi, 1997;

Urbanek, 1989; etc.) (Figure 2.4B). Seventy-six HPI, hyphae can progress across the

outer surfaces of the lemma and glume to reach the developing caryopsis (Figure 2.4C)

A

5 µm

H H

M

C

5 µm

B

20 µm

H

Page 37: PhD Fusarium

14

(Kang and Buchenauer, 2000). Throughout the later stages of this biotrophic infection

process, mycelial growth progresses to the point where fungal reproductive structures

(conidiophores) are established on the surface of infected tissue (Pritsch et al., 2000).

Fusarium hyphae follow various routes to reach interior surfaces of infected plant

tissue (Bushnell et al., 2003). These routes of entry include direct wall penetration of the

epidermal cell wall by a hyphal peg (Figure 2.5A), entry through stomates (Figure 2.5B),

or colonization of openings between lemma and palea during the brief period of

dehiscence when cereal florets are open (Lewandowski and Bushnell, 2001). Following

primary penetration, Fusarium hyphae exhibit a subcuticular growth pattern with flat,

branched hyphae growing between the cuticle and epidermal cell walls of host tissue

(Pritsch et al., 2000).

Figure 2.5 Penetration of lemma of wheat (variety = Agent) by F. culmorum. A,

Transmission electron micrograph (TEM) showing penetration peg

(arrowhead) from an infection hyphae (IH) invading epidermal cell wall

(EW) at 36 HPI. B, Scanning electron micrograph (SEM) showing growth

of hyphae (H) through a stomatal (S) opening on the inner surface of the

lemma 48 HPI (Kang and Buchenauer, 2000).

A B

S

H

1 µm 5 µm

Page 38: PhD Fusarium

15

2.2.2.2 Systemic Infection

Once fungal colonization is established, Fusarium hyphae infect adjacent florets on

the same spikelet through a systemic and necrotic infection of the rachis and rachis node

(Figure 2.6). Systemic infection is marked by increased fungal colonization and a rapid

necrotic deterioration of host-cell tissue because invasive hyphae progress from floret to

floret (Ribichich et al., 2000; Schroeder and Christensen, 1963). Thereafter, infected

florets cease to develop as host cell tissue and organelles become completely fragmented

as hyphae spread vertically and horizontally within the rachis and rachis node (Figure

2.6).

Figure 2.6 Schematic diagram of the first two florets of a wheat spikelet, showing the

relationship among the ovary (O), lemma (L), glume (G), palea (P),

rachilla (RL), rachis node (RN), and rachis (R) in terms of initial

Fusarium infection sites (red arrowheads) and systemic infection

pathways (blue arrows) of F. culmorum. Dotted lines and dotted areas

indicate vascular tissues within the RN (Kang and Buchenauer, 2000).

O

RL

O P

G L

RN

R

Page 39: PhD Fusarium

16

Continued necrotic infection leads to a degeneration of plant cytoplasts, cell walls,

chloroplasts and other plastids, which limit the ability of the phloem and xylem to deliver

nutrients to developing tissue. FHB infection is characterized by premature death of

infected spikelets, resulting in bleached and shriveled kernels often referred to as

tombstones or Fusarium damaged kernels (Figure 2.1).

2.2.3 FHB Control Strategies

2.2.3.1 Agronomic and Cultural Practices

FHB epidemics are initiated by inoculum (mostly ascospores for F. graminearum)

produced within crop residues on the soil surface. Any measure to reduce crop residue

accumulation and longevity on the soil surface such as plowing or burning excess residue

has the potential to reduce primary inoculum levels (Dill-Macky and Jones, 2000;

Fernandez et al., 1993). Furthermore, agronomic techniques to stagger seeding dates and

maintain long rotation periods between susceptible crops (e.g. cereals or maize) within a

given area are also recommended practices to reduce inoculum levels and limit FHB

development (Dill-Macky and Jones, 2000; McMullen et al., 1997).

Unfortunately, it is difficult to predict the effectiveness of such practices to limit

FHB (Meier et al., 2000) as disease establishment within a susceptible crop is highly

dependent on climate and irregular in terms of disease incidence and severity. In many

cases, environmental conditions associated with the year of production tend to be the

single largest factor associated with variation in disease impact (Schaafsma et al., 2001).

Page 40: PhD Fusarium

17

2.2.3.2 Fungicides

Fungicide applications are commonly used to limit yield losses associated with FHB

infection and mycotoxin contamination of small grain cereals (reviewed by Mesterházy,

2003b). Among the currently-available fungicides registered for FHB control and

mycotoxin suppression, products based on triazole chemistry (e.g., tebuconazole,

metconazole, or prothioconazole) are considered to be the most efficacious (Jennings,

2002; Mesterházy, 2003b; etc.). However, commercial experience and extensive trial

data from various sources have shown that fungicide efficacy is variable at best (e.g.,

D‟Mello et al., 1998; Homdork et al., 2000; Jennings et al., 2000; Simpson et al., 2001).

Chemical disease control is dependent on timing and method of application, host crop

FHB susceptibility, as well as prevailing environmental conditions, and Fusarium

inoculum levels (Magan et al., 2002; Mesterházy, 2003b). Given the complexity

associated with all of the variables that affect FHB pathogenesis, fungicide treatments are,

therefore, recommended and considered most effective when used as part of an integrated

crop management strategy (Milus and Parsons, 1994; Teich, 1989) that is based on an

accurate disease forecasting system and sound agronomy (McMullen et al., 1997;

Schaafsma et al., 2001).

2.2.3.3 Biological Control Agents

A common approach to biological control strategies is to apply non-pathogenic

organisms in advance of Fusarium infection of cereals as part of a competitive exclusion

strategy to disrupt the FHB disease cycle upon spikelet colonization and infection

(reviewed by da Luz et al., 2003). Numerous research groups have tested a wide range of

Page 41: PhD Fusarium

18

biological control agents against diseases attributed to Fusarium spp. (e.g., Huang et al.,

1993; Kempf and Wolf, 1989; Shi and Wang, 1991; Stockwell et al., 2000; etc.);

however, very few researchers have reported successful large-scale applications of this

strategy. Additional work and screening is required to identify biological control strains

and application technologies to limit FHB colonization and mycotoxin accumulation

under various field conditions (Chen et al., 2000; da Luz et al., 2003).

2.2.3.4 Crop Breeding

The standard approach to develop cultivars with enhanced FHB resistance has been

to cross cultivars with resistance or enhanced tolerance to Fusarium infection with a

susceptible, but agronomically superior and locally adapted, cultivars or crop varieties to

generate progeny with a favorable phenotype. Unfortunately, progress to date in

developing cereal cultivars with enhanced resistance to FHB-related yield loss and

mycotoxin contamination has been limited (reviewed in: Bai et al., 2003; Miedaner,

2002; Mesterházy, 2003a; Steffenson, 2003). Lack of progress using classical plant

breeding efforts has been attributed to: i) limitations associated with practical techniques

to evaluate FHB resistance within large breeding programs, ii) difficulties in establishing

consistent levels of Fusarium inoculum within FHB nurseries on a cost-effective basis,

iii) limited availability of cereal germplasm with high levels of innate resistance, and iv)

inconsistent financial support for public research (Dill-Macky, 2003; Sutton, 1982).

Sources of resistance to FHB in wheat are based on spring wheat genotypes with

innate resistance. Varieties such as: „Sumai 3‟, „Nobeoka Bozu‟, „Shanghai 7-31B‟,

„Nyubai‟, „Ning 7840‟, and „Frontana‟ are commonly use in breeding programs around

Page 42: PhD Fusarium

19

the world to develop regionally-adapted cultivars with enhanced FHB and mycotoxin

resistance (Buerstmayr et al., 1996; Diaz de Ackermann and Kohli, 1997b; Gilchrist et

al., 1997; Snijders, 1990b; etc.). Relatively few barley cultivars have been identified with

a high level of innate resistance to FHB and mycotoxin accumulation (Steffenson, 2003).

Unfortunately, most accessions from Fusarium-resistant varieties exhibit unfavorable

agronomic traits which render them largely unsuitable for widespread adoption within

FHB affected regions (Chen et al., 2000). Furthermore, most new accessions produced by

backcrossing resistant varieties with regionally-adapted germplasm tend to have limited

and often unstable FHB resistance (Mesterházy, 2003a).

2.2.4 Classifications of FHB Resistance

Classic descriptions of host-plant resistance to initial infection and to spread of

Fusarium within the spike were initially categorized as „Type 1‟ and „Type 2‟, resistance,

respectively (Schroeder and Christensen, 1963). Type 1 resistance is regarded as an

important component of head blight resistance of barley, since disease resistance given by

percentage of florets infected is inherently more important than spread from floret to

floret. Type 2 resistance is believed to be more important for wheat as FHB tends to

spread more readily from spikelet to spikelet (Bushnell et al., 2003).

Since the development of this classification system, three other types of resistance

have been postulated; „Type 3‟ resistance, which is based on an ability to limit

accumulation of trichothecene toxins in kernels (Miller et al., 1985), „Type 4‟ resistance

based on kernel infection (Mesterházy, 1995), and „Type 5‟ resistance based on increased

tolerance to yield loss in the presence of FHB (Mesterházy, 1995). Unfortunately, beyond

Page 43: PhD Fusarium

20

the descriptions of Types 1 and 2, there is little agreement on the order and general

designation of various mechanisms of FHB resistance in cereals.

2.3 Trichothecene Mycotoxins

Trichothecenes are low molecular weight sesquiterpenoid mycotoxins characterized

by a tricyclic ring structure with a double bond at C-9,10 and an epoxide group at C-12,13

(Mirocha et al., 2003) (Figure 2.7).

Trichothecene MW Oxygenation and Esterification at Position

b

R1 R2 R3 R4 R5

T-2 Toxin 466 -OH -OAC -OAC -H -OIsoval

HT-2 Toxin 424 -OH -OH -OAC -H -OIsoval

4,15-Diacetoxyscripenol 366 -OH -OAC -OAC -H -H

Nivalenol 312 -OH -OH -OH -OH =O

DON 296 -OH -H -OH -OH =O

15-AcDON 338 -OH -H -OCH3 -OH =O

3-AcDON 338 -OCH3 -H -OH -OH =O a MW = Molecular Weight (Daltons).

b OAc (Acetyl Ester = - O-CO-CH3), OIsoval (Isovalerate Ester = -OCOCH2CH(CH3)2).

Figure 2.7 Diagram of the general structure, numbering system of the tetracyclic

trichothecene nucleus, and specific side-chain groups of naturally occurring

trichothecenes of the genus Fusarium (Desjardins, 2006).

Type A and B structural classes can be grouped based on type of substituent group

at C-8 (Chelkowski, 1989; Desjardins, 2006; Foroud and Eudes, 2009; Mirocha et al.,

1

6

16

R5

10

11

6

7 8

9

R3

15

13

5

14

12

2 O

1 H

O

3

4

R2

R1

H

H

R4

Page 44: PhD Fusarium

21

2003; Perkowski et al., 1995). Type A trichothecenes (e.g., T-2 toxin, HT-2 toxin,

diacetoxyscirpenol) have hydrogen, hydroxyl, or ester groups at C-8 position. Type B

trichothecenes (e.g., nivalenol, DON, 15-AcDON) have a keto substituent at C-8 (Figure

2.7).

The name denoted to trichothecene mycotoxins is derived from the fungus

Trichothecium, which produces the natural product called trichothecin (Freeman and

Morrison, 1948). In pure crystalline form, trichothecenes tend to be colourless,

odourless, relatively soluble in polar solvents, and very stable within a wide range of

matrices (Scott, 1991; Trenholm et al., 1991; 1992).

2.3.1 Mode of Action

The biochemical mode of action and cytotoxic effects of trichothecene mycotoxins

within eukaryotic cells has been extensively reviewed (e.g., Canady et al., 2001;

Desjardins, 2006; Prelusky et al., 1994; Rotter et al., 1996; Ueno, 1983). In general,

trichothecene cytotoxicity is attributed to the disruption of peptidyl-transferase activity

given by mycotoxin binding to ribosomal protein L3 (RPL3) within the 60S subunit of

eukaryotic ribosomes (Feinberg and McLaughlin, 1989; McLaughlin et al., 1977).

Although binding to RPL3 has been specifically attributed to the presence of the C-

12,13 epoxide group common to all trichothecenes (Figure 2.7) (Carter and Cannon,

1977; Rocha et al., 2005; Rotter et al., 1996), the number and position of substituent

groups on the tetracyclic nucleus (Figure 2.7) has a profound influence on the relative

toxicity of various trichothecene mycotoxins. Given the inherent diversity of the various

side chain substituents that comprise both Type A and B trichothecenes, a wide range of

Page 45: PhD Fusarium

22

physico-chemical properties and cytotoxic effects of trichothecenes has been exhibited on

various types of eukaryotic cells (Desjardins, 2006; Desjardins and Hohn, 1997; Kuiper-

Goodman, 1994; Mirocha, 1983; Nishiuchi et al., 2006; Ohtsubo, 1983; Otokawa, 1983;

Prelusky et al., 1994; Terao, 1983; Ueno, 1983; etc.).

The presence of different side chains on the trichothecene nucleus is, therefore, a

key determinant of relative cytotoxicity values and specific cytotoxic effects for

trichothecene mycotoxins. Depending on the test system, different substituents can lead

to diverse effects including inhibition of protein synthesis during translation,

transcription, or elongation as well as various secondary effects such as inhibition of

DNA and RNA synthesis, necrosis and inhibition of cell division (Feinberg and

McLaughlin, 1989), inhibition of mitochondria enzymes, immunotoxicity (Creppy, 2002;

Sudakin, 2003).

2.3.2 Mechanisms of Fungal Protection

Although trichothecenes have no function related to the growth of the producing

organism, they are involved in the biological adaptation and ecology of Fusarium species

to various environments (Desjardins, 2006). Part of this adaptation mechanism involves

various specialized genetic adaptations by the producing organism during the biosynthesis

of trichothecenes as a means of „self-preservation‟ (Cundliffe, 1989).

Genes that encode for detoxification mechanisms and/or molecular efflux pumps to

limit cytotoxicity to the parent organism are believed to be the principal means by which

virulent Fusarium species protect themselves from trichothecene-specific cytotoxicity.

For example, the F. sporotrichioides gene Tri12, which encodes an efflux pump, is

Page 46: PhD Fusarium

23

responsible for pumping trichothecenes out of, and away from, the fungal cell (Alexander

et al., 1999). Genes to help limit the metabolic cytotoxicity of trichothecenes have also

been demonstrated as a defense response within Fusarium species. For example, the gene

Tri101 encodes an acetyl-transferase enzyme responsible for the addition of an acetyl-

ester side-chain group (R-OHCH3) to trichothecene mycotoxins such as DON as a

mechanism to reduce molecular cytotoxicity to the parent organism (Kimura et al.,

1998b).

2.3.3 Effects on Plants

The toxicity of trichothecenes to plant cells was first reported in 1961 (Brian et al.,

1961). Since this initial work, trichothecene toxins are well established as phytotoxic

compounds that produce wilting, chlorosis, necrosis, and various other non-specific

effects within plant tissue (Cutler and Jarvis, 1985, Desjardins, 2006; Nishiuchi, et al.,

2006; Packa, 1991; Wakulinski, 1989, etc.). Inhibition of ribosomal protein synthesis is a

common mechanism of phytotoxicity; however, some trichothecenes affect electron

transport chains, cellular membranes, leaf chlorophyll synthesis and other functions

within plant tissue. However, many aspects related to the specific effects of

trichothecenes within plant systems, particularly host cell defense mechanisms, remain

unknown (Desjardins, 2006).

Genetic disruption techniques to produce „knock-out‟ strains of virulent Fusarium

species have been invaluable tools to clarify the relationship between Fusarium

pathogenicity and trichothecene production within host plant cell tissue. Desjardins et al.

(1989) demonstrated that inoculation of parsnip root discs with a T-2 producing wild type

Page 47: PhD Fusarium

24

strain of F. sporotrichioides caused extensive root rot, while the Tri4 mutant, which

produced little or no trichothecene, caused significantly less root tissue damage. Similar

studies conducted with a mutant strain of F. graminearum, where the trichodiene

synthase (Tri5) gene was altered to impair trichothecene biosynthesis, were initiated to

examine the effect of trichothecene production on seedling emergence and height of

Fusarium-inoculated wheat, maize, oat and rye (Proctor et al., 1995). Relative to

trichothecene producing (Tri5+) wild-type strains, non-trichothecene producing (Tri5-)

mutant strains had reduced incidence and severity of disease (i.e., bleaching of heads,

yield reduction, etc.) and reduced fungal spread from single spikelet inoculations

(Desjardins et al., 1996; Mirocha et al., 1997; Proctor et al., 1995). Similar results were

noted by Harris et al. (1999a) who found that mutant strains of F. graminearum, which

did not produce trichothecene, were less virulent on inoculated maize. A comprehensive

summary of these as well as subsequent Tri5 disruption studies provided by Desjardins

(2006) clearly demonstrates that trichothecenes act to enhance the ability of Fusarium to

cause disease on susceptible host plants.

2.4 DON, 3-AcDON, and 15-AcDON

Most phytopathogenic F. graminearum species are believed to produce 15-AcDON

or 3-AcDON as labile, acetylated precursors to DON (Figure 2.8). Production of either

acetylated compound is geography-dependent since 3-AcDON chemotypes tend to

dominate in Asia and Europe, while Fusarium species which produce 15-AcDON have

been generally more prevalent in North America (Goswami and Kistler, 2004; Moss and

Thrane, 2004). Therefore, the accumulation of DON within plant tissue is believed to be

Page 48: PhD Fusarium

25

a metabolic process conferred by fungal and plant carboxyl-esterases, which continuously

deacetylate either 15-AcDON or 3-AcDON within plant tissue (Evans et al., 1997;

Mitterbauer and Adam, 2002; Scott et al., 1984).

Figure 2.8 Structure of DON, 15-AcDON, and 3-AcDON (Desjardins, 2006).

2.4.1 Contamination of Food and Feed

After FHB infection of cereals, DON, 15-AcDON and 3-AcDON accumulate within

grain as thermally stable and persistent contaminants through storage, handling and

processing of food and feed (Canady et al., 2001; Desjardins, 2006; D'Mello et al., 1999,

Visconti and De Girolamo, 2002). Therefore, DON and either 3-AcDON or 15-AcDON

are considered food- and feed-borne contaminants that are hazardous to human and

animal health (reviewed in Canady et al., 2001; D'Mello et al., 1999; Desjardins, 2006).

The effects of chronic exposure to DON within small-grain cereals has been widely

tested within domestic livestock. Among animals tested, swine are the most sensitive to

DON exposure (Trenholm et al., 1994). In fact, swine producers termed DON

“vomitoxin” when they associated observed emesis (vomiting) following the

Page 49: PhD Fusarium

26

consumption of Fusarium-infected maize well before the biochemical effects were clearly

evident (Mirocha, 1983). In subsequent research, more sophisticated chronic feeding

studies with swine have shown decreased skin temperature, depressed feed intake,

reduced thyroid size, altered stomach conditions, increased albumin levels, and decreased

-globulin levels after consumption of dry feed with as little as 1 - 3 mg·kg-1

DON

(Jacobsen et al., 1993; Rotter et al., 1994).

Although type B mycotoxins are among the least acutely toxic trichothecenes (e.g.,

DON, LD50 = 46 mg·kg-1

mice, oral), chronic exposure is considered a significant risk for

both humans and domestic animals (Canady et al., 2001). Most countries involved in the

export and import of grain have imposed strict guidelines to regulate the trade of DON-

contaminated cereals. In March, 2004, the Codex Committee on Food Additives and

Contaminants reaffirmed the 2001 Joint FAO/WHO Expert Committee on Food

Additives (JECFA) provisional maximum tolerable daily intake (PMTDI) for DON of 1

µg·kg-1

·day-1

for cereals for use within human food or food processing

(http://ec.europa.eu/food/fs/ifsi/eupositions/ccfac/archives/ccfac_04366_item14g_en.pdf).

This regulatory limit for DON, and closely-related mycotoxins, will undoubtedly continue

to evolve as more accurate risk-assessment models are developed based on more precise

toxicological end-points (Miller, 2008).

2.4.2 Mechanism of Virulence for FHB

DON is regarded as a principal component of the necrotrophic phase of Fusarium

pathogenesis of plant tissue (Atanasoff et al., 1994; Desjardins et al., 1996; Desjardins et

al., 2000; Desjardins, 2006; Harris et al., 1999a; McCormick, 2003; Mesterházy, 2002;

Page 50: PhD Fusarium

27

Miller, 1994; Miller et al., 1985; Mirocha et al., 1997; Mudge, et al., 2006; Proctor et al.,

1995; 1997; Snijders and Perkowski, 1990; Wang and Miller, 1987; 1988; etc.). Aside

from inhibition of protein synthesis, as previously discussed, DON can induce a range of

phytotoxic symptoms including light-dependent bleaching of detached plant tissues based

on loss of chlorophyll, carotenoids (Bushnell et al., 2002; Seeland and Bushnell, 2001),

and electrolytes within exposed plant-cell tissues (Cossette and Miller, 1995).

Electron microscopy and immunogold labeling techniques provide anecdotal

evidence of DON, 15-AcDON and 3-AcDON toxicity in Fusarium pathogenicity (Kang

and Buchenauer, 1999). Toxin diffusion into and within susceptible plant tissue occurs

well in advance of Fusarium hyphal invasion (Figure 2.9). Without exception, toxin

accumulation was well correlated with symptoms of Fusarium-related plant cell damage.

Figure 2.9 Immunogold localization of DON, 3-AcDON, and 15-AcDON within wheat

spikelet (cv. Agent) infected with F. culmorum. Toxins detected within: A,

Plant host cell wall (HCW) and ahead (see arrow) of hyphal fungal cell (FC)

1.5 days after inoculation (DAI). B, HCW of plant cells within the rachis 6

DAI. C, Parenchyma cells (PC) within a young kernel located three

spikelets above inoculated spikelets 10 DAI. Secondary thickening (ST)

noted within PC (Bars = 0.5 µm (Kang and Buchenauer, 1999).

5 µm 5 µm 5 µm

A B C

Page 51: PhD Fusarium

28

Analytical methods to measure DON concentrations have also provided valuable

input in terms of quantifying DON within developing cereal tissue. For example, Sinha

and Savard (1997) used a competitive direct ELISA technique to measure DON

concentrations within developing wheat heads and found that the rachis had the highest

DON concentrations (93 ppm), followed by floral chaff (50 ppm), kernels (25 ppm), and

peduncles (16 ppm). In a follow-up study, DON was detected below the site of injection

into single florets at concentrations of hundreds of ppm, i.e. it was found within the

kernels and rachis (Savard et al., 2000). The results of these studies are complemented by

fungal colonization experiments based upon quantitative PCR assays of plant tissue

before and after Fusarium pathogenesis. Nicholson et al. (1998) used quantitative PCR

based on oligonucleotides from known F. graminearum DNA sequences to demonstrate

greater fungal colonization of plant tissue by trichothecene-producing than non-producing

mutant strains.

2.5 Transgenic Approaches to Limit Trichothecene Toxicity

The study and subsequent application of innate and transgenic mechanisms to instill

enhanced “Type III” resistance to DON and structurally-similar trichothecenes within

higher order plants is a complex and difficult task. Many variables (both known and

unknown) affect host plant responses to Fusarium pathogenesis (Desjardins, 2006;

Foroud and Eudes, 2009; Muehlbauer and Bushnell, 2003). For this reason, novel

trichothecene-specific mechanisms are often initially evaluated within yeast and/or

Arabidopsis as model eukaryotic test systems prior to evaluation within higher order

plants. Such model eukaryotic test systems offer increased efficiency in terms of overall

Page 52: PhD Fusarium

29

convenience and decreased cost with minimal complexity, variability, and time to initial

evaluation (Mitterbauer and Adam, 2002; Urban et al., 2002).

Four general mechanisms have been proposed as transgenic approaches to limit

trichothecene, particularly DON, cytotoxicity within plants and eukaryotic test systems;

1) inhibition of trichothecene biosynthesis, 2) alteration of the target ribosomal site, 3)

metabolic trichothecene detoxification, and 4) inhibition of trichothecene biosynthesis

(McCormick, 2003; Mitterbauer and Adam, 2002). A fifth mechanism attributed to

plantibodies and mimotopes as been added to this literature review based on novel

applications of recombinant antibody engineering technology to limit mycotoxin-specific

cytotoxicity.

2.5.1 Inhibition of Trichothecene Biosynthesis

Biosynthesis of trichothecenes within in vitro test systems can be blocked by the

addition of flavonoids, furanocoumarins, and various other naturally-occurring soluble

phenolic compounds derived from plants (Bakan et al,. 2003; Desjardins et al., 1988).

For example, xanthotoxin and various other furanocoumarins, which are produced by a

variety of plants, have been shown to be effective inhibitors of trichothecene biosynthesis

at the preliminary step of trichodiene oxygenation. Unfortunately, the efficacy of such

compounds is often limited as many fungi are capable of expressing enzymes that

detoxify and limit the effectiveness of trichothecene biosynthesis inhibiting compounds

(Spencer et al., 1990).

Page 53: PhD Fusarium

30

2.5.2 Alteration of Target Protein

Given that trichothecene mycotoxins inhibit protein synthesis by binding to RPL3, a

possible mechanism to attenuate trichothecene cytotoxicity would be to develop

ribosomal genes whose products do not bind trichothecene toxins. Some of the earliest

work in this area was reported by Hobden and Cundliffe (1980) who reported ribosomal

resistance to trichothecene produced by Myrothecium verrucaria. In fact, this work

coupled with related research, initially confirmed that the RPL3 protein was the primary

target of trichothecenes (Fernandez-Lobato et al., 1990; Fried and Warner, 1981).

Harris et al. (1999b) introduced an altered RPL3 protein from rice (Oryza saliva)

into tobacco (Nicotiana tabacum) cells to instill enhanced mycotoxin resistance.

Expression of a rice cDNA encoded with a modified RPL3 based on a change from

tryptophan to cysteine at residue 258 provided enhanced trichothecene resistance within

tobacco (Harris and Gleddie, 2000; 2001, US Patent 606046). A similar approach was

reported by Adam et al. (2000) who engineered a mutant tomato Rpl3 gene to provide

resistance to DON in yeast. Unfortunately, results based on modified plant Rpl3 cDNA

expression within transgenic tobacco plants were less promising as constitutive

mycotoxin resistance was not achieved. Limited efficacy was attributed to a post-

transcriptional effect which led to a preferential utilization of endogenous RPL3 protein

as well as limitations associated with stoichiometry of RPL3 protein expression relative

to rapidly elevated mycotoxin levels within infected tissue (Mitterbauer et al., 2004).

Page 54: PhD Fusarium

31

2.5.3 Metabolic Trichothecene Detoxification

In planta detoxification of DON, and structurally similar trichothecenes, based on

expression of trichothecene detoxification and resistance genes is a rapidly-emerging and

important field of study with many practical commercial applications. Comprehensive

reviews of enhanced metabolic resistance to Fusarium mycotoxins within plants is

described in additional detail by Berthiller et al. (2007) and Boutigny et al. (2008).

In planta expression of metabolic detoxification genes such as Tri101 is a common

approach to instill enhanced trichothecene tolerance. Tri101 is a gene located outside the

principal trichothecene biosynthetic gene cluster that encodes trichothecene C-3

acetyltransferase, an enzyme which adds an acetylester group to the C-3 hydroxyl group

on trichothecenes (outlined in Section 2.3.2). Constitutive expression of Tri101 within

plants and other model eukaryotic organisms enables a metabolic detoxification of DON

to 3-AcDON, a compound with reduced eukaryotic cytotoxicity (Kimura et al., 1998a;

1998b). Tri101 has been expressed in a range of plants with promising results to show

decreased trichothecene cytotoxicity and prevention of FHB yield loss (reviewed in:

Alexander, 2008; Kimura et al., 2006). Unfortunately, expression of Tri101 alone may

not provide complete resistance as both Fusarium and plants commonly produce

deacetylation enzymes that can convert 3-AcDON back to DON. In fact, Mitterbauer and

Adam (2002) stated that the efficacy of Tri101 expression within plants could be limited

by a „futile cycling‟ whereby plant carboxylesterases, as well as fungal enzymes such as

Tri8 (McCormick and Alexander, 2002), could play a significant part in the metabolic

Page 55: PhD Fusarium

32

conversion of acetylated compounds back to more toxic C-3 hydroxyl precursors (e.g., 3-

AcDON to DON).

The identification of trichothecene detoxification genes is not solely limited to those

isolated from Fusarium species. Enzymes capable of trichothecene metabolism have also

been isolated from other organisms. For example, potato tissue can rapidly metabolize,

and limit the cytotoxicity of the trichothecene diacetoxyscirpenol based on an initial

removal of the acetyl groups followed by cleavage of the trichothecene skeleton

(Desjardins et al., 1992). In addition, constitutive over-expression of a UDP

glycosyltransferases (UGTs) isolated from Arabidopsis thaliana can also decrease plant

sensitivity to DON and 15-AcDON. Expression of UGTs enables the conjugation of

sugars to trichothecenes thereby forming less toxic DON-3O-glucoside and 15-AcDON-

3O-glucoside conjugates (Poppenberger et al., 2003). This work is supported by the

earlier work of Miller and Arnison (1986), which showed that FHB-resistant wheat

cultivars (e.g., „Frontana‟) metabolized DON to a less toxic compound based on a

biochemical conversion to at least three metabolites, one of which was a DON-glycoside

conjugate.

Another means of enhanced, and perhaps more complete, metabolic resistance may

involve the expression of bacterial genes responsible for the cleavage of the trichothecene

epoxide group at C-12,13 (Figure 2.7). Although such de-epoxidation activity has been

successfully demonstrated in bacteria (Fuchs et al., 2002), application of this technology

within plants to consistently yield favorable results remains an area of future research.

Page 56: PhD Fusarium

33

2.5.4 Reduction of Intracellular Concentrations

The identification and application of candidate genes that encode for molecular

efflux pumps may also be used to limit trichothecene cytotoxicity within plant cells. For

example, Adam and Lemmens (1996) demonstrated enhanced trichothecene resistance

based on in planta expression of the ABC (ATP Binding Cassette) transporter protein

encoded by a Pleiotropic Drug Resistance gene (i.e. Pdr5) isolated from S. cerevisae

(Balzi et al., 1994). Pdr5 is one of nine members of the (NBF-TMS6)2 subclass of ABC

encoding genes present within the yeast genome. ABC transporter proteins are located in

the plasma membrane of host cells and remove toxic substances using the energy of ATP

hydrolysis (Decottignies and Goffeau, 1997; McCormick et al., 1999).

Expression of Pdr5 genes is a promising strategy to complement other approaches.

Muhitch et al. (2000) successfully demonstrated that co-expression of the Fusarium

acetylation enzyme (TRI101), and Saccharomyces transporter protein (PDR5) resulted in

increased resistance to the trichothecene 4,15-diacetoxysciprenol during germination and

plantlet development of transgenic tobacco.

2.5.5 Plantibodies and Mimotopes

The expression of functional full-size antibodies and antibody fragments within

plants has received extensive research interest within the last 15 years. Plant-produced

antibodies, or “plantibodies” have been evaluated and applied mainly for production of

biopharmaceuticals and immunodiagnostic agents as well as biocontrol agents to improve

resistance against plant pathogens and cytotoxic herbicides (reviewed in Section 3.6.2.3).

Page 57: PhD Fusarium

34

Various antibodies with specificity to a wide range of mycotoxins are commonly

used within standard immunoassay formats (e.g., Enzyme-Linked Immunosorbent

Assays) to assess mycotoxin levels within contaminated grain (reviewed in Mirocha et al.,

2003). However, despite reports of favorable efficacy, very few have been used as

plantibodies to instill enhanced in planta resistance against Fusarium pathogenesis

(Peschen et al, 2004). Of those tested, most are Fusarium-specific plantibodies expressed

as fusion proteins to antifungal peptides (reviewed in Hu et al., 2008). In these cases, the

Fusarium-specific antibody targets the pathogen while the fusion protein destroys it.

Surprisingly, only a very limited number of recombinant plantibodies have been

applied to limit, or immuno-modulate the cytotoxic effects of low molecular weight

mycotoxins. Yuan et al. (2000) reported the first in planta expression of an anti-

mycotoxin gene. The functional, zearalenone-specific scFv fragment produced within the

cytoplasm of transgenic Arabidopsis-leaf tissue was shown to have a similar binding

affinity to a bacterially-produced scFv and its parent monoclonal Ab. Likewise, Yuan et

al. (1999) were the first to report expression of two functional anti-idiotype peptides, or

mimotope peptides for a low molecular weight mycotoxin (DON). Both mimotopes were

antagonistic to DON-induced protein synthesis inhibition and cellular damage within a

model eukaryotic test system.

These preliminary studies provide insights into the vast potential that antibody

engineering can provide to design and express new and novel peptides that reduce (or

perhaps eliminate) mycotoxin-specific cytotoxicity. The next literature review chapter

provides a comprehensive overview of antibody engineering as applied to agricultural

Page 58: PhD Fusarium

35

biotechnology. Subsequent research chapters describe experiments at the heart of this

Ph.D. project which were designed to develop, isolate and assess the efficacy of a unique

single-domain antibody fragment with specificity for the mycotoxin 15-AcDON.

Page 59: PhD Fusarium

36

3 LITERATURE REVIEW 2: ANTIBODY ENGINEERING IN

AGRICULTURAL BIOTECHNOLOGY

3.1 Introduction

Antibodies (Abs) are gamma globulin proteins produced by the immune system of

vertebrates to recognize and bind to a virtually limitless repertoire of antigens (Ags). Abs

are inherently unique, as they possess high affinity and binding specificity for a target Ag.

These properties have made Abs invaluable reagents within various biotechnological and

biomedical applications ranging from standard diagnostic immunoassays (e.g., ELISA) to

integrated immunoaffinity-based systems designed to isolate, enrich, and purify specific

target reagents (e.g., proteins, immuno-conjugates, etc.).

Initial efforts to produce in vitro Ag-specific Abs led to the introduction of

hybridoma technology in 1975 in which Ag-stimulated B-cells were fused with their

myloma counterparts to generate stable hybrid or “hybridoma” cell lines to enable large

scale production of Ag-specific monoclonal Abs (mAb) (Köhler and Milstein, 1975).

Subsequent advances in the fields of genetics, molecular biology and immunology have

greatly enhanced applications of novel technologies required for the development,

isolation, and application of recombinant Ab (rAb) fragments for an expanded range of

applications. For example, routine use of phage display technology to screen

combinatorial rAb libraries (reviewed in Winter et al., 1994; Hoogenboom, 2005)

coupled with new software tools to support an expanding structure-function database

(e.g., Harding et al., 2004) has opened unlimited opportunities to develop tailor-made rAb

fragments. Moreover, on-going improvements associated with rAb optimization and

subsequent expression in recombinant hosts offers the possibility of large-scale and low-

Page 60: PhD Fusarium

37

cost production platforms as an alternative to more expensive, laborious and often

cumbersome methods associated with the mAb development using mammalian cell lines

Antibody engineering has emerged as a new discipline focused on the identification,

cloning, optimization and expression of Ag-specific rAb genes within various prokaryotic

and eukaryotic host systems (Filpula, 2007; Holliger and Hudson, 2005; Jain et al., 2007;

Lo, 2004; Maynard and Georgiou, 2000). This review presents an overview of

established and emerging technologies available for the de novo generation of Ag-

specific, rAb fragments as an alternative to traditional immunoassays based on

monoclonal antibodies or polyclonal serum. Applications and future research needs

specific to agricultural biotechnology and genetically-modified plants are presented as

concluding remarks.

Key Words

Antibody; antigen; immunoassay; immunoaffinity; ELISA; recombinant antibody (rAb);

antibody engineering; rAb fragment; phage display; combinatorial libraries; rAb

optimization; rAb expression; Ab engineering.

3.2 Recombinant Antibody Fragments

The production of Ag-specific rAb fragments derived from full-size conventional

Abs was first reported in the late 1980‟s following the discovery of the genetic

mechanism associated with Ab gene rearrangements. Concurrent advances in subcloning

and expression systems coupled with the introduction of polymerase chain reaction (PCR)

technology further enabled the recombinant expression of rAb fragments within bacterial

hosts (initial reviews in Plückthun and Skerra, 1989; Winter and Milstein, 1991). In

Page 61: PhD Fusarium

38

general terms, the production of rAb fragments through these new processes involved the

isolation of mRNA for specific Ab genes derived from either hybridoma, spleen cells, or

lymph node leukocytes followed by reverse transcription PCR (RT-PCR) to form

complementary DNA (cDNA) and eventual PCR amplification by using gene-specific

primer sets to generate a complete Ab gene sequence (Maynard and Georgiou, 2000).

Given the inherent reliance on PCR amplification, the most significant initial

advancements were related to the application of optimized oligonucleotides or so-called

primers to anneal and amplify target rAb genes. Within ten years of adoption of PCR,

numerous primer sets were published based on N-terminal sequences of isolated rAb

genes (Benhar and Pastan, 1994), Ab leader sequences (Larrick et al., 1989), and known

variable domain framework sequences.

A wide variety of rAb fragments have been produced, characterized, and applied in

various biological systems (reviewed in Hudson, 1998; 1999; Little et al., 2000; Holliger

and Hudson, 2005; Yau et al., 2003a). The following section provides a general

description of the three most common and well documented rAb fragments with potential

applications in agricultural biotechnology.

3.2.1 Fab Fragment

The term „Fab‟, or fragment for antigen binding (Better et al., 1988), refers to a

relatively large (MW ≈ 50 – 55 kDa) heterodimer molecule comprised of two polypeptide

chains, one containing the constant and variable light chain domains (CL + VL) of a parent

Ab paired with its corresponding variable and first constant regions (CH1 + VH) (Jeffrey

et al., 1993) (Figure 3.1).

Page 62: PhD Fusarium

39

Figure 3.1 Schematic representation (left) and ribbon diagram (right) of recombinant

antibody fragment (Fab). Constant and variable light chain domains (CL +

VL) and corresponding first constant and variable heavy chain domains

(CH1 + VH), and target Ag (i.e., Digoxin) are labeled accordingly (adapted

from Joosten et al., 2003 and Jeffrey et al., 1993).

Fabs have the advantage of enhanced durability within in situ applications, as they

are inherently more stable and less prone to dissociation and proteolytic degradation

relative to other conventional rAb fragment formats (e.g., scFv). Enhanced stability is

attributed to the presence of a covalent disulfide bond which serves to retain tertiary

folding and to facilitate more extensive interface and non-covalent interactions between

the two paired immunoglobulin chains or four Ab domains (Maynard and Georgiou,

2000). Despite their relatively large size relative to other rAb fragment types, Fab

proteins are amendable to standard Ab engineering techniques to improve attributes

associated with biological function, recombinant expression, affinity for target Ag, etc.

(reviewed by: Filpula, 2007; Holliger and Hudson, 2005; Maynard and Georgiou, 2000).

3.2.2 scFv Fragment

The term single-chain variable fragment (scFv) refers to a rAb fragment in which

the variable heavy and light domains (VH and VL, respectively) of a parent

immunoglobulin (reviewed in Chapter 1) are joined by a hydrophilic and flexible peptide

Page 63: PhD Fusarium

40

linker (Figure 3.2) (Bird et al., 1989; Huston et al., 1988). The order of the domains can

be designed for expression as either VH-linker-VL or VL-linker-VH (Huston et al., 1991;

1995). In most cases, the length of the peptide linker is 15 amino acids based on a

(Gly4Ser)3 configuration (reviewed in Huston et al., 1995).

Figure 3.2 Schematic representation (left) and ribbon diagram (right) of recombinant

single chain variable fragment (scFv) antibody. Variable light and heavy

chain domains (VL and VH, respectively) and synthetic peptide linker are

labeled accordingly (adapted from Joosten et al., 2003 and Filpula, 2007).

Specific length and composition of the peptide linker can have a dramatic impact on

post-translational folding and valency of scFv fragments (Robinson and Sauer, 1998;

Turner et al., 1997). For example, Atwell et al., (1999) demonstrated that three amino

acid linkers often form scFv dimers whereas linkers with two residues or less induce the

formation of trimers. In general, increasing the length of linker will lead to monomeric

variants. However, monomeric and multimeric scFv variants have been reported with

linkers of up to 30 residues (Desplancq et al., 1994; Nieba et al., 1997).

Page 64: PhD Fusarium

41

Once valency is accounted for, one cannot assume that all scFv fragments possess

comparable Ag-binding affinity to larger Ab formats (e.g., mAb and Fab) because they all

have the same VH / VL domain pairing. Numerous publications, based on various Ags,

have demonstrated that relative scFv affinity is best described on case by case basis. For

example, Choi et al. (2004) noted that a scFv fragment raised against deoxynivalenol

(haptenic Ag, MW ≈ 296 Da) had a binding affinity that was approximately two orders of

magnitude less than the parent mAb. However, based on a similar comparison for the

same Ag, Wang et al. (2007) noted no differences between mAb and scFv binding

affinity.

Similar variability has been noted for scFv stability and efficiency of expression. In

general terms, post-translational scFv folding and function is greatly influenced by

specific expression systems and growth conditions (i.e., choice of expression vector and

recombinant host, etc.) as well as method of purification. In addition, factors such as

proteolytic degradation of the linker sequence (Whitlow et al., 1993) and formation of

insoluble inclusion bodies (Choi et al., 2004) have great influence on scFv stability and

expression.

Various techniques have been used to overcome such limitations. For example,

Reiter et al. (1994) reported on the formation of disulfide-stabilized Fvs (i.e., dsFvs)

through the introduction of cysteine residues between the VL and VH domains which

resulted in the formation of an inter-chain disulfide bond which thereby conferred greater

rAb stability. As well, molecular chaperones have been used as part of scFv expression

Page 65: PhD Fusarium

42

strategy to assist in post translational scFv folding and to increase efficiency of

expression (reviewed in Arbabi-Ghahroudi et al., 2005).

3.2.3 VHH Fragment

The term “VHH” refers to the variable domain of camelidae family heavy chain Abs

(HCAbs). VHH fragments are among the smallest intact Ag-binding fragments known (

average 14 to 15 kDa, 118 - 136 amino acid residues) (Figure 3.3).

Figure 3.3 Schematic representation (left) and ribbon diagram (right) of recombinant

HCAb variable domain (VHH) of antibody “AMD9”. CDR 1, 2, and 3 in

ribbon diagram are labeled accordingly (adapted from Joosten et al., 2003

and Desmyter et al., 2002).

The hallmark of VHH domain resides in key hydrophobic to hydrophilic amino acid

substitutions at former VL interface which imparts increased solubility and stability (e.g.,

resistance to aggregation). Despite the absence of VL and VH combinatorial diversity,

single domain VHH fragments have demonstrated high affinity binding to a wide range of

Ag types (Ghahroudi et al. 1997; Muyldermans, 2001). HCAbs are believed to

compensate for the loss of VL domain through extended CDR3 loop regions and a higher

rate of somatic hypermutations to form a VHH paratope that can form a large antigen-

Page 66: PhD Fusarium

43

binding repertoire and additionally can penetrate into and conform to clefts and cavities

of Ag epitopes (reviewed in Muyldermans, 2001). An example of this unique ability is

VHH binding to active sites of enzymes that are not typically recognized by conventional

rAb fragments (e.g., Desmyter et al., 1996; 2002). Numerous reviews are available which

document Ag binding characteristics and potential applications of VHH fragments (e.g.,

Holliger and Hudson, 2005; Joosten et al., 2003; Muyldermans, 2001).

Single domain VHH fragments offer a number of advantages relative to larger and

more complex rAb fragments such as scFv or Fab. DNA encoding the variable domain of

HCAb can be readily isolated from peripheral leukocytes and subcloned into various

phage display vectors for highly efficient VHH library generation since no synthetic linker

is required for the pairing of VH-VL domains (Ghahroudi et al., 1997). Another beneficial

characteristic of VHH is its hydrophilic amino acid substitutions within FRs

(Muyldermans, 2001). Consequently, expression in hosts such as E. coli tends to produce

high yields of soluble, stable VHH protein that is inherently free of problems associated

with protein aggregation and proteolytic degradation common to scFv expression

(Arbabi-Ghahroudi et al., 2005). Finally, recent experiments have demonstrated that

VHH fragments possess enhanced functionality and stability over a range of adverse

temperature and solvent regimes where current immunoassays do not typically function

(e.g., Goldman et al., 2006; Ladenson et al., 2006).

The understanding of amino acid sequences of VHH fragments provides interesting

and useful techniques to improve expression of non-dromedary Abs. For example, it is

possible to replace hydrophobic residues on the VH side of a conventional rAb fragment

Page 67: PhD Fusarium

44

with „VHH-like‟ residues to minimize limitations associated with insolubility and non-

specific binding. This process is known as „camelization‟ and can be used with VH

fragments derived from various animal sources (Davies and Riechmann, 1994; Davies

and Riechmann, 1996a; Tanha et al., 2001).

3.3 Development and Types of Antibody Libraries

The concept of Ab libraries was established following the elucidation of the genetic

mechanisms associated with the creation of immunoglobulin gene diversity (Tonegawa,

1983) and the advancement of technologies associated with PCR for amplification of the

variable domains of immunoglobulin genes (Orlandi et al., 1989; Songsivilai et al., 1990).

In principle, rAb libraries may be classified as immunized, naïve, semi-synthetic, or

synthetic according to the genetic material used for library construction.

3.3.1 Immunized Libraries

Immunized rAb libraries are constructed from DNA isolated from peripheral

leukocytes of a host that has been repeatedly exposed to a single target immunogen or

group of immunogens. Library construction occurs through a stepwise process whereby

the B cell genes encoding Ab variable domain regions are amplified by PCR and

subcloned into vectors to enable expression of different rAb fragment formats including

VHH (Ghahroudi et al., 1997; Goldman et al., 2006; Ladenson et al., 2006), scFv

(Clackson et al., 1991; McCafferty et al., 1990) and Fab (Huse et al., 1989; Orum et al.,

1993; Persson et al., 1991).

The inherent advantage of an immunized library format resides in the fact that

peripheral leukocytes, and therefore high concentration of corresponding mRNA used for

Page 68: PhD Fusarium

45

library construction, are the product of the process of affinity maturation (Hoogenboom et

al., 1998). Although the development of a rAb library based on an immunogen-specific

response improves the probability of isolating Ag-specific rAbs, overall library size and

diversity is often limited relative to other non-biased library formats (Burton et al., 1991;

Clackson et al., 1991).

Depending on the nature of the Ag and overall test system, the development of

hyper-immunized libraries can present a number of technical challenges and potential

drawbacks associated with isolating rAbs against self-Ags or against Ags that are

extremely toxic or chemically unstable. Immunological tolerance is another factor which

can reduce the diversity of natural in vivo repertoire. In addition, one must account for

the various logistical, financial and ethical considerations associated with the

immunization protocol to ensure a robust and predictable response to the Ag of interest

(Willats, 2002; Hoogenboom, 1997).

3.3.2 Naïve Libraries

Naïve libraries are prepared according to the same methodology used for

construction of immune libraries except that the animal has not been hyperimmunized

with the Ag of interest. The major source of immunoglobulin genes are peripheral blood

lymphocytes (PBLs), bone marrow, tonsils, and spleen cells. IgM is the preferred

immunoglobulin isotype as it is more diverse and has not been subjected to bias or

previous Ag selection (Dörsam et al., 1997; Griffiths et al., 1993; Little et al., 1993;

Marks et al., 1991; Schier et al., 1995).

Page 69: PhD Fusarium

46

Library size plays a vital role in successfully isolating Ag-specific rAb fragments.

This concept has been proven in several reports in which naïve libraries of medium size

have only resulted in the isolation of Abs with micromolar affinities (Marks, et al., 1991;

Griffiths et al., 1994). Accordingly, a library size of approximately 1010

is likely to yield

Abs with affinities in the low nanomolar range (Vaughan et al., 1996). These results

demonstrate that size and heterogeneity of such libraries have critical roles in isolating

high affinity Abs without prior immunization. An advantage of naïve libraries, in

particular larger-sized ones, is that they are often not antigenically pre-disposed to self-

antigens. As such, large, diverse naïve libraries are a fit as a universal source of Ab

binders (Vaughan et al., 1996).

3.3.3 Synthetic / Semi-Synthetic Libraries

Methodologies associated with synthetic libraries combine the advantages of naïve

libraries (e.g., structural diversity, etc.) with on-going advancements associated with

improved knowledge of rAb structure and function. Accordingly, these technologies

enable in vitro production of high affinity rAbs with application against a range of target

Ags that are not affected by inherent controls which may limit expression or bias an

immunogenic response to a target Ag (Fellouse et al., 2007; Knappik et al., 2000; Winter

and Milstein, 1991).

One of the first semi-synthetic libraries was generated by using a repertoire of VH

genes, combined with a synthetic CDR3 construct, and displayed on phage surface as

either scFv (Hoogenboom et al., 1992) or Fab (Barbas et al., 1992) fragments. The

complexity and diversity was further expanded using a repertoire of VL genes in

Page 70: PhD Fusarium

47

conjunction with initial library formats (de Kruif et al., 1995; Nissim et al., 1994).

Second generation libraries evolved from the development of synthetic Ab scaffolds that

express well in E. coli to help overcome limitations associated with post-translational rAb

folding (Knappik and Plückthun, 1995). It is also possible to construct fully synthetic

rAb libraries using available DNA sequences of human immunoglobulins (Andris et al.,

1995; Cook and Tomlinson, 1995; Tomlinson et al., 1992; Winter, 1998) followed by

total gene synthesis for expression in E. coli based on optimized rAb scaffolds (Griffiths

et al., 1994; Knappik et al., 2000; Krebs et al., 2001).

3.4 Display Systems

High throughput screening of rAb libraries is made possible through techniques that

establish a direct physico-chemical link between a gene (i.e., genotype) and the resultant

rAb fragment (i.e., phenotype). This linkage allows isolation of an Ab fragment with

desired Ag binding properties from a large, pooled rAb repertoire, while the genetic

material is available for further isolation and engineering. rAb fragments are most

commonly displayed on phage, bacteria, yeast, and ribosomes (Li, 2000; Yau et al.,

2003a).

3.4.1 Phage Display

Phage display was first demonstrated by Smith (1985) who reported expression of

foreign peptides as a fusion with pIII viral coat protein on the surface of non-lytic

filamentous phage. Within five years of Smith‟s demonstration of a direct link between

phenotype, i.e., as protein expressed on the phage surface, to genotype, i.e., as foreign

DNA within phage genome, McCafferty et al. (1990) used phage display techniques to

Page 71: PhD Fusarium

48

isolate Ag-specific scFv fragments from a diverse filamentous phage-scFv display

repertoire. Since the time of these reports, phage display technology has matured into a

widely adopted technique for selection of rAb fragments from very large phage-display

libraries.

Phage display techniques are now commonly used to produce Ab fragments with

optimized binding affinity, stability, activity (reviewed by Forrer et al., 1999) and to

isolate Abs which were previously considered to be very difficult to obtain through

conventional methods (Griffiths et al., 1993; Hoogenboom and Chames, 2000).

Numerous reviews highlight the diverse applications of phage-display technologies (e.g.,

Hoogenboom et al., 1998; Paschke, 2006; Smith and Petrenko, 1997; Willats, 2002; etc.).

3.4.1.1 Filamentous Bacteriophage

Filamentous bacteriophage (Ff) are viruses (e.g., F1, M13, fd, etc.) that can infect

and replicate within gram-negative bacteria (e.g., E. coli) without killing the host cell

(Russel et al., 2004). These viruses are ideal for applications within rAb engineering

since the Ff genome is inherently small and tolerates insertions into non-essential regions

without loss of bacterial infectivity. During phage assembly, fusion proteins are

expressed on a particular viral coat protein while genetic information encoding the

displayed protein is packaged within the single-stranded DNA (ssDNA) of nascent phage

particles (Smith and Pertrenko, 1997). Direct genotype to phenotype coupling ensures

that phage produced within the same infected bacterial cell are comprised of identical

clones (Paschke, 2006).

Page 72: PhD Fusarium

49

The defining characteristic of Ff structure is a circular 6.4 kbp ssDNA genome

encased by a long, flexible tube comprised of ≈ 2,700 copies major coat protein (pVIII),

with a minor coat proteins (pIII, pVI, pVII, and pIX) expressed at phage tips (Figure 3.4)

(Yau et al., 2003a). Of these coat proteins, pIII is the most extensively used and well

characterized phage-display format since it is more amendable than the other coat

proteins to large protein insertions without loss of phage infectivity (reviewed in

Hoogenboom et al., 1998; Hoogenboom and Chames, 2000; Russel et al., 2004; Smith

and Pertrenko, 1997; etc.).

3.4.1.2 Phage vs. Phagemid Vectors

Phage display rAb libraries are constructed by cloning a diverse PCR amplified Ab

repertoire into either a phage or phagemid vector (Hoogenboom et al., 1992; Winter et al.,

1994). Regardless of library type (i.e., phage or phagemid), the salient goal is to

construct a heterogeneous and robust mixture of phage clones which collectively display

rAb–phage fusions capable of target or “Ag-specific” recognition and replication to

produce identical progeny phage in large scale (Smith and Petrenko, 1997).

Page 73: PhD Fusarium

50

Figure 3.4 Schematic representation of a filamentous bacteriophage and its protein

coats (adapted from Willats, 2002; Yau et al., 2003a).

Both phage and phagemid vectors are designed so that the target rAb gene is

inserted between a signal sequence and the coat protein gene (e.g., pIII). Likewise, both

types of vectors carry antibiotic resistance selectable markers, as well as a phage-derived

origin of replication to enables packaging of foreign DNA into viral ssDNA and

production of nascent phage progeny which display rAb fused to surface phage coat

protein (Figure 3.5) (Russel et al., 2004; Smith and Pertrenko, 1997).

Phage vectors are derived directly from the Ff genome whereby the recombinant

protein is cloned as a fusion to phage coat protein (e.g., pIII) for production of multivalent

rAb phage protein progeny. Conversely, a phagemid vector is a plasmid-based sequence

harbouring gene III and intergenic region of Fd phage fusion gene under control of a weak

to moderate bacterial promoter such as LacZ. Phagemid vectors also have a second

Page 74: PhD Fusarium

51

origin of replication to allow propagation as a plasmid within host E. coli (Figure 3.5).

Many phagemid vectors also have an amber stop codon inserted between end of desired

protein and pIII gene to enable soluble rAb expression within non-suppressor strains of E.

coli (e.g., HB2151) (Russel et al., 2004; Smith and Pertrenko, 1997).

Figure 3.5 Phage (left) and phagemid (right) vectors to construct rAb phage display

libraries through the minor coat protein pIII. Ff ori = filamentous phage

origin of replication. Plasmid ori = plasmid origin of replication. TAG =

Amber stop codon. Sig. = filamentous phage leader sequence (adapted

from Russel et al., 2004).

Phage production using phagemid vectors is only possible when additional phage

proteins are provided by helper phages through a process known as “phage-rescue”

(reviewed within Paschke, 2006, etc.) (Figure 3.5). Use of helper phage is, therefore,

Page 75: PhD Fusarium

52

very critical as improves the relative proportion of univalent hybrid phage progeny

carrying rAb-phagemid genome to wild-type or “bald” phage based only on phage

genome (Russel et al., 2004). Helper phage can also impact resultant phage valency

within pIII phagemid systems to promote production of monovalent phages carrying only

one rAb fragment pIII fusion protein per phage particle (Paschke, 2006; Russel et al.,

2004).

Finally, regardless of vector type, during library construction, it is essential to

maintain stringent control of fusion protein expression using controllable bacterial

promoter such as LacZ (for example by using Glucose as repressor) through selectable

markers and consistent techniques during all propagation steps as continuous expression

of rAb-phage coat fusion proteins increases the metabolic burden to host E. coli cells.

Such techniques are important to ensure consistent selection pressure to limit any

ecological advantage for non-functional clones which limit library efficiency and function

(Paschke, 2006).

3.4.1.3 Panning

The selection of Ag-specific Abs from a phage display library occurs through a

procedure known as „bio-panning‟ or simply „panning‟. Although this process can

involve many variations and modifications, it relies mainly on the stepwise isolation and

amplification of phage displaying Ag-binding Ab fragments (reviewed in Maynard and

Georgiou, 2000; Paschke, 2006; Smith and Pertrenko, 1997; etc.).

After exposure of the full phage-display rAb repertoire to the target Ag (e.g., coated

onto a solid surface or present in solution or even on the cell surface), washing steps are

Page 76: PhD Fusarium

53

used to remove phage carrying non-specific Abs from those displaying Ag-specific Abs.

The latter are desorbed through low or high pH elution prior to amplification in phage

host (typically E. coli) (Figure 3.6). After each round of panning, amplified phage are

subject to increased selection pressure and washing to continue phage enrichment and

amplification based on Ag-binding properties of phage-displayed rAbs (Paschke, 2006).

Figure 3.6 Phage display panning cycle: 1.) rAb-displaying phages are added to

immunotubes coated with target Ag; 2.) Washing is performed to remove

non-binding phages; 3.) Positive-binding phage are eluted; 4.) Eluted

phage are used to infect E. coli; 5.) Positive clones are plated as E. coli

plaques or colonies; 6.) Helper phage (if phagemid system) is added for

phage rescue to amplify positive clones; 7.) Positive phage are purified for

reintroduction into next round of panning (de Bruin et al., 1999).

High affinity phage display Abs are commonly selected within three to four rounds

of panning (de Bruin et al., 1999). After panning, rAb fragments are typically expressed

in E. coli and purified for subsequent characterization, and application.

Page 77: PhD Fusarium

54

3.4.2 Bacteria Display

Peptide display on the surface of bacteria was first described well over a decade ago

(Georgiou, et al., 1993). Since the initial reports of recombinant peptide expression in

prokaryotes, fully functional Ab fragments have been displayed on surface of E. coli

(Francisco et al., 1993; Fuchs et al., 1991). As noted for phage display, bacterial display

libraries can also be used to select rAbs with improved affinity for target Ag (Daugherty

et al., 1998; 1999).

A limitation associated with the expression of foreign peptides on the surface of

gram-negative bacteria (e.g., E. coli) is the physiological barrier associated with an

extensive network of macromolecules within the bilayer cell envelope. However, this

limitation can be circumvented: peptidoglycan chaperones have been used to facilitate

transport and anchoring of recombinant Ab fragments through both the inner and outer

membranes without causing an adverse effect on cell growth and viability (Fuchs et al.,

1991). Another way to address this problem is to select an appropriate leader sequence

and a surface-localized carrier protein (Yau et al., 2003a).

It is also possible to express rAbs on the surface of gram-positive bacteria. For

example, scFv expression has been reported on the surface of Staphylococcus xylosus and

S. carnosus (Gunneriusson et al., 1996) Ab display on gram-positive bacteria has been

shown to provide efficient rAb secretion and post-translational folding.

3.4.3 Yeast Display

As in bacterial surface display, the phenotype and genotype linkage for yeast surface

display models is comprised by the protein on the cell surface and the corresponding

Page 78: PhD Fusarium

55

DNA within the cell. Given this direct linkage, an additional subcloning step is not

required after selection of an Ag-binding cell (Yau et al., 2003a).

Yeast display of rAbs offers many of the advantages of bacterial display. Yeast has

a thick, rigid cell wall which enables stable maintenance of surface-displayed proteins

(Georgiou et al., 1997). In contrast to typical prokaryotic expression systems (e.g., E.

coli), yeast possess protein folding and secretory mechanisms of higher order eukaryotic

(e.g., mammalian) systems. As such, yeast display alleviates some biases associated with

prokayotic protein expression and folding (Boder and Wittrup, 1997).

Despite these advantages, experience has shown that yeast-display Ab libraries tend

to have limited transformation efficiency, leading to smaller and less diverse repertoires

relative to phage or bacterial display (Yau et al., 2003a). Nevertheless, high affinity rAb

fragments have been isolated from yeast display libraries. For example, Boder and

Wittrup (1997) successfully used yeast-surface display to screen a diverse peptide library

to select binders to a soluble fluorescein tracer. Likewise, Kieke et al., (1997) reported

successful isolation of an anti-T cell receptor scFv using a yeast-surface display library.

3.4.4 Ribosome Display

Ribosome display represents an alternative to expression of Ab fragments on the

surface of phage, yeast or bacterial cells. As in other display systems, ribosome display

libraries are designed based on a direct link between peptide / Ab phenotype and

genotype. Unlike other library formats, ribosome display is entirely in vitro or cell-free

(reviewed in Hanes and Plückthun, 1997; Schaffitzel et al., 1999). Ribosome display is

unique as the phenotype to genotype construct is designed to include a spacer sequence

Page 79: PhD Fusarium

56

with no stop codon such that, when the nascent Ab fragment is translated from mRNA,

both the mRNA and the Ab remain bound to the ribosome molecule to form an „ARM‟

(Antibody-Ribosome-mRNA) complex (Figure 3.7) (He and Taussig, 1997).

Figure 3.7 Ribosome display panning cycle. 1.) Double-stranded DNA (dsDNA)

transcription to form messenger RNA (mRNA). 2.) In vitro translation of

mRNA to form ARM (Antibody-Ribosome-mRNA) complexes. 3.)

Exposure of ARM complexes to immobilized target Ag for affinity

selection or Ag-specific panning. 4.) Elution and isolation of mRNA from

Ag-specific ARM complex(es). 5.) Reverse transcription PCR

amplification of target mRNA to form dsDNA for continued panning or

transformation into host organism (e.g., E. coli) (adopted from Yau et al.,

2003a).

A key advantage of cell-free systems is that very large libraries can be handled since

transformation efficiency is not a limiting factor as it is for other systems. In addition, in

vitro systems are not impacted by ongoing random mutation of foreign DNA imparted by

Page 80: PhD Fusarium

57

the host organism which may limit library diversity. As an example, the typical size limit

associated with transformation efficiency for bacterial display libraries ranges from 1010

1011

clones (Yau et al., 2003a). With ribosome display, however, libraries with up to 1015

members can be created and screened (Roberts, 1999).

Ribosome display also enables the incorporation of continuous diversification

during each panning cycle using PCR-based mutagenesis. As a result, ribosome display

is a good format to select Ab fragments with improved KD (dissociation constants) and

robust Ag-binding affinities (Yau et al., 2003a). For example, Hanes et al. (1998)

reported the isolation of scFvs from a ribosome-display library with affinities to target Ag

in the picomolar range.

3.5 Optimization of Recombinant Antibodies

Ab fragments such as scFv, Fab, and VHH often display lower affinity than their

parental, full-size counterparts (Adams and Schier, 1999; Huston et al., 1996). A number

of strategies can be used to overcome this limitation, thus enabling isolation of high-

affinity Ag-specific rAb fragments. For example, panning procedures vs. target Ag can

be optimized and in vitro mutagenesis (where the natural affinity maturation process is

mimicked) can be performed (Adams and Schier, 1999).

3.5.1 Optimized Panning Procedures

Although optimization of the selection or panning process (Figure 3.6) does not

inherently improve rAb characteristics, it allows for more efficient recovery or selection

of high affinity Ag-specific rAb fragments from a diverse library. Panning strategies used

to increase the probability of isolating high-affinity rAbs have generally consisted of

Page 81: PhD Fusarium

58

gradually decreasing the Ag concentration during successive rounds of panning (Strachan

et al., 2002). In the case of panning against low molecular weight haptenic Ags, panning

strategies often use soluble or free hapten to elute the rAb from target Ag. Likewise,

structurally-similar hapten analogues can be preincubated with the phage-display rAbs

prior to exposure to the target hapten conjugate. Finally, subtractive panning, where the

rAbs are incubated with the carrier protein prior to incubation with the hapten, can also be

used to improve the probability of selecting Ag-specific, high affinity rAbs (reviewed in

Sheedy et al., 2007).

As more knowledge is gained on Ab structure through X-ray crystallography,

specific mutations can be targeted to modify and improve rAb specificity and affinity for

target Ag (Korpimäki et al., 2003; Lamminmäki et al., 1999). Ab engineering provides

excellent tools to tailor the properties of rAb fragments with respect to affinity, specificity

and performance for different applications (Valjakka et al., 2002).

3.5.2 In vitro Affinity Maturation

Because of the relatively low affinity of rAbs isolated from naïve libraries, the

process of somatic hypermutation must be mimicked through antigen-driven affinity

maturation (Irving et al., 1996). In vitro affinity maturation by site-directed or random

mutagenesis is not limited by the restrictions inherent to the natural somatic

hypermutation mechanisms (Parhami-Seren et al., 2002). Affinity maturation in vitro

can, therefore, generate extended and functional variability in rAb molecules (Borrebaeck

and Ohlin, 2002), and lead to selection of rAb fragments with increased affinity and

specificity for target Ag (Roberts et al., 1987; Wu et al., 1998; 1999).

Page 82: PhD Fusarium

59

Strategies to affinity mature Abs and fragments thereof in vitro can be grouped into

the following categories: random mutagenesis, site-directed mutagenesis, and shuffling.

Random mutagenesis consists, as its name implies, of the introduction of mutations

randomly throughout the gene. Random mutagenesis can be subgrouped into error-prone

PCR and bacterial mutator strains. Site-directed mutagenesis includes mutational hot

spots and parsimonious mutagenesis, as well as any mutational strategy which assigns the

mutations to given positions or residues within an Ab‟s wild-type gene. Shuffling

encompasses chain shuffling, DNA shuffling and staggered extension process. The most

frequently used strategies are those of site-directed mutagenesis, where mutations are

directed to specific CDRs or framework regions (Irving et al., 1996), and error-prone

PCR, where mutations are randomly introduced across the gene.

3.5.2.1 Random Mutagenesis

3.5.2.1.1 Error-Prone PCR

Error-prone PCR uses low-fidelity polymerization PCR conditions to introduce

point mutations randomly over a gene sequence (Stemmer, 1994). The introduction of

random mutations in vitro is an efficient method for increasing both specificity and

affinity, as long as a strong selection pressure is applied during the panning stage

(Miyazaki et al., 1999).

3.5.2.1.2 Bacterial Mutator Strains

Affinity maturation of rAbs and increase in their expression levels has been

achieved by using E. coli mutator cells (Coia et al., 2001a; 2001b; Irving et al., 1996;

2002). This approach consists of the selection of rAbs from a library followed by gene

Page 83: PhD Fusarium

60

diversification or mutagenesis through amplification in a bacterial mutator strain of E.

coli such as MutD5, a conditional mutant which produces single base substitutions

(transversions or transitions) at high frequency compared to normal E. coli cells. Such

mutator strains can produce a large number of mutant rAbs which can then be selected by

phage-display or other methods (Irving et al., 1996). For example, the affinity of an scFv

for the glycoprotein glycophorin was increased by passaging the rAb construct through

such a bacterial mutator strain. A single point mutation resulted in a 10-fold increase in

the scFv binding affinity, and another point mutation located in a framework region near

the CDR3 of the VL resulted in a 1,000-fold increase in affinity (Irving et al., 1996). The

effects of these point mutations illustrate the power of random mutagenesis in generating

high-affinity mutants that could not be obtained by site-directed mutagenesis. However,

four to ten rounds of mutation or passage through mutator cells are required to obtain

high affinity mutants (Coia et al., 2001a).

3.5.2.2 Focused Mutagenesis

3.5.2.2.1 Site-Directed Mutagenesis

Improvements in affinity can be achieved in vitro by site-directed mutagenesis,

where specific or selected residues are mutated at predetermined locations within target

rAb genes. Following mutagenesis, a library is constructed and screened for high-affinity

mutants. For example, Davies and Riechmann (1996b) have investigated the effect of

randomizing CDRs 1 and 2 residues in VH domains specific for both haptens and protein

antigens. This research was successful in terms of generating randomized repertoires

which were displayed on phage and affinity selected to improve Ag binding.

Page 84: PhD Fusarium

61

3.5.2.2.2 Mutational Hot Spots

Chowdhury and Pastan (1999) have developed a strategy that enables the isolation

of rAb fragments with increased affinity from small phage-display libraries. Their

approach is based on the fact that the DNA encoding the variable domains of Abs

contains “mutational hot spots”, or nucleotide sequences naturally prone to

hypermutations during the in vivo affinity maturation process. Several types of hot spots

have been suggested, such as direct and inverted repeats, palindromes, secondary

structures and certain consensus sequences (Chowdhury and Pastan, 1999). Several

consensus hot spot sequences have been studied in great details; one of these sequences is

the tetranucleotide A/G-G-C/T-A/T (Chowdhury and Pastan, 1999). The serine codons

AGY, where Y can be either C or T, are other hot spot consensus sequences found in Ab

genes that can be used as targets for mutagenesis.

Hot spot mutagenesis techniques have been used to affinity mature rAb fragments

as a precursorary step to develop Ag-specific high affinity rAbs (e.g., Chowdhury and

Pastan, 1999; Li et al., 2001). For example, hot spot mutagenesis was used to generate a

scFv fragments with high affinity to mesothelin, a cancer protein Ag. The affinity of the

mesothelin-specific scFv PE38 was found to be 11 nM (Chowdhury and Pastan, 1999).

Improved affinity was desired to increase its cytotoxicity so that it could serve as a potent

immunotoxin. DNA sequences present in the rAb variable domains (hot spots) were

selected and random mutations were introduced in the light chain CDR3 (Chowdhury and

Pastan, 1999). Thirty-two hot spots were identified in the scFv, 14 being located in the

variable heavy domains, and 18 in the variable light domains. A library with the affinity-

Page 85: PhD Fusarium

62

matured clones was constructed and panning of the library yielded several mutants with a

15- to 50-fold increase in affinity. Mutagenesis of the same original library but outside of

the hotspots resulted in a mutated library from which the highest-affinity clone selected

only had a four-fold increased affinity compared to the wild-type Ab (Chowdhury and

Pastan, 1999).

3.5.2.2.3 Parsimonious Mutagenesis

Parsimonious mutagenesis was developed by Balint and Larrick (1993) as a

technique whereby all three CDRs of a variable region gene are simultaneously and

thoroughly searched for improved variants in libraries of manageable size (Balint and

Larrick, 1993; Schier et al., 1996a; 1996b; 1996c). In parsimonious mutagenesis,

synthetic codons are used to mutate about 50 % of all targeted amino acids while keeping

the other 50% of targeted residues intact (wild type) (Chames et al., 1998). rAb libraries

can be constructed with low-redundancy “doping” codons and biased nucleotide mixtures

designed to maximize the abundance of combining sites with predetermined proportions

of pre-selected sets of alternative amino acids (Balint and Larrick, 1993).

This technique has several advantages over standard mutagenesis procedures, one of

which is the total number of substitutions per Ab gene is reduced thereby resulting in a

larger number of well-folded and potentially active binders in the library (Chames et al.,

1998). Random mutagenesis may target residues essential to Ag-binding, leading to a

library of mutants that will be mostly non functional, whereas parsimonious mutagenesis

will preserve these essential residues, resulting in a library of mainly functional clones

(Chames et al., 1998).

Page 86: PhD Fusarium

63

3.5.3 Optimized Panning Procedures

Shuffling of rAb genes can be accomplished in several ways: chain shuffling, DNA

shuffling and staggered-extension process (StEP), and through variations of these

techniques. Chain shuffling consists in shuffling heavy and light chain variable regions

of Abs. In DNA shuffling, the Ab DNA is digested with DNase I, and randomly

reassembled and amplified by PCR. StEP is also a PCR-based method, where template

switching, caused by shortened extension time, shuffles various portions of several

parental Ab genes.

3.5.3.1 Chain Shuffling

Chain shuffling can generate high-affinity rAbs (e.g., scFv) from immunized

animals quite rapidly (Kang et al., 1991). Since shuffling approaches mimic somatic

hypermutation, they are believed to be more efficient than random or site-directed

mutagenesis in producing functional rAbs (Ness et al., 1999; 2002). The heavy and light

chains of rAbs isolated from an immune library can be recombined, thereby generating a

vast number of functional rAbs from an initially limited genetic repertoire (Kang et al.,

1991).

For chain shuffling to be an effective mutagenesis strategy, the pre-selection of Ag-

binding rAbs from an immune library is required prior to invoking the shuffling process.

In other words, chain shuffling is only feasible to improve rAb affinity within immunized

libraries (Park et al., 2000). Accordingly, chain shuffling is not applicable to naïve rAb

libraries since heavy and light chains available within the library have not been exposed

to target Ag. Moreover, experiments have shown that individual chains might

Page 87: PhD Fusarium

64

discriminate among partners, rendering the shuffling process difficult between naïve and

somatically mutated chains (Kang et al., 1991).

3.5.3.2 DNA Shuffling by Random Fragmentation and Reassembly

DNA shuffling (Stemmer, 1994; Stemmer et al., 1995) is based on repeated cycles

of point mutagenesis, recombination and selection, which should allow in vitro molecular

evolution of complex sequences such as proteins. It somewhat mimics the natural

mechanisms of molecular evolution (Minshull and Stemmer, 1999), but with a faster rate

(Patten et al., 1997; Stemmer, 1994).

The method involves the digestion with DNase I of a large Ab gene to create a pool

of random DNA fragments. These fragments can then be reassembled into full-length

genes by repeated cycles of annealing in the presence of DNA polymerase (Stemmer,

1994). The fragments prime each other based on homology, and recombination occurs

when fragments from one copy of a gene prime another copy, causing a template switch.

Because of the template switching events, a certain level of homology is required among

the parental genes to be suitable for this method (Stemmer, 1994).

DNA shuffling offers several advantages over more traditional mutagenesis

strategies. Compared to methods such as error-prone PCR and site-directed mutagenesis,

DNA shuffling can be used with longer DNA sequences, and also allows for the obtention

and selection of clones with mutations outside of the binding or active site of the protein,

whereas site-directed mutagenesis is limited to a given region of the protein due to the

limitation in library size that can be efficiently transformed to construct the mutant library

(Stemmer, 1994).

Page 88: PhD Fusarium

65

DNA shuffling was first performed with 1 kb-long interleukin 1β genes. The genes

were broken into 10- to 50-bp fragments and reassembled to their original size and

function (Stemmer, 1994). Sequencing of clones following the shuffling process revealed

that reassembly introduced point mutations at a rate of 0.7%, a rate similar to that of

error-prone PCR. A similar method was used to shuffle β-lactamase genes, where three

cycles of shuffling followed by selection yielded a mutant with 32,000-fold increase in

enzymatic activity. Error-prone mutagenesis of the same genes resulted only in a 16-fold

increase (Stemmer, 1994).

3.5.3.3 Staggered Extension Process (StEP)

DNA shuffling by staggered extension process was developed in 1998 by Zhao et

al., (1998). StEP consists in the priming of template sequences followed by several

cycles of denaturation and shortened annealing/polymerase-catalyzed extension. During

each cycle, the DNA fragments can anneal to different templates based on sequence

complementarity, and extend further to create recombinant genes. Because of the

template switching, the growing polynucleotide chains can contain information from one

or various parental clones (Zhao et al., 1998). The whole process can be performed in a

single PCR reaction, and results in a pool of mutants, the majority of which are

functional.

3.6 Expression and Purification

There is a growing demand for rAbs for treatment of human diseases, in vitro

diagnostic tests, and affinity purification methods, placing pressure on current production

capacity which is based largely on bacterial and tissue culture techniques. Alternative

Page 89: PhD Fusarium

66

systems, such as yeast (Horwitz et al., 1988), baculovirus (zu Putlitz et al., 1990) and

plants (Hiatt and Ma, 1992), are very attractive, both for cost effectiveness and individual

advantages such as scalability and safety for instance, that are absent in bacterial or

mammalian systems. Several plant-produced Abs (plantibodies) are undergoing clinical

trials and the first commercial approval could be only a few years away. The

performance of the first generation of products has been very encouraging so far (Stoger

et al., 2005). Ongoing studies are addressing further biochemical constraints, and aim to

further improve yields, homogeneity and authenticity (e.g. its binding characteristics

compared to parent Abs). There is no universal expression system that can guarantee

high yields of recombinant product, as every Ab will pose unique challenges. The choice

of an expression system will depend on many factors: the type of rAb being expressed,

sequence of the individual rAb and the investigator‟s preferences.

Given that the focus of this book is application of immunoassays within agricultural

biotechnology, studies and experimental findings which describe mammalian expression

and post-translational modification, etc. support rAbs as therapeutic treatments, will not

be addressed.

3.6.1 Prokaryotic Systems

3.6.1.1 E. coli Cytoplasmic Expression

The expression of rAb fragments in the reducing environment of the cytoplasm of

E. coli often leads to the formation of insoluble inclusion bodies, which contain unfolded

protein (Verma et al., 1998). This necessitates the development of refolding protocols to

recover functional Abs. In addition, accumulation of foreign protein in bacterial cells

Page 90: PhD Fusarium

67

may lead to their poor growth. Inducible promoters can, therefore, reduce the risk of cell

toxicity. lac, trp, and their hybrid tac promoters, which are regulated by the lac repressor

and induced by IPTG, are such examples. Another popular promoter is the λPL promoter

responsible for the transcription of the λ DNA molecule, which is regulated by a

temperature sensitive repressor. The T7 RNA promoter can also be used to obtain tightly

controlled, high level expression. Together with the T7 RNA polymerase (encoded by T7

gene1), it is one of the most powerful systems currently employed for recombinant

protein productions. The efficiency of T7 RNA polymerase in processing gene transcripts

is superior to that of native E. coli translation machinery which has made it a very

attractive for high level production of recombinant proteins (see review by Terpe, 2006).

Even under optimized expression conditions with a suitable promoter system,

cytoplasmic expression of functional rAbs and fusion proteins can be very challenging.

For example Peipp et al. (2004) reported that functional rAbs and fusion proteins was

only obtained in a minority of the cases for which it has been attempted.

3.6.1.2 E. coli Periplasmic Expression

Expression of Ab fragments into the periplasmic space of E. coli is the most

promising route to produce functional rAb fragments. This methodology was first

developed for Fv (Skerra and Plückthun, 1988) and Fab (Better et al., 1988; Plückthun

and Skerra, 1989) fragments. The periplasmic route is similar to protein synthesis

pathway in eukaryotes, in which the nascent proteins go through the endoplasmic

reticulum (ER) and into the Golgi apparatus. The transport of secretory proteins to the

periplasm of E. coli is comparable to that of ER and can lead to proper folding. Signal

Page 91: PhD Fusarium

68

sequences such as pelB (pectate lyase), derived from the pelB gene of Erwinia carotovora

are added in frame with the genes encoding the H and L chains of Ab fragments, resulting

in simultaneous secretion of both chains into the periplasmic space (Plückthun and

Skerra, 1989).

The advantage of periplasmic secretion over cytoplasmic expression is that it leads

to the production of an assembled, fully functional product without having to refold the

protein in vitro. The oxidative environment of the periplasm allows for disulfide bond

formation and the low protease environment permits stable folding of the recombinant

proteins (Better et al., 1988). Both intra- and inter-chain protein folding and heterodimer

associations occur in the periplasm, which is necessary for proper folding of a functional

Ab (Plückthun and Skerra, 1989). The bacterial environment, however, is not efficient in

producing full length Ab molecules.

3.6.2 Eukaryotic Systems

Production of Ab fragments in eukaryotes has also been widely studied, from

unicellular yeast cultures (Freyre et al., 2000; Gach et al., 2007) to higher plants (Hiatt et

al., 1989). In this section, different eukaryotic expression systems used for recombinant

antibody production will be described and their individual pros and cons will be

discussed.

3.6.2.1 Yeast Expression

The main advantages of yeast over bacterial systems are related to the fact that yeast

is both a microorganism and a eukaryote. Frenken et al. produced both scFv (1998) and

VHH (2000) in E. coli and Saccharomyces cerevisiae and found that VHH production

Page 92: PhD Fusarium

69

levels in the latter organism was up to 100 mg·L-1

, i.e., 1000-fold higher than E. coli.

Furthermore, E. coli-produced VHH exhibited less specific activity (higher fraction of

incorrectly folded soluble species present in the periplasmic preparation) than yeast. In

another study (Huang and Shusta, 2006), two scFvs were expressed at 0.25 mg·L-1

in E.

coli, while yields in Pichia pastoris were between 60 - 250 mg·L-1

.

Yeast provides folding pathways for heterologous proteins and, when yeast signal

sequences are used, correctly folded proteins can be secreted into culture medium.

Moreover, yeasts rapidly grow on simple growth media, and, therefore, represent an

attractive option for industrial-scale production of rAbs. Proteins which may accumulate

as insoluble inclusion bodies in E. coli are often soluble when expressed in yeast (Freyre

et al., 2000). In addition, the degradation of heterologous proteins, often a problem in E.

coli, is usually reduced in yeast. Another unique feature of yeast is that it has the option

of stable transformation without the use of antibiotic resistance, like ampicillin or

kanamycin resistance commonly used with E. coli. E. coli uses episomal vectors which

propagate extra-chromosomally, whereas in yeast, the vector carrying the gene of interests

is integrated into the host genome by homologous recombination (Joosten et al., 2003).

Secretion of functional Fab and whole immunoglobulin (IgG) Abs in yeast was first

demonstrated by Horwitz et al. (1988) in S. cerevisiae, although expression levels were

rather low (approx. 200 ng Fab·mL-1

). Many efforts have been devoted to improving the

expression level of Ab in yeast cells. Yeast secretion yields have been reported to be as

high as 1.2 g·L-1

for scFv fragment in fed batch fermentation cultures (Freyre et al.,

2000). Elements that control the expression level of foreign protein in yeast have been

Page 93: PhD Fusarium

70

well studied. The yeast invertase signal sequence, the PGK promoter and the

polyadenlylation signal were used in the study of Fab and IgG expression (Horwitz et al.,

1988). The proteins were correctly folded and whole Ab and Fab were purified from the

culture supernatants. The antibodies behaved indistinguishably from their lymphoid cell-

derived counterparts in both direct and competition binding assay. The chimeric whole

Ab from yeast exhibited the same ADCC activity as the chimeric Ab from Sp2/0 cells.

Codon optimization and lower repetitive AT and GC content also improve the expression

levels of some rAbs by 5- to 10- fold in standard shake flask cultures (Sinclair and Choy,

2002; Woo et al., 2002), but this is not the case for all rAbs (Outchkourov et al., 2002).

Non-conventional species of yeasts and filamentous fungi have been tested for

expression of Abs and rAb fragments. Abdel-Salam et al. (2001) reported the expression

of Fab in Hansenula polymorpha. Nevertheless, Ab monomers (kappa and gamma

chains) did not assemble into a heterodimer and were poorly secreted. Yarrowia

lipolytica and Kluyveromyces lactis have also been reported for scFv production

(reviewed by Joosten et al., 2003). Filamentous fungus Trichoderma reesei was also

reported to have secreted 1 mg·mL-1

of Fab into the growth medium under shake-flask

conditions and the yield could be increased to 150 mg·L-1

in bioreactor cultures by fusing

the heavy Fv chain to T. reesei cellubiohydrolase I (CBHI) enzyme (Nyyssönen et al.,

1993; Nyyssönen and Keränen, 1995).

A strong preference has been given to P. pastoris for its aerobic growth; a key

physiological trait that greatly facilitates culturing at high cell densities compared with

the fermentative baker‟s yeast S. cerevisiae (Cregg et al., 1993). An scFv-green

Page 94: PhD Fusarium

71

fluorescent protein was expressed by methylotrophic yeast P. pastoris under the

methanol-inducible alcohol oxidase 1 (AOX1) promoter and secreted into the growth

medium. The soluble fusion protein was properly folded without additional renaturation

or solubilization (Petrausch et al., 2007). Rahbarizadeh et al. (2006) found that high

inoculum densities limit growth potential but gave rise to a higher level of VHH

production in P. pastoris. Medium composition and pre-induction osmotic stress were

found to have the greatest influence on yield, but can be improved when casamino acid or

EDTA are included in growth medium. However, glycerol supplementation during

induction resulted in increased growth rates and biomass accumulation, but expression of

scFv was repressed (Hellwig et al., 2001). Gasser et al. (2006) first engineered the

protein disulfide isomerase (PDI) and the unfolded protein response (UPR) transcription

factor HAC1 to be constitutively over-expressed in P. pastoris (Mattanovich et al., 2004).

While the over-expression of HAC1 led to a moderate increase of Fab secretion, i.e., 1.3-

fold, PDI enabled an increase of the Fab secretion level by 1.9 fold. Hence, the formation

of interchain disulfide bonds can be seen as a major rate limiting factor to Fab assembly

and subsequent secretion.

3.6.2.2 Insect Cell Expression

Insect cell expression systems have been shown to be a viable alternative to

standard microbial and mammalian systems designed to express rAb fragments and full-

size mAbs, particularly for Abs designed for use as therapeutic proteins (Demangel et al.,

2005; Guttieri et al., 2000; Verma et al., 1998). The baculovirus-mediated gene

expression in insect cells not only produces large amounts of the foreign protein while

Page 95: PhD Fusarium

72

allowing it to retain its functional activity, it has a highly restricted host range and is less

likely to have contamination during the downstream process and cause harmful effects in

end users. Between 1 to 500 mg of recombinant protein per liter of infected cells have

been reported (Filpula, 2007; Guttieri et al., 2000). However, the success of a foreign

gene expression in insect cells depends on a number of factors. High quality growth

media and careful culturing is required and, for optimal results, the insect cells should be

highly viable and in the log phase of growth. Unfortunately, expression of the foreign

protein is controlled by a very late viral promoter and peaks while the infection

culminates in death of the host cell, thereby allowing for only transient expression of the

rAb. Previous studies also suggested that, during the late phase of baculovirus infection,

the host‟s secretory pathway can become impaired (Reavy et al., 2000).

An alternative to baculovirus-mediated expression is based on stable transformation

of the gene, under the control of an appropriate promoter, into insect cells. For example,

the Drosophila metallothionein promoter has been used to control expression and found

to be tightly regulated, directing high levels of transcription when induced by heavy

metals, such as cadmium or copper (Guttieri et al., 2000). Moreover, the polyhedrin

promoter is considerably stronger than most other eukaryotic promoters, and therefore

enables the gene to be transcribed at a high level, causing recombinant proteins to be

secreted in the insect cell culture in large amounts (Tan and Lam, 1999). To allow for the

continuous expression of Ab genes, Ab DNA can be inserted into insect cell

chromosomes under the control of the baculovirus immediate to early gene promoter

(IEI), which is recognized by the insect cell RNA polymerase (Guttieri et al., 2000). The

Page 96: PhD Fusarium

73

concentration of IgG recovered from the transformed insect cell culture medium was

considerably lower (approx. 0.06 µg of IgG·mL-1

) than the level generated by infection

with the baculovirus recombinant (approx. 9 µg of IgG·mL-1

) and with the predicted yield

from hybridoma cells (1 – 10 µg of IgG·mL-1

). Nevertheless, the baculovirus system is

still considered a viable alternative to microbial expression systems for whole Ab

molecule expression and when post-translational modification, e.g. glycosylation, is

required.

3.6.2.3 In planta Expression

“Plantibodies”, a contemporary term to describe full size Ab or rAb fragments

expressed in plant tissues, was first reported almost twenty years ago by Hiatt et al.,

(1989). Since then many researchers have been trying to express and scale up the

production of Ab or fragments in plants (reviewed within Fischer et al., 1999; Peeters et

al., 2001; Stoger et al., 2002). Using plants as bioreactors presents many advantages

related with manufacturing facilities, production costs and biosafety, among others.

However, despite the production costs reduction and the biocomparability of plantibodies

with their conventional Ab counterparts, contamination risk exists with mycotoxins,

alkaloids, allergenic and immunogenic proteins, especially if the expression host is a

tobacco plant. However, the favorable properties of plants are likely to make them a

useful alternative for small, medium and large scale production throughout the

development of new Ab-based pharmaceuticals or diagnostic reagents. For example,

Khoudi et al. (1999) reported the production of a functional, purified anti-human IgG

mAb through expression of its encoding genes in perennial transgenic alfalfa.

Page 97: PhD Fusarium

74

The deposition and storage of a scFv rAb fragment in pea seeds was reported using

a seed specific USP promoter (Saalbach et al., 2001), in cereals (Stoger et al., 2000),

tobacco plants (Ko and Koprowski, 2005) or suspension cells (Yano et al., 2004). In

particular, seeds offer special advantages, such as ease of handling and long-term storage

stability. Nevertheless, most of the plantibody studies have focused on expression in

leaves. Arguably, there may not be a high enough demand for diagnostic Ab that requires

the large-scale Ab production possible within plant expression, nonetheless, the idea of

expressing functional rAb molecules in plant has already provided a new way of

understanding plant/virus interaction (Sudarshana et al., 2007). The studying and/or

altering the function of an antigen in vivo, also termed immuno-modulation, has also

gained new insights into plant physiology (Artsaenko et al., 1995; 1998; Conrad and

Fiedler, 1998; Tavlodoraki et al., 1993), plant pathogen resistance (Peschen et al, 2004)

or potentially confers herbicide resistance to plants (Almquist et al., 2004; Weiss et al.,

2006).

Currently, Agrobacterium and particle bombardment are the most commonly used

technologies for plant transformation. A single plant binary vector carrying genes

encoding heavy and light chains under two different promoters has been used to express

both genes and to assemble functional full-size Ab (Düring et al., 1990). The major

drawback of the binary vector is low transformation efficiency due to the large size of the

Ab DNA, in addition to the enormous binary vector required (Komori et al., 2007).

Instead of molecularly stacking the heavy and light Ab genes in the same vector, crossing

two transgenic lines separately expressing the heavy and light chain provides an

Page 98: PhD Fusarium

75

alternative that is less demanding in terms of molecular biology (Schillberg et al., 2003).

The particle bombardment approach can insert genes into the genome of the plant nucleus

or the plastid genome, depending on the vector construct. Chloroplasts have been used

for stable expression of mAb in the chloroplast genome. Advantages of targeting the

chloroplast genome include no position effect, no gene silencing, high

expression/accumulation, and minimized environmental concerns (Daniell et al., 2004).

Chloroplasts can process proteins with disulfide bridges, which is required for proper

folding of proteins. Although stably transformed (transgenic) plants are able to express

correctly folded and functional Ab of both the IgG and IgA classes (Ma et al., 1995) or its

fragments (Owen et al., 1992), yields are generally very low (usually in the range of 1-40

µg·g-1

of fresh biomass). In addition, the time necessary to generate the first grams of

research Ab material is very long, requiring > 2 years (Giritch et al., 2006).

To shorten the time required to obtain the functional antibodies from plants,

transient expression using agro-infiltration or plant viral vector system have been used

(Giritch et al., 2006; Ko and Koprowski, 2005). Using the subgenomic promoter of TMV

coat protein and tobacco mild green mosaic virus variant U5 coat protein, two separate

virus vectors were used to co-infect Nicotiana benthamiana leaf cells. Full length heavy

chain and light chain proteins were produced and assembled into glycosylated functional

full size Ab (Verch et al., 1998). Regeneration time is much shorter than that for stable

transformation, and different host plants can be infected by the same viral vector,

allowing time-efficient screening for recombinant gene expression. More importantly, it

Page 99: PhD Fusarium

76

saves the labor-intensive step of crossing different transgenic plants producing different

Ab subunits.

Many different Ab formats have been expressed successfully in plants. These

include full-size antibodies (Giritch et al., 2006; Ma et al., 1995), camelid heavy-chain

antibodies (Ismaili et al., 2007), Fab fragments (Weiss et al., 2006; Yano et al., 2004),

and scFvs (Owen et al., 1992; Saalbach et al., 2001; Stoger et al., 2000). Regardless of

which form of Ab fragments are being produced by the plant systems, ER is an important

site of the major bio-functions of synthesis, assembly, and glycosylation of these protein

molecules. Antibodies are often targeted to subcellular compartments or the apoplastic

space since most proteins are more stable in the subcellular compartment than in the

cytosol. The greatest accumulation of full size antibodies has been obtained by targeting

to the apoplastic space and the greatest accumulation of scFv antibodies was obtained

when the antibodies were retained in the ER (Daniell et al., 2004).

The average yield of the recombinant protein using plant secretion sequence (SS) is

usually 0.1 - 2% of total soluble protein (Yano et al., 2004). So far, three different leader

sequences have been tested: human derived leader sequence (LS), dicotyledonous

calreticulum derived SS, and monocotyledonous hordothionin derived SS. The latter did

not consistently result in mAb expression, while plants transformed with the dicot SS

construct grew more vigorously and expressed the antibodies more consistently than

transgenic cells made with human LS. The promoter is another critical element

determining the expression level, for examples, Cauliflower Mosaic virus 35S

(CaMV35S) promoter for dicotyledonous plants (McLean et al., 2007; Olea-Popelka et

Page 100: PhD Fusarium

77

al., 2005) and the maize ubiquitin-1 promoter preferable in monocotyledonous plants.

Petruccelli et al. (2006) successfully produced a full length anti-rabies mAb molecule in

transgenic tobacco using CaMV35S to drive the heavy chain, while the potato proteinase

inhibitor II (pin2) promoter controlling light chain gene produced functional mAb. An

effective plant production system for recombinant Ab fragments also requires the

appropriate control of post-translational processing of recombinant products. The ER

retention signal sequence (KDEL) has little effect on the accumulation of Ab in the

transgenic leaves, but leads to higher Ab yields in seeds. The proteins purified from

leaves contain complex N-glycans, including Lewis a epitopes, as typically found in

extracellular glycoproteins and consistent with an efficient ER retention and the cis-Golgi

retrieval of the Ab. The glycosylated proteins purified from the seed were partially

secreted and sorted to protein storage vacuoles (PSVs) in seeds and not found in the ER

(Ramirez et al., 2003). More importantly, full size antibody (McLean et al., 2007) or

scFv fragments (Almquist et al., 2004, 2006; Makvandi-Nejad et al., 2005) produced in

Tobacco have been proved to be functional. This further supports the fact that

“plantibodies” could be an alternate source of diagnostic as well as therapeutic antibodies.

3.7 Application to Immunoassays and Agricultural Biotechnology

3.7.1 Immunoassays

Given the widespread adoption of genetically modified (GM) crops within global

agriculture and strict regulations governing their co-existence with non-GM crops

(Demont and Devos, 2008), mandatory product labeling (e.g., CODEX alimentarius, EU,

2003; etc.) and international trade, there is an increased need to standardize and

Page 101: PhD Fusarium

78

continuously upgrade the analytical methods used to monitor and verify the presence of

GM traits within food and feed (Dong et al., 2008).

Immunoassays for the detection of protein expression within GM crops are currently

limited to enzyme linked immunosorbent assays (ELISA) based on either trait-specific

polyclonal serum or mAbs, as well as rapid lateral-flow / immuno-chromatographic

diagnostic strip kits (typically based on polyclonal serum) (e.g., Lin et al., 2001; Ma et al.,

2005; Tripathi, 2005; Van den Bulcke et al., 2007). Both immunoassay formats are

widely used in the US, Canada, and other countries which produce and export GM crops

as a primary method for determining minimum thresholds of GM products within

commercial food and feed shipments (Stave, 2002).

A wide array of immunoassay products are currently available to assess GM

expression within agricultural crops. These testing formats are developed within private,

or in-house programs or are available as commercial products from companies such as:

Agdia Inc. (Elkart, IN, USA), EnviroLogix Inc. (Portland, ME, USA), and Strategic

Diagnostics Inc. (Newark, DE, USA). Although these platforms are very useful

qualitative tools to detect the presence of GM traits within commercial crops, the results

generated by current immunoassay products are often limited by detection limit and

overall precision, or consistency, of results. For example, Ma et al. (2005) reported that

both ELISA and DNA-based (PCR) tests were capable of distinguishing samples with

GM concentrations between 0.1 to 0.5%, but the precision at this range was very low as

results were highly inconsistent. Another consideration is the disparity between the time,

costs, and resources required to develop GM-specific immunoassay reagents based on

Page 102: PhD Fusarium

79

traditional mAb and polyclonal platforms relative to the rapid progression of GM crops

toward newer events based upon multiple / stacked traits.

Antibody engineering has evolved rapidly over the past 20 years to the point where

it is possible to isolate and optimize high-affinity and Ag-specific rAb fragments as a

cost-effective alternative to established mAb technologies (biomedical applications

reviewed in Holliger and Husdon, 2005). In this regard, rAb fragments (i.e., Fab, scFv,

VHH) could represent a means to complement, and potentially displace, polyclonal serum

and mAbs used within immunoassay formats. In fact, one could argue that the

application of rAb technology to detect GM proteins within plant matrices would be a

natural extension of published research which demonstrated effective use of rAb

technology within immunoassay formats to detect ligands ranging from: mycotoxins

(Wang et al., 2007; 2008) to caffeine (Ladenson, 2006) to large complex protein toxins

such as cholera toxin, ricin and staphylococcal enterotoxin B (Goldman et al., 2006). The

application of rAb technology within current ELISA and lateral flow stick GM

immunoassay formats would obviously depend on any benefits, or advantages, imparted

by this new technology. The following is an initial (and by no means exhaustive) list of

“key traits” that rAb fragments could exhibit relative to current mAb and polyclonal

technology: enhanced GM trait specificity based on recognition of a single target protein;

improved sensitivity to target protein; decreased development costs and timelines; ability

to combine rAb reagents to enable simultaneous and customized detection of stacked GM

traits; improved durability and speed of deployment impart convenient and real-time

immunoassay results.

Page 103: PhD Fusarium

80

Apart from the advantages associated with improved affinity, cost-effective

expression and Ag-specificity (as discussed within this chapter), rAb technology also

imparts unique opportunities to optimize the physical characteristics of rAb fragments to

suit the intended use pattern. For example, it is possible to express single-domain VHH

fragments in a multimeric pentamer format as a means of increasing antibody avidity

(Zhang et al., 2004) with potential use in lateral flow devices and ELISA kits. Depending

on the intended application and immunoassay design, it is also possible to couple rAb

fragments to larger indicator molecules (e.g., fluorophores, etc.) to impart visual GM

detection in real time. Finally, given that VHH fragments have been shown to have high

affinity for large, complex enzymes (e.g., Desmyter et al., 1996; 2002), such fragments

may be particularly suited for detection of GM traits which express enzymes which confer

tolerance to the herbicide glyphosate (e.g., GAT, EPSPS, etc.).

3.7.2 Research Applications to Agricultural Biotechnology

Apart from standard immunoassay detection procedures, it is also possible to apply

rAb technology to study structure-function relationships between GM traits and the target

organism. For example, Fernández et al. (2008) reported the use of scFv phage-display

libraries and various bio-panning techniques to characterize epitopes that mediate binding

of Cry1Ab and Cry11Aa toxins with target protein receptors within Manduca sextra and

Aedes aegypti, respectively. Such results could provide insights into the mechanism of

insect specificity and mode of action of Cry toxins to enable strategies designed to

improve target insect toxicity and specificity. Ongoing research in this area could be

useful to study, and potentially improve upon, the precise mechanism and toxicity by

Page 104: PhD Fusarium

81

which target transgenic events work within the host crop and efficacy versus the target

organism. Finally, rAbs are also well suited to study and modify metabolic pathways

within plants as a means of developing new transgenic traits and GM products (reviewed

in Nölke et al., 2006).

Page 105: PhD Fusarium

82

4 RESEARCH CHAPTER 1: CLONING, EXPRESSION, AND

CHARACTERIZATION OF A SINGLE DOMAIN ANTIBODY

FRAGMENT WITH AFFNIITY FOR 15-ACETYL-DEOXYNIVALENOL.

4.1 Abstract

A single-domain variable heavy chain (VHH) antibody fragment specific to the

mycotoxin 15-acetyldeoxynivalenol (15-AcDON) was obtained after immunization of a

llama (Llama glama) with the protein conjugate 15-DON-BSA plus TiterMax™ Classic

adjuvant. After confirmation of a polyclonal response to DON toxin in both conventional

(cIgG) and heavy chain antibody (HCAb) fractions, a VHH library was constructed from

amplified cDNA by nested PCR. VHH fragments with binding affinity for the mycotoxin

were selected by panning of the phagemid library against microtiter plates coated with 15-

DON-OVA. The dominant clone (NAT-267) was expressed in E. coli and was purified

as a VHH monomer (mNAT-267) at a final concentration of 1.3 mg∙mL-1

. Isolated NAT-

267 VHH DNA was fused to the homopentamerization domain of the B subunit of

verotoxin to generate the pentabody format of single-domain antibody (sAb). The VHH

pentamer (pNAT-267) was expressed in E. coli and was purified at a final concentration

of 1.0 mg∙mL-1

. Surface Plasmon Resonance (SPR) analysis of soluble mNAT-267

binding kinetics to immobilized 15-DON-HRP (Horse Radish Peroxidase) indicated a

dissociation constant (KD) of 5 μM. Competitive Direct Enzyme-Linked Immunosorbent

Assay (CD-ELISA) and Fluorescence Polarization Assay (FPA) inhibition experiments

with monomer and pentamer confirmed binding to 15-AcDON. Competitive inhibition

FPA tests with mNAT-267 and pNAT-267 IC50 values for 15-AcDON hapten were 1.24

μM and 0.50 μM, respectively. These values were similar to the IC50 value of 1.42 μM

for 15-AcDON given by polyclonal llama serum sampled 56 days after immunization.

Page 106: PhD Fusarium

83

Competition formats for structurally related trichothecenes resulted in no cross reactivity

to: DON; 3-acetyldeoxynivalenol (3-ADON); neosolaniol (NEO); diacetoxyscirpenol

(DAS); and T-2 toxin. Our study confirmed that recombinant VHH fragments capable of

binding low molecular weight haptens can be produced through the creation and panning

of hyper-immunized single domain (sdAb) libraries.

Key Words

VHH, Hapten, Mycotoxin, 15-acetyl-deoxynivalenol, Phagemid Library, Surface

Plasmon Resonance, CD-ELISA, Fluorescence Polarization Assay.

4.2 Introduction

Deoxynivalenol (3,7,15-trihydroxy, 12,13-epoxy-trichothec-9-en-8-one), or DON, is

one of many members of the trichothecene group of mycotoxins produced by Fusarium

species. DON is a low molecular weight hapten (M.W. 296 Da) containing one primary

and two secondary hydroxyl groups conferring limited solubility in water and high

solubility in various polar solvents such as methanol and acetonitrile (Desjardins, 2006).

Of the trichothecene mycotoxins that accumulate within cereal grain, DON toxin and its

acetylated metabolites (i.e., 15-acetyl-deoxynivalenol (15-AcDON) and 3-acetyl-

deoxynivalenol (3-AcDON)) are considered among the most commonly and widely

distributed food and feed contaminants (Abramson et al., 2001; Greenhalgh et al., 1986).

Immunoassay techniques using polyclonal and monoclonal antibodies (mAb) have

been developed to detect DON and other mycotoxins within grain, food, feed, and animal

serum (e.g., Maragos and McCormick, 2000; Sinha et al., 1995). These systems typically

use anti-DON mAbs from hybridoma cell lines producing murine immunoglobulins.

Page 107: PhD Fusarium

84

Although these tests provide robust and precise results, mAb development requires highly

specialized equipment and labor-intensive procedures to select, culture, and maintain

optimal hybridoma cell lines (Sinha et al., 1995).

Heavy chain antibodies found within sera of the camelidae (i.e., camels,

dromedaries and llamas) (Hamers-Casterman et al., 1993) are unique in that they are

devoid of the variable light chain (VL) and first constant region (CH1) which are common

to larger conventional immunoglobulin (cIgG) structures (Nguyen et al., 1999). The

paratope of HCAbs is comprised of a single heavy chain variable domain (often called

„VHH‟) which is directly linked to the second constant domain (CH2) by a unique hinge

region (Hamers-Casterman et al., 1993).

Extended hinge regions coupled with deletion of the CH1 domain, and amino acid

substitutions within the variable domain are believed to impart increased hydrophilicity

and solubility of HCAbs within serum (Desmyter et al., 2001; Nguyen et al., 2000; Vu et

al., 1997). These properties coupled with expanded complementarity determining region

(CDR) loops within the VHH paratope may expand the antigen (Ag) - binding repertoire

within a camelid immune system by enabling HCAbs to bind otherwise inaccessible

epitopes (Muyldermans, 2001; Nguyen et al., 2000).

Despite their lack of variable light and heavy chain pairing, single domain

antibodies (sdAb) recognize and bind a wide variety of epitopes and Ag types (Nguyen et

al., 2000; Sheedy et al., 2006; van der Linden et al., 2000). Recombinant sdAb fragments

derived from phage libraries are regarded as among the smallest intact Ag-binding

fragments currently known (Muyldermans, 2001) and demonstrate robust and stable

Page 108: PhD Fusarium

85

expression in bacteria (e.g., Desmyter et al., 2001; Ghahroudi et al., 1997; Sheedy et al.,

2006; etc.) and yeast (e.g., Dolk et al., 2005; Frenken et al., 2000; etc.) relative to other

rAb formats (e.g., Fab, Fv, scFv, mAb, etc.).

The advantages associated with ease of expression, stability, and solubility of VHH

fragments can be attributed to key differences relative to standard rAb fragments (e.g.,

scFv, Fv, Fab, etc.). VHH fragments are small (≈ 14 – 15 kDa), typically have hydro-

philic amino acid substitutions within the second framework region (FR-2) and by their

single-domain nature, do not require the linker associated with the standard scFv format

which can lead to problems associated with aggregation, proteolytic degradation and

variable expression within recombinant hosts (Arbabi-Ghahroudi et al., 2005). Improved

functionality over various temperature and solvent regimes (Goldman et al., 2006; van

der Linden et al., 1999) coupled with high levels of recombinant expression combine to

make VHH fragments useful reagents within next-generation immunoassays (Goldman et

al., 2006, Ladenson et al., 2006) and for expression within recombinant hosts to evaluate

potential in vivo mechanisms of immunomodulation (Muyldermans, 2001).

As with other rAb fragments, VHH fragments isolated from hyper-immunized or

naïve libraries are highly specific based on the recognition of unique epitopes on target

Ags (Ghahroudi et al., 1997; Goldman et al., 2006; Ladenson et al., 2006; Sheedy et al.,

2006; van der Linden et al., 2000). A wide-range of affinities has been published for Ags

ranging from large immunogenic proteins to enzymes to low-molecular weight haptens

(reviewed in Muyldermans, 2001). A growing number of VHH fragments have been

isolated which demonstrate a range of binding affinities against an array of different

Page 109: PhD Fusarium

86

hapten types (Alvarez-Rueda et al., 2007; Goldman et al., 2006; Ladenson et al., 2006;

Sheedy et al., 2006; Spinelli et al., 2000; 2001; Yau et al., 2003b). These results serve to

validate potential uses of this new technology in terms of small molecule binding.

The objective of this research was to isolate and characterize mycotoxin-specific

recombinant VHH fragments from a hyper-immunized phage-display library. The

identification of mycotoxin-specific fragments will provide additional information to

further characterize the nature and binding kinetics of VHH antibody fragments to

haptens. Such fragments will also aid in the development of low cost, durable reagents

required for effective and robust toxin detection systems (Goldman et al., 2006; Ladenson

et al., 2006). Finally, with favorable binding affinity and expression, mycotoxin-specific

VHH fragments can also be used in eukaryotic systems in vivo (e.g., yeast and plant cells)

to evaluate their potential for immunomodulation of the cytotoxic effects of DON and

related trichothecene mycotoxins.

4.3 Materials and Methods

4.3.1 Reagents and Solutions

Restriction enzymes were purchased from New England Biolabs (Mississauga, ON).

Ampicillin, kanamycin, bacto-tryptone, bacto-yeast extract, isopropyl β-D-1-

thiogalactopyranoside (IPTG), phenylmethylsulphonyl fluoride (PMSF), isopropanol, 6-

aminofluorescein (fluoresceinamine isomer II), bovine serum albumin (BSA), ovalbumin

(OVA), TiterMax™ Classic, N-(2-hydroxyethyl)-piperazine-N-2-ethanesulfonic acid

(HEPES), Tris HCl, sodium dodecyl sulfate (SDS), triethylamine, polyethylene glycol

(PEG), and polyoxyethylene sorbitan mono-laurate (Tween 20) were purchased from

Page 110: PhD Fusarium

87

Sigma Chemical Co. (St. Louis, MO). Skim milk was purchased from Difco (Oakville,

ON). PCR primers were synthesized by Gibco BRL (Burlington, ON). Mouse anti-6-His

monoclonal IgG and goat anti-mouse alkaline phosphatase conjugate were purchased

from Qiagen (Mississauga, ON). PVDF membrane and NBT (nitro blue tetrazolium

chloride) / BCIP (5-Bromo-4-chloro-3-indolyl phosphate, toluidine salt) were purchased

from Roche Diagnostics (Laval, PQ). 96 well Maxisorp™ microtiter-plates were

purchased from Nalge Nunc Inc. (Naperville, IL). Rabbit anti-6-HIS-HRP conjugate was

purchased from QED Bioscience Inc (San Diego, CA). AP and HRP development

substrates were purchased from Kirkegaard & Perry Laboratories (Gaithersburg, MD).

4.3.2 Immunogen and Llama Immunization

All DON-based reagents were obtained from the Eastern Oilseed and Cereal

Research Center (ECORC) of Agriculture and AgriFood Canada (AAFC) Ottawa,

Ontario, Canada. DON was isolated from large-scale fermentations of Fusarium

graminearum liquid culture followed by purification as described by (Greenhalgh et al.,

1986). 15-AcDON was prepared by selective hydrolysis of 3,15-diacetyl-DON, which

was obtained by acetylation of DON or 3-AcDON (Savard, 1991). Neosolaniol and T-2

Toxin were isolated according to standard methods (Greenhalgh et al., 1986).

Diacetoxyscirpenol (DAS) toxin was purchased from Sigma-Aldrich (Oakville, Ontario).

Synthesis of DON-protein conjugates 15-DON-BSA, 15-DON-OVA, 3-DON-HRP and

15-DON-HRP was based on standard methods (Bauminger and Wilcheck, 1980) with

modifications as previously described (Sinha et al., 1995). DON-protein conjugates were

Page 111: PhD Fusarium

88

purified by size exclusion chromatography on Sephadex G-15 (2.5 x 21-cm column) with

distilled deionized water at a flow rate of 4 mL∙min-1

.

A two-year-old male llama was immunized by sub-cutaneous (Sub-Q) lower-back

injection of 15-DON-BSA (250 μg∙mL-1

) conjugate in 1.0 mL PBS + 1.0 mL of TiterMax

Classic adjuvant. The llama was boosted with a second injection three weeks after the

first immunization followed by three subsequent injections every two weeks for a total of

five immunizations over a nine-week period.

Pre-immune serum was collected as a negative control, and 15.0 mL of blood was

collected one week after each immunization to measure titer at 7, 28, 42, 56 and 70 days

after the first immunization. Serum was prepared by storing blood samples overnight at

4ºC followed by centrifugation at 2,700 x g for 10 minutes at 4ºC. Samples of fresh

blood (20 mL) were collected one week after the third and fifth immunizations from

which leukocytes where isolated, i.e., 42 and 70 days after immunization, respectively.

The isolation of RNA from peripheral leukocytes was performed using a QIAamp RNA

Blood Mini Kit (Cat.No. 52304 – Qiagen Inc., Mississauga, Canada).

4.3.3 Fractionation of Polyclonal Serum

Llama serum from a pre-immune sample and from samples taken one week after the

third and fifth immunizations (42 and 70 day samples, respectively) were fractionated by

protein G and protein A chromatography (Hi Trap, Pharmacia, Upsala, Sweden) and

separated by acidic elution as previously described (Hamers-Casterman et al., 1993).

Serum fractions A1 (HCAb), A2 (HCAb), G1 (HCAb), and G2 (cIgG) from each

collection date were dialyzed against PBS and subsequently stored at 4°C with 0.01%

Page 112: PhD Fusarium

89

NaN3 as a preservative. The IgG content of the isolated serum fractions was measured

using the BCA-protein assay (Pierce, Rockford, IL, USA).

4.3.4 Confirmation of Titer

Polyclonal serum was screened against 15-DON-OVA or OVA (control) by indirect

ELISA as based on a modified standard protocol (Sinha et al., 1995). Briefly, a standard

96-well microtiter plate was coated with 100 μL 15-DON-OVA (5 μg∙mL-1

in 0.15 M

carbonate-bicarbonate buffer, pH 9.6). Blank wells were coated with 100 µL OVA (20

μg∙mL-1

in PBS). After incubation overnight, plates were washed with PBST, and

blocked with 1.0% OVA (2 hours, 37°C) followed by washing with PBST. Non-

immunized (control) wells for 15-DON-OVA and OVA (blank) were coated with pre-

immune polyclonal serum. Treatment wells were coated with 42- and 70-day serum

samples. Immune and non-immune polyclonal sera were diluted serially from 1:3 to

1:59,049 in PBS for both 15-DON-OVA and OVA (blank) coated wells. After

incubation at room temperature (1.5 hours) and washing with PBST, 100 μL of goat anti-

llama IgG (1:1,000 dilution PBS) was added. Plates were incubated at 37°C for 1 hr and

washed five times with PBST. Swine anti-goat HRP conjugate (100 µL; 1:3,000 dilution

in PBS) was added followed by incubation and washing as described. Titer was

determined with standard colourimetric buffer followed by 1 M H3PO4 and measurement

of optical density at 450 nm on a standard ELISA plate reader (Dynatech MR5000). Best

titers were defined as the greatest sample dilution giving twice the background

absorbance of OVA control.

Page 113: PhD Fusarium

90

Titer of cIgG and HCAb fractions were tested against 15-DON-OVA or OVA

(control) by indirect ELISA as described above with one modification. As an additional

blocking step, equal volumes and concentrations of processed cIgG and HCAb fractions

were „pre-incubated‟ with 2% OVA in PBS. Antibody fractions were added to 15-DON-

OVA and control wells and diluted in PBS to establish a dilution series from 1:2 to

1:1,024 and processed according to the previously described method (Sinha et al., 1995).

4.3.5 Library Construction

Total RNA was isolated from the leukocyte fraction of Peripheral Blood

Lymphoctes (PBL) of whole blood samples collected one week after the third and fifth

immunizations (42 and 70 days, resp.) using a QIAamp RNA Blood Mini Kit (Cat. No.

52304 – QIAGEN Inc., Mississauga, Canada). EDTA (dipotassium salt) at 1-2 mg ml-1

blood was used as an anticoagulant. The VHH library was constructed according to

Ghahroudi et al., (1997) with minor modifications. Briefly, first strand complementary

DNA (cDNA) was derived from 33 µL of total RNA (approx. 3 µg∙20 µL-1

) isolated from

42 and 70 day PBLs using oligo dT primer and a „First-Strand cDNA Synthesis Kit‟

(Amersham Biosciences, Buckinghamshire, UK). Resultant synthesis of VH and VHH

dsDNA occurred through nested PCR using proofreading Taq polymerase with sense and

anti-sense hinge-specific primer mixtures “MJ1.2.3 Back”, and “CH2 + CH2b3”,

respectively (Table 4.1). After amplification, doublet PCR bands between 620 – 670 bp

corresponding to VHH – CH2 hinge HCAb dsDNA were agarose gel-purified. Gene-

specific primers: MJ7 (Sense) and MJ8 (Antisense) (Table 4.1) were used to amplify the

Page 114: PhD Fusarium

91

VHH region of HCAb and add two SfiI restriction sites flanking the 5‟ and 3‟ termini of

VHH coding sequences. PCR products were then pooled and agarose gel-purified.

Table 4.1 Nucleotide sequences of primers for PCR amplification of HCAb VHH

genes.

Type Name Nucleotide Sequencea

PCR Isolation and Amplification

Forward

MJ1.2.3

MJ1 Back 5’-GCCCAGCCGGCCATGGCCSMKGTGCAGCTGGTGGAKTCTGGGGGA-3’

MJ2 Back 5’-GCCCAGCCGGCCATGGCCCAGGTAAAGCTGGAGGAGTCTGGGGGA-3’

MJ3 Back 5’-GCCCAGCCGGCCATGGCCCAGGCTCAGGTACAGCTGGTGGAGTCT-3’

Reverse CH2 5’-CGCCATCAAGGTACCAGTTGA-3’

CH2b3 5’-GGGGTACCTGTCATCCACGGACCAGCTGA-3’

Addition of SfiI

Forward MJ7 5’-CATGTGTAGACTCGCGGCCCAGCCGGCCATGGCC-3’

Reverse MJ8 5’-CATGTGTAGATTCCTGGCCGGCCTGGCCTGAGGAGACGGTGACCTGG-3’

Colony PCR

Forward PN2 5’-CCCTCATAGTTAAGCGTAACGATCT-3’

Reverse MJ13RP 5’-CAGGAAACAGCTATGAC-3’

a Key to symbols: M=A+C, K=G+T, S=G+C.

VHH dsDNA was ligated into pMED1 phagemid vector (Ghahroudi et al., 1997)

using the LigaFast Rapid DNA ligation system (Promega) according to standard methods.

Briefly, amplified VHH fragments were digested with SfiI restriction enzyme along with

pMED1 phagemid DNA overnight at 50°C. To reduce the probability of self-ligation of

the phagemid vector and to improve the functional size of the VHH library, pMED1 DNA

was further digested with PstI and XhoI restriction enzymes (2 hours, 37°C). Once the

ratio of insert to vector was optimized, VHH leukocyte DNA was ligated into the pMED1

phagemid vector at the SfiI restriction site prior to ligation to create the library.

Page 115: PhD Fusarium

92

After transformation into TG1 E. coli (Stratagene), VHH library size and diversity

were estimated by plating on LB + ampicillin (100 mg∙mL-1

) agar plates followed by

colony PCR using universal reverse primer “MJ13RP” and forward primer “PN2” (Table

4.1). 30 positive clones which produced a PCR product of approximately 600 bp (400 bp

VHH plus 200 bp oligo DNA) were nucleotide sequenced. Positive clones were also

digested with the random 4-mer restriction endonuclease enzyme BstN1 followed by

agarose gel electrophoresis. Clones were regarded as unique if the pattern of digestion

produced a DNA „fingerprint‟ that was different from other randomly selected clones.

4.3.6 VHH Library Panning

The VHH library was subjected to four rounds of panning on 96-well Maxisorp

plates. Target Ag was coated as: 20.0 µg, 17.5 µg, 15 µg, and 10.0 µg 15-DON-OVA per

well for rounds 1, 2, 3, and 4, respectively. Two replicates of target Ag were used in each

round of panning. Blank wells (one replicate) were maintained during panning by coating

wells with PBS. Antigen and control wells for all panning rounds were coated with 100

μL∙well-1

and incubated overnight at 4°C. Blocking buffer (2% w/v OVA in PBS) was

used for panning rounds 1 and 2. To limit non-specific binding for rounds 3 and 4,

blocking buffer was switched to casein (1% w/v in PBS). As well, phage from each

round of panning were also pre-incubated with the respective blocking solution (100 µL)

for 1 hour at 37°C before 100 μL of phage were added to 15-DON-OVA and PBS coated

wells. After incubation of phage for 2 hours at 37°C, antigen and control wells were

washed with PBST. Stringency of washing was increased by washing 5, 6, 7, and 8 times

with 200 µL PBST for rounds 1, 2, 3, and 4 of panning, respectively.

Page 116: PhD Fusarium

93

Bound phage were eluted by adding 100 µL of freshly prepared triethylamine

solution (35 µL TEA + 2.50 mL sterile H2O). Eluant was neutralized after incubation for

10 minutes by adding 200 µL of 1.0 M Tris HCl. Eluted phages after each round of

panning were used to infect 2 mL E. coli TG1 cells for subsequent amplification as

described in (Ghahroudi et al., 1997). Briefly, infected cells were incubated at 37°C for

30 min without shaking. A 2 μL aliquot was taken for phage titration, the remaining

culture volume was transferred to 6 mL 2xYT medium + ampicillin (100 μg∙mL-1

)

followed by incubation and shaking (37°C, 30-60 min). The culture was subsequently

superinfected with 900 μL of helper phage M13 K07 (1 x 10 11

cfu∙mL-1

) (New England

Biolabs) followed by incubation with no shaking (37°C, 15 min.) and incubation with

shaking (37°C, 30-60 min). Cells were transferred to 92 ml 2xYT medium containing

ampicillin and kanamycin (final concentrations of 100 mg∙mL-1

and 50 μg∙mL-1

,

respectively). After amplification of eluted phages overnight (37°C with shaking), phage

titers were estimated by plating infected cells on 2xYT + ampicillin 100 mg∙mL-1

agar

plates. Input and eluted phage were quantified by serial dilution with PBS and plating on

LB-AMP (data not shown). The number of colonies in each plate were counted and

compared with the colony numbers from subsequent rounds of panning.

4.3.7 Phage ELISA

Individual colonies obtained after four rounds of panning were tested against 15-

DON-OVA conjugate in a phage ELISA according standard procedures (Ghahroudi et al.,

1997). Briefly, clones were grown in 2xYT medium + ampicillin (100 mg∙mL-1

) + 0.1 %

glucose medium to OD600 = 0.3 – 0.5, and infected with M13K07 helper phage (37°C no

Page 117: PhD Fusarium

94

shaking, 30 min) followed by kanamycin (50 μg∙mL-1

) and amplification overnight (37°C

with shaking). Cultures were centrifuged (5,000 rpm, 20 min., 4°C) to pellet the cells,

followed by the addition of 100 μL of supernatant (recombinant phage particles) to pre-

coated microtiter plate wells. Treatments were two replicates of 15-DON-OVA (0.5

μg∙well-1

in PBS or 0.15 M carbonate-bicarbonate buffer). Blank wells were OVA (2.0

μg∙well-1

PBS) and BSA (2.0 μg∙well-1

PBS). Negative control was 100 μg∙well-1

of

M13K07 in PBS. After 2-hour incubation at 37°C, microtiter plate wells were washed

five times with PBST followed by addition of anti-M13 HRP conjugate (1:5000). VHH-

phages bound to DON were detected by the addition of 100 μL of HRP substrate (KDL)

followed by colourimetric development and assessment as previously described.

4.3.8 Colony PCR and Sequencing

Universal primers M13R and PN2 were used to amplify the VHH fragments of the

positive clones determined by phage ELISA. Sequencing was carried out using the ABI

PRISM™ Big Dye® Terminator Cycle Sequencing Reaction Kit (Perkin-Elmer,

Wellesley, MA) on a 373 DNA sequencer apparatus (Perkin-Elmer). Sequencing data

were aligned and numbered according to the Kabat numbering system (Kabat et al.,

1991). The unique nucleotide sequence for the dominant clone (repeated in 38 of 100

clones) that bound to 15-DON-OVA and not to OVA control was termed “NAT-267”.

4.3.9 Monomer Expression and Purification

Restriction enzyme sites BbsI and BamHI were added to the 5‟ and 3‟ ends of the

NAT-267 VHH DNA fragment using a PCR involving gene-specific sense primer VHH-

BbsI (5‟-TATGAAGACACCAGGCCCAGGTGCAGCTGGTGGAGTCT-3‟) and anti-

Page 118: PhD Fusarium

95

sense primer VHH-BamHI (5‟-CGCGGGATCCTGAGGAGACGGTGACCTGGGT-3‟).

The amplified DNA was then digested with BbsI and BamHI restriction enzymes and

ligated into digested pSJF2 vector using standard techniques (Tanha et al., 2003).

Competent E. coli TG1 cells were transformed and clones expressing DON-specific

recombinant VHH were grown in 1-liter cultures of 2xYT medium + ampicillin (100

mg∙mL-1

) with 0.1% glucose to an OD600 of 0.8. Cultures were induced with 0.5 mM

IPTG and grown overnight on a rotary shaker at 28°C. After confirmation of expression

by SDS-PAGE and Western blotting, recombinant mNAT-267 VHH protein was extracted

from the bacterial periplasm by standard methods (Anand et al., 1991) and purified by

immobilized metal affinity chromatography (IMAC) and quantified as described

elsewhere (MacKenzie et al., 1994). The state of aggregation of the purified protein was

checked by size exclusion chromatography on Superdex 200 (Amersham Biosciences).

4.3.10 Pentamerization of VHH Monomer

Restriction enzyme sites BbsI and ApaI were added to the 5‟ and 3‟ ends of the

NAT-267 VHH DNA fragment using a PCR involving the gene-specific sense primer

VHH-BbsI and antisense primer VHH-ApaI (5‟-ATTATTATGGGCCCTGAGGAGACG

GTGACCTGGGTC-3‟). Amplified NAT-267 DNA was digested with BbsI and ApaI

restriction enzymes and ligated into digested pVT2 using techniques as described in

(Zhang et al., 2004). A VHH pentamer of the dominant clone was formed by fusion of

monomer DNA to the verotoxin B subunit encoded within the pVT2 expression vector.

pNAT-267 DNA was then used to transform E. coli TG1 according to standard

techniques.

Page 119: PhD Fusarium

96

4.3.11 Pentamer Expression and Purification

The same conditions which were used to express the mNAT-267 VHH protein, were

applied for expression of pNAT-267 VHH. In accordance with standard protocols (Zhang

et al., 2004), expressed proteins were extracted by whole-cell lysis. Briefly, cells were

pelleted by centrifugation (20 min, 6,000 xg, 4°C) and resuspended in 25 mL of ice-cold

lysis buffer (10 mM Hepes, 500 mM NaCl; 20 mM Imidazole; pH 7.4) followed by

centrifugation (1 hour, 6,000xg, 4°C). Supernatants were filtered (0.22 µm Millipore

filter) and protein extracts were centrifuged again (1 hour, 6,000 xg, 4°C). After

confirmation of protein expression by SDS-PAGE and Western Blotting, pentabodies

were purified by IMAC as described above. The state of aggregation was checked by size

exclusion chromatography on Superdex 200 (Amersham Biosciences).

4.3.12 Determination of Binding Affinity

4.3.12.1 Fluorescence Polarization Assay

DON-fluorescein tracer was prepared according to standard methods (Maragos et

al., 2002). Briefly, DON (0.625 mg in 100 μL acetone) was added to 1,1-carbonyl-

diimidazole (2 mg) and allowed to mix (2 hours, 24C). 6-aminofluorescein (10 mg) was

dissolved in 1 mL sodium carbonate (1 mL, 0.1 M). 6-aminofluorescein (100 µL) was

added to DON-1,1-carbonyl-diimidazole, final volume 200 μL. After reaction for 48

hours at 2 - 8C, DON– fluorescein solution (20 μL; approx. 20 mM) was spotted onto

LK6F silica gel plates (1,000 m Preparative layer thickness 20 x 20 cm; Whatman)

along with samples of DON (20 mM), DON-1,1-carbonyl-diimidazole (20 mM), and 6-

aminofluorescein (20 mM), as reference standards. Thin layer chromatography (TLC)

Page 120: PhD Fusarium

97

plates were developed in chloroform:methanol:acetic acid (90:10:1 v/v/v). Each product

visualized at 366 nm was scraped from the plate into a vial containing 0.2 mL methanol

(2 - 8C) incubated over-night and stored at -20C until required.

Fluorescence polarization was measured with a Perkin Elmer Envision 2100

multilabel reader. To determine the operating dilution of fluorescent tracer within the

assay, DON-fluorescein was serially diluted 1:2 in 1 mg∙mL-1

BSA from 1:100 to

1:102,400 and analyzed by fluorescence polarization (100 μL∙well-1

) with an excitation

wavelength of 488 nm and an emission wavelength of 530 nm. The operating dilution of

DON-fluorescein conjugate was the dilution yielding a total fluorescence within the linear

range of the above titration, nearest the lower plateau to represent the lowest accurately-

measurable concentration of DON-fluorescein conjugate, thus requiring the least amount

of VHH to saturate the system. To determine the dilution of mNAT-267 and pNAT-267

VHH fragments, DON- fluorescein conjugate was diluted in 1 mg∙mL-1

BSA and mixed

with serial dilutions of VHH in PBS. These mixtures were allowed to react (10 min.,

22C) in the dark and followed by fluorescence polarization (100 μL∙well-1

) analysis with

excitation and emission wavelengths of 488 nm and 530 nm, respectively.

A standard curve of competition between DON-fluorescein (1:8,000 dilution in

PBS) and free DON in solution at concentrations ranging from 5 μg∙mL-1

to 9.8 ng∙mL-1

was prepared using a competition assay. VHH antibody fragments were diluted 1:50 (v/v)

in PBS containing DON serially diluted 1:2 (v/v) from an initial concentration of 10

μL∙mL-1

(six replicates for each concentration). Mixtures were allowed to react for 1

hour at 22C after which they were applied (80 μL∙well-1

) to a black 96-well microtitre

Page 121: PhD Fusarium

98

plate. DON-fluorescein was diluted 1:640 (v/v) in 1 mg∙mL-1

BSA and added (20

μL∙well-1

) to the serum-DON solution in each well. Mixtures reacted for 10 minutes at

22C in the dark and were analyzed by FPA. Cross reactivity with other trichothecene

mycotoxins (i.e., 3-AcDON, 15-AcDON, DAS, T2, and NEO) was determined by

substitution of DON with each mycotoxin within the serial dilutions as previously noted.

4.3.12.2 Surface Plasmon Resonance

The interaction of mNAT-267 VHH and immobilized 15-DON-HRP conjugate was

determined by SPR using the BIACORE

3000 biosensor system (Biacore, Inc.,

Piscataway, NJ). Data were analyzed with BIAevaluation 4.1 software. Approximately

500 resonance units (RUs) of 15-DON-HRP conjugate and 190 RUs HRP (control) were

immobilized on a CM5 research grade sensorchip (BIACORE), respectively.

Immobilizations were carried out at protein concentrations of 50 μg∙mL-1

for conjugate

and 200 μg∙mL-1

for HRP in 10 mM acetate (pH 4.0) using an amine coupling kit

supplied by the manufacturer. Before analysis, the mNAT-267 VHH was passed through

a Superdex 75 (GE Healthcare) column. mNAT-267 VHH was injected over HRP

reference and 15-DON-HRP surfaces at concentrations of: 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10

μM for 6 sec. All analyses were carried out at 25ºC in 10 mM HEPES, pH 7.4,

containing 150 mM NaCl, 3 mM EDTA and 0.005% surfactant P20 at a flow rate of 100

μL∙min-1

with 6-sec pulse injection of mNAT-267 VHH followed by 6-sec. injection of

10 mM sodium borate, pH 8.5, in 1 M NaCl (BIACORE) for regeneration. Steady state

binding (Req) at each concentration of VHH monomer was determined for KD analysis.

Page 122: PhD Fusarium

99

To evaluate competitive inhibition of mNAT-267 to free DON, 15-DON-HRP

conjugate and 190 RUs of HRP (control) were immobilized on a CM5 research grade

sensorchip (BIACORE) as noted above. All analyses were conducted at 25°C in 10 mM

HEPES, pH 7.4, containing 150 mM NaCl, 3 mM EDTA and 0.005% surfactant P20 at a

100 μL∙min-1

flow rate with 6 sec pulse injection of VHH monomer in absence or presence

of free DON (50 and 500 μM).

pNAT-267 VHH and control pentabody „C3C-BSA8‟ were captured on a Ni2+

charged sensorchip NTA (BIACORE) at densities of 10,000 RUs and 6,000 RUs,

respectively. DON toxin was injected over both surfaces at a flow rate of 20 μg∙min-1

at

concentrations ranging from 0.1 - 2 mM. Pentamers were regenerated with 1-min

injection of 350 mM EDTA in 10 mM HEPES pH 8.5 containing 150 mM NaCl and

0.005% P20 surfactant. All analyses were conducted at 25ºC in 10 mM HEPES, pH 7.4

containing 150 mM NaCl and 0.005% surfactant P20.

4.3.12.3 Competitive Inhibition ELISA

Microtiter plates were coated with either 15- DON-OVA conjugate (0.5 μg∙well-1

,

0.15 M bicarbonate buffer) or OVA (2.0 μg∙well-1

, PBS) and blocked with 1.0% casein

(PBS) (2 hours, 37°C). Eleven serial dilutions of DON ranging from 0.005 to 10.00

μg∙mL-1

(17 nM to 34 µM, respectively) were prepared in PBS. Purified soluble mNAT-

267 or pNAT-267 VHH (100µL) were added in equal volumes to each dilution series for a

final DON concentration ranging from 0.0025 to 5 μg∙mL-1

(or 8.4 nM to 17 µM,

respectively). After vortexing and incubation (2 hours, 37°C), DON–VHH solutions were

added to the ELISA-plate wells and incubated (1 hour, 37°C) followed by five washes

Page 123: PhD Fusarium

100

with PBST. Rabbit anti-6 His-HRP conjugate (100 µL; 1:5,000 in PBS) was added to

each well followed by incubation (1 hour, 25°C). HRP substrate (KPL, Gaithersburg,

MD) was added followed by the addition of 1 M H3PO4. Absorbance at (450) nm was

measured with an ELISA plate reader. Cross reactivity with other trichothecene

mycotoxins (i.e., 3-AcDON, 15-AcDON, DAS, T2, and NEO) was tested by substitution

of DON-standards with each mycotoxin using the serial dilutions previously noted.

4.4 Results

4.4.1 Polyclonal Response

4.4.1.1 Direct ELISA

ELISAs using polyclonal llama sera sampled either 42 or 70 days after initial

immunization with 15-DON-BSA + TiterMax Adjuvant confirmed a robust humoral

response over a dilution series ranging from 1:1 to 1:2,097,152 (Figure 4.1). Polyclonal

titer, expressed as 50% of maximum absorbance for pre-immune, 42, and 70 day sera

were estimated to be 1:250; 1:70,800; and 1: 56,200, respectively. Overall response was

regarded as mycotoxin-specific as the response of polyclonal sera to control antigen (2%

OVA) was markedly less than to wells coated with 15-DON-OVA (Figure 4.1).

Page 124: PhD Fusarium

101

0.0

0.5

1.0

1.5

2.0

0.00000010.0000010.000010.00010.0010.010.11

Log of Dilution

Absorbance

(450 nm)

Pre: 15-DON-OVA

42d IgG: 15-DON-OVA

72d IgG: OVA Control

72d IgG: 15-DON-OVA

Figure 4.1 ELISA of polyclonal llama serum response to 250 μg 15-DON-BSA

conjugate. 42-and 70-day serum samples each taken one week after third

and fifth immunizations, respectively. X-axis shows dilutions from 1 to

1:2,097,152 in PBS. Antigen wells were coated with 15-DON-OVA (0.5

μg•well-1

). Control wells were coated with OVA (2.0 μg·well-1

).

4.4.1.2 Fluorescence Polarization

TLC of DON-fluorescein conjugate yielded three distinct bands (Rf = 0.29, 0.24 and

0.18). Llama anti-DON serum had affinity for each conjugate using FPA (data not

shown). DON-fluorescein conjugate corresponding to Rf = 0.29 was used for the

development of an optimized FP assay (FPA) since it was the most plentiful product and

had the most consistent results.

A competitive polyclonal FPA was established with 56-day polyclonal serum based

on a plot of polarization/maximum polarization observed (i.e. P/Po) against DON

concentration. The linear working range of the FPA was 10 – 5,000 ng∙mL-1

with limits

of detection and quantitation of 19 ng∙mL-1

and 50 ng∙mL-1

, respectively (data not

shown). The IC50 from the competitive FPA to DON was 229 ng∙mL-1

, a value which is

Page 125: PhD Fusarium

102

significantly higher than previously published values based on similar polyclonal assays

to assess hapten binding (e.g., Furzer et al., 2006; Maragos et al., 2002).

4.4.2 HCAb and cIgG Response

4.4.2.1 Protein A and G Chromatography

Fractionation of pre-immune, 42- and 70-day serum samples confirmed an increase

in HCAb and cIgG content after immunization with 15-DON-BSA. A greater proportion

of HCAb relative to cIgG was noted within fractionated pre-immune serum, while

immunization with 15-DON-BSA increased the relative proportion of cIgG in the 42- and

70-day hyper-immunized sera (Table 4.2). Absolute and relative quantities of HCAb and

cIgG fractions were in agreement with published values (Muyldermans, 2001; van der

Linden et al., 2000).

Table 4.2 HCAb and cIgG fractions of llama serum. Volumes of pre-immune, 42-

day and 70-day samples = 12.1, 12.5 and 15.5 mL, respectively.

IgG G1 (HCAb) G2 (cIgG) A1 (HCAb) A2 (HCAb)

Pre 42d 70d Pre 42d 70d Pre 42d 70d Pre 42d 70d

Vol. (mL) 2.3 2.5 2.8 6.0 6.4 8.5 1.8 1.9 2.1 2.0 1.8 1.7

Ratio* (% v/v) 19.0 19.8 18.3 49.6 50.8 56.4 14.9 15.1 14.0 16.5 14.3 11.3

Conc. (mg·mL-1

) 0.11 0.17 0.13 0.05 0.71 0.21 0.10 0.10 0.14 0.16 0.12 0.16

Amount (mg) ** 0.25 0.43 0.36 0.30 4.54 1.79 0.18 0.19 0.29 0.32 0.22 0.27

* % v/v ratio calculated as volume of fraction ÷ total volume.

** Amount = Conc. (mg·mL-1

) x Vol. (mL).

4.4.2.2 ELISA on Polyclonal Serum Fractions

Relative to pre-immune fractions, both cIgG and HCAb fractions from 42- and 70-

day sera had significantly greater binding to 15-DON-OVA than to OVA. The cIgG

Page 126: PhD Fusarium

103

fractions of 42- and 70-day serum samples had the greatest overall binding to 15-DON-

OVA. However, HCAb fractions also showed significant binding to 15-DON-OVA (data

not shown). Based on the positive HCAb ELISA results, a recombinant VHH phage-

display library was constructed using HCAb cDNA from both the 42- and 70-day sera.

4.4.3 Library Construction

The initial step in VHH library construction was PCR amplification of a mixture of

42- and 70-day variable heavy chain leukocyte DNA. Sense primer mixture “MJ1.2.3

Back” and hinge-specific anti-sense primer mixture “CH2b3 + CH2” (Table 4.1) annealed

to conserved regions of cIgG and HCAb DNA to produce PCR fragments of 620 – 690 bp

and 900 bp, respectively. VH/VHH–specific primers MJ7 and MJ8 (Table 4.1) amplified

620 – 690 bp HCAb DNA and to add SfiI restriction sites to produce 400 – 450 bp PCR

products which were gel purified and ligated into pMED1 phagemid. Library diversity

was determined by sequencing of 30 recombinant clones and by concurrent digestion of

PCR-amplified VHH fragments from individual colonies with 4-mer BstN1 restriction

endonuclease. Library size was determined to be approx. 109 primary recombinants.

4.4.4 Panning and Phage ELISA

The recombinant phage-displayed VHH library was panned against 15-DON-OVA

and OVA (control) through four rounds of panning. Favorable ratios of input and output

phage specific to the target mycotoxin were maintained through a continuous reduction in

15-DON-OVA conjugate coating for each round of panning and a switch in blocking

agents from OVA to casein between panning rounds two and three.

Page 127: PhD Fusarium

104

Phage ELISA conducted on ten randomly selected clones from the fourth round of

panning revealed three clones (phage # 2, 6, and 7) that bound to 15-DON-OVA, but not

to control proteins (i.e., OVA, BSA). Further phage ELISA screening of 100 randomly

selected output clones from the fourth round of panning produced 38 clones that bound to

15-DON-OVA, but not to control proteins, which suggested that ~ 40% of the phage

clones were hapten-specific (data not shown).

4.4.5 VHH Sequence Results

Nucleotide sequencing of positive phage ELISA clones revealed a single dominant

nucleotide sequence („NAT-267‟) encoding a VHH which bound to 15-DON-OVA, but

not protein controls. Relative to previously published VHH sequences, the NAT-267

sequence was unique (Figure 4.2).

FR-1 (1-25)

CDR-1 (26-35)

FR-2 (36-49)

CDR-2 (50-65)

1 10 20

.........│.........│.....

30

....│.....

40

....│.........

50 60

│..a.......│.....

QVQLEESGGGLVQAGGSLRLSCAAS GRTSSNTFVG WFRQAPGKEREFVA AIRRSDDRTYYAASVRG

FR-3 (66-94)

CDR-3 (95-102)

FR-4 (103-113)

70 80 90

....│.........│..abc.......│....

100

.....│abcdef..

110

.......│...

RFTISGDSAKNVVALQMSSLRPEDTAVYYCAA TRTWLVTGQSDYPY WGQGTQVTVSS

Figure 4.2 Amino acid sequence of NAT-267 VHH from fourth round of panning.

Recombinant VHH CDR (underlined) and FR delimitations as well as

amino acid insertions (52a, 82a,b,c, 100a,b,c,d,e,f) defined according to

Kabat numbering scheme (Kabat et al., 1991).

Although the sequence of the Framework Region (FR) was similar to that of the

hapten binder „RR6-R2‟ (Spinelli et al., 2001), residues within the three complementarity

determining regions (CDR) were novel. As commonly observed for VHH hapten binders,

Page 128: PhD Fusarium

105

NAT-267 did not contain cysteine residues within CDR 1 or 3, which is commonly

believed to serve as the basis of an interloop disulfide bridge within recombinant VHH

protein structure (Figure 4.2). Except for clones that bound only to control proteins, all

VHH sequences isolated had relatively long predicted CDR-3 regions (12 – 18 amino

acids) (data not shown).

4.4.6 Expression of Soluble VHH Protein

Recombinant protein fractions of approximately 16 kDa in size, containing the

mNAT-267 VHH were pooled. From the absorption measurement at 280 nm, a yield of 3

to 6 μg of purified protein per liter of E. coli culture was calculated. The purified protein

was concentrated, by ultrafiltration, to 1.3 mg∙mL-1

in PBS; no aggregation was observed.

Purification of pNAT-267 VHH of approximately 130 kDa in size was successful

using standard IMAC procedures. Aggregated pentabodies were removed by size

exclusion chromatography on Superdex 200 column. Overall yield of pNAT-267 was

determined to be 3 to 9 μg∙L-1

of E. coli culture. Ultrafiltration was used to obtain a final

pentamer concentration of 1.0 mg∙mL-1

in PBS; no aggregation was observed.

4.4.7 VHH Affinity Measurements

4.4.7.1 Fluorescence Polarization

Initial FPA analysis based on step-wise titrations of both mNAT-267 and pNAT-

267 confirmed identical binding of both recombinant proteins to DON-fluorescein tracer

diluted 1:8,000 in skim milk (1 mg∙mL-1

) (data not shown). Plots, based on P/PO versus

concentration of free hapten, were used to determine IC50 values, or concentration

required for 50% inhibition of binding. Competitive FP inhibition assays determined an

Page 129: PhD Fusarium

106

IC50 value of 0.82 μM for free DON versus 56-day polyclonal llama serum (Figure 4.3A).

Neither the monomer nor pentamer bound to soluble DON. Likewise, FP inhibition with

3-AcDON determined an IC50 value of 0.62 μM for 56-day polyclonal llama serum, but

no binding for NAT-267 monomer or pentamer (Figure 4.3B). FP inhibition with 15-

AcDON determined an IC50 value of 1.42 μM for 56-day polyclonal serum, and IC50

values of 1.24 and 0.50 μM for NAT-267 monomer and pentamer VHH, respectively

(Figure 4.3C). FP inhibition assays with diacetoxyscirpenol (DAS), neosolaniol (NEO),

and T-2 trichothecene toxins indicated no specific binding to polyclonal serum, mNAT-

267 or pNAT-267 VHH (data not shown).

4.4.7.2 Surface Plasmon Resonance

Binding kinetics of mNAT-267 VHH binding to 15-DON-HRP were determined by

SPR (BiaCore™) analysis. Monomer was injected over HRP control and 15-DON-HRP

bound to a SPR sensor chips at concentrations of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10 μM to

generate an overlay sensogram of mNAT-267 binding to 15-DON-HRP conjugate (Figure

4.4). Trace amounts of aggregate mNAT-267 VHH contamination were likely responsible

for second phase binding to 15-DON-HRP conjugate, which could be seen on sensorgram

traces as trends with slightly positive slope. To minimize this effect, data were collected

at the highest flow rate (100 μL∙min-1

), and binding kinetics were measured as soon as

first phase binding reached steady state, or „Req‟ (Figure 4.4).

Page 130: PhD Fusarium

107

0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.00.00

0.25

0.50

0.75

1.00

1.25Serum

Monomer

Pentamer

Log[DON ng/mL]

P/P

o

0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.00.00

0.25

0.50

0.75

1.00

1.25Serum

Monomer

Pentamer

Log[DON ng/mL]

P/P

o

0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.00.00

0.25

0.50

0.75

1.00

1.25Serum

Monomer

Pentamer

Log[DON ng/mL]

P/P

o

Figure 4.3 FP inhibition of mNAT-267 and pNAT-267 VHH binding and 56-day

polyclonal llama serum binding to DON-fluorescein tracer with DON

toxin (A); 3-AcDON (B); and 15-AcDON (C) from 5 μg∙mL-1

to 9.8

ng∙mL-1

. IC50 of llama serum binding to: DON; 3-AcDON; and 15-

AcDON = 0.82 μM; 0.62 μM; and 1.42 µM, respectively. IC50 for mNAT-

267 and pNAT-267 binding to 15-AcDON = 1.24 μM and 0.50 μM,

respectfully. For each assay, error bars represent ±1 SD, number of

replicates = 6. DON- fluorescein tracer within each assay diluted 1:8,000

in PBS.

A

B

C

Page 131: PhD Fusarium

108

0 5 10 15 20

0

50

100

150

200

Time (s)

Re

sp

on

se

(R

U)

Figure 4.4 Overlay sensorgrams of mNAT-267 VHH steady state (Req) binding to 15-

DON-HRP conjugate. mNAT-267 VHH injected over HRP reference and

15-DON-HRP surfaces at the concentrations of 1 (lowest trace), 2, 3, 4, 5,

6, 7, 8, 9 and 10 (highest trace) μM for 6 seconds.

An assessment of steady state binding (Req) relative to concentration of mNAT-267

VHH confirmed first-order binding kinetics (Figure 4.5A). A Scatchard plot of Req

binding at each mNAT-267 VHH concentration relative to actual binding demonstrated

binding to immobilized 15-DON-HRP with a dissociation constant (KD) of 5 µM (Figure

4.5B). Independent association and dissociation rates were too fast to calculate.

Free DON (50 and 500 μM) was mixed with mNAT-267 VHH (5 μM) to test

competitive inhibition of monomer binding to bound 15-DON-HRP. No competitive

inhibition was observed indicating that mNAT-267 had no affinity for soluble DON.

This observation was further confirmed by SPR analysis of the interaction between

pNAT-267 VHH and soluble DON. After capture of pNAT-267 to the SPR sensor chip,

no binding interaction was noted between the pentamer and concentrations of soluble

DON ranging from 0.1 to 2 mM (data not shown).

Req

Page 132: PhD Fusarium

109

0 2 4 6 8 10 120

50

100

150

200

Conc (M)

Ste

ad

y s

tate

bin

din

g (

RU

)

0 25 50 75 100 125 150 1750

10

20

30

40

50

Steady state binding (RU)

Ste

ad

y s

tate

bin

din

g/C

(R

U/

M)

Figure 4.5 (A) Steady state (Req) affinity binding (RU) mNAT-267 VHH to 15-DON-

HRP conjugate for ten monomer concentrations, as shown in Figure 4.4.

(B) Scatchard plot of steady state „Req‟ binding (RU) relative to Req / C

(mNAT-267 VHH concentration) (RU·μM-1

). Dissociation constant (KD)

= 5 μM.

SPR analysis was not used to assess interaction of mNAT-267 or pNAT-267 to 3-

AcDON and 15-AcDON. Unfortunately, it was not possible to prepare functional

solutions since the relative water solubilities of these acetylated toxins were much lower

than DON. Furthermore, it was impractical to dissolve the acetylated toxins in organic

solvent (e.g., acetone) followed by dilution in PBS since the solvent adversely affects

SPR measurement relative to control assessments.

Page 133: PhD Fusarium

110

4.4.7.3 CI ELISA

Checkerboard immunoassay titrations confirmed optimum binding of NAT-267

VHH (relative to OVA controls) based on dilution of monomer and pentamer at 0.38

μg∙mL-1

within ELISA wells coated with 15-DON-OVA at 0.5 μg∙well-1

. Although CD

ELISA consistently showed that 15-AcDON inhibited mNAT-267 and pNAT-267

binding to 15-DON-OVA, it was not possible to precisely define the respective IC50

values below 10 µM (data not shown). Binding of monomer and pentamer to 15-DON-

OVA coated plates was not affected by competition with serial dilutions of DON or 3-

AcDON at concentrations ranging from 8.4 nM (0.0025 μg∙mL-1

) to 17 μM (5.0 μg∙mL-1

).

4.5 Discussion

DON toxin and its acetylated metabolites are low molecular weight (296 to 338 Da)

highly stable trichothecene mycotoxin contaminants of maize and small grain cereals

(Desjardins, 2006). Trichothecene mycotoxins, a group of over 148 related compounds,

are known to cause severe toxicosis in humans and farm animals. For example, swine

vomit and display a wide variety of other symptoms of toxicosis including

immunosuppression, after consumption of contaminated feed (Canady et al., 2001). Most

countries have legislation and strict guidelines regarding acceptable levels of mycotoxins

in contaminated cereals (Rotter et al., 1996).

Immunoassay techniques that can detect and quantify DON and structurally-similar

mycotoxins use polyclonal serum and monoclonal antibodies. These assays provide

highly accurate mycotoxin measurement. However, the development of toxin-specific

immunoglobulins is difficult since it typically involves the repeated immunization of

Page 134: PhD Fusarium

111

animals and the use of highly specialized, laborious techniques to produce specific

polyclonal sera or mAbs with high specificities and affinities.

The development of toxin-specific recombinant antibody (rAb) fragments such as

single-chain variable fragment (scFv) and VHH provides an alternative means of

producing low-cost and abundant quantities of high affinity binders within common

recombinant expression hosts. Although scFv fragments often display binding affinities

equivalent to the parent mAb (e.g., Wang et al., 2007), in practical terms, difficulties in

obtaining purified protein, and challenges associated with protein degradation and the

formation of insoluble aggregates and inclusion bodies within bacterial cultures (Choi et

al., 2004), has limited large-scale application of this technology (Arbabi-Ghahroudi et al.,

2005). We pursued the development of DON-specific single domain VHH fragments as

an alternative to the conventional IgG mAb and scFv rAb formats, as VHH are smaller

and generally have favorable expression from microbial cells.

This study describes the successful isolation and cloning of a single-domain VHH

gene from a hyperimmunized llama library, followed by soluble expression as a

monomeric and pentameric VHH protein, and characterization of binding kinetics to

various DON toxins and their respective protein conjugates. To our knowledge, this is

the first example of the development of a single-domain VHH binder to a mycotoxin.

Before VHH library construction and panning, we confirmed a robust and toxin-

specific response to immunization with 15-DON-BSA within peripheral leukocytes

(Figure 4.1). Serial dilution of whole and cIgG and HCAb fractionated serum tested by

ELISA confirmed a polyclonal response specific for 15-DON-OVA. An initial

Page 135: PhD Fusarium

112

competitive FP inhibition assay confirmed that whole llama serum had affinity for free

DON toxin (IC50 = 229 ng·mL-1

). Based on these results, we concluded that the methods

and reagents used to generate a polyclonal response were optimized. Although the

fractionated llama serum demonstrated a greater increase in cIgG content relative to

HCAb fractions (Table 4.2), we concluded that the HCAb response was pronounced

enough to warrant construction of a hyper-immunized VHH library.

Peripheral leukocytes were used as sources of mRNA for the cloning of VHH coding

sequences to construct the hyperimmunized library (reviewed in Muyldermans, 2001).

Variable regions of HCAb and IgG immunoglobulins were amplified by nested PCR

based on hinge-specific primers. VHH-specific PCR products were excised from an

agarose gel and re-amplified with hinge-specific primers. The latter primers added the

required SfiI restriction endonuclease enzyme site onto amplified VHH DNA to enable

digestion and ligation into the pMED1 phagemid system. Results based on colony PCR,

DNA fingerprinting, and successive plate dilutions of transformed TG1 E. coli showed

the VHH library to be diverse and of sufficient size to proceed with biopanning.

Four rounds of panning against decreasing concentrations of 15-DON-OVA coupled

with increased washing stringency yielded a single dominant VHH clone (termed NAT

267) at a frequency of 38%. Subsequent phage ELISA confirmed that NAT-267 bound to

15-DON-OVA with little or no binding to OVA (control). The panning process was

deemed successful despite the fact that it was not based on a process of affinity selection

through elution with free DON hapten which may have improved the selection of high

affinity toxin-specific VHH binders (Sheedy et al., 2007).

Page 136: PhD Fusarium

113

The NAT-267 VHH gene was well expressed in monomeric and pentameric forms

within E. coli. The relatively high concentrations of expressed VHH are comparable to

the values published for scFv and Fab (Maynard and Georgiou, 2000). The high yield of

monomer and pentamer forms of NAT-267 was attributed to the soluble and inherently

robust nature of recombinant VHH proteins (Ghahroudi et al., 1997; Goldman et al., 2006;

Ladenson et al., 2006). Direct ELISA assays confirmed that soluble VHH monomer and

pentamer bound to 15-DON-OVA, but not to OVA (control). Given identical microtiter

plate dilution regimes, the ELISA signal for the pentamer was consistently higher than for

equivalent monomer concentrations. This result suggested that the dominant VHH clone

was selected based on avidity of a multivalent phage during panning to hapten-protein

conjugate rather than affinity to free hapten, which is a limitation when panning with

multivalent Ab-displaying phage.

A series of assays to determine VHH monomer and pentamer binding specificity

confirmed that purified NAT-267 monomer and pentamer fragments recognized 15-

AcDON (Figure 4.3C) but not DON (Figure 4.3A), 3-AcDON (Figure 4.3B), or the other

trichothecene toxins tested (diacetoxyscirpenol, neosolaniol, and T-2 toxin).

The free hydroxyl group at position 15 of DON and 3-AcDON may have interfered

with NAT-267 VHH binding. Conversely, the presence of an acetyl ester group (≈ 42 Da)

at position 15 of the trichothecene structure (Figure 4.6) may have altered the molecular

structure and conformation to impart VHH binding to 15-AcDON. Given that the DON

toxin was conjugated to carrier proteins by a hemi-succinate linker at position 15 for both

the antigen (15-DON-BSA) and the coating conjugate (15-DON-OVA) used for panning,

Page 137: PhD Fusarium

114

it was postulated that the VHH paratope recognized an epitope at or in close proximity to

position 15 of the DON molecule.

Trichothecene MW

a

(Da)

Oxygenation and Esterification at Positionb

C-3 (R1) C-4 (R2) C-7 (R3) C-8 (R4) C-15 (R5)

3-Acetyl-DON 338 OAc H OH =O CH2-OH

15-Acetyl-DON 338 OH H OH =O CH2-OAc

Deoxynivalenol 296 OH H OH =O CH2-OH

Diacetoxyscirpenol 366 OH OAc H H CH2-OAc

Neosolaniol 382 OH OAc H OH CH2-OAc

T-2 Toxin 466 OH OAc H OIsoval CH2-OAc a MW = Molecular Weight

b OAc = acetyl ester. OIsoval = isovalerate ester.

Figure 4.6 Structure, composition, and molecular weight of trichothecene mycotoxins

used in this study (Desjardins, 2006).

Highly-specific VHH binding to 15-AcDON was therefore regarded as a function of

both the linker used to fix DON to carrier proteins and the panning methods, while the

physico-chemical properties of these ligands, such as water solubility and molecular size,

may have had little impact on VHH specificity. It would be interesting to test this

hypothesis by panning the VHH library against a DON-protein conjugate with different

linker chemistry and/or a different conjugation position (e.g., 3-DON-OVA).

Page 138: PhD Fusarium

115

Based on competitive FP assays, IC50 values determined by competition with 15-

AcDON were 1.24 and 0.50 μM for the VHH monomer and pentamer, respectively.

These values are comparable to IC50 of 1.42 µM for 15-AcDON using 56-day polyclonal

llama serum in a competitive FP assay (Figure 4.3C). Unfortunately, it was not possible

to validate FP results by CD ELISA since 15-AcDON inhibition values for both the

monomer and pentamer could not be precisely determined. The inherent difficulty in

establishing consistent IC50 values by CD ELISA was attributed to the relatively fast

dissociation rate of both the NAT-267 monomer and pentamer coupled with the

continuous removal of VHH from the non-equilibrium binding system by successive

washing steps after initial binding. Furthermore, the limited solubility of 15-AcDON

prevented SPR analysis of its binding to chip-immobilized NAT-267 VHH. However,

SPR analysis of NAT-267 monomer binding to chip-immobilized 15-DON-HRP yielded

a dissociation constant (KD) of 5 µM, which is similar to the IC50 determined in the

competitive FP assay using NAT-267 (1.24 μM).

Unlike reports associated with poor scFv expression and isolation (Choi et al.,

2004), our monomeric and pentameric VHH fragments were found to be well expressed,

stable and not prone to aggregation or formation of insoluble inclusion bodies.

Furthermore, unlike scFv for DON toxins, NAT-267 VHH was highly specific to the 15-

AcDON moiety with no cross-reactivity to 3-AcDON or DON. Such specificity could aid

in toxin-specific detection as a compliment to existing mAb assays. For example, the

mAb (IgG) developed by (Maragos and McCormick, 2000) was reported to be very

specific for DON and 3-AcDON with little or no affinity for 15-AcDON. When used in

Page 139: PhD Fusarium

116

conjunction this mAb-based ELISA, NAT-267 VHH would allow quantification of all

three toxins (i.e., DON, 3-AcDON, and 15-AcDON). Furthermore, the mAb-based assay

developed by Sinha et al. (1995), which detects both 15-AcDON and DON (i.e., 15-

AcDON + DON), could be used in conjunction with NAT-267 VHH to determine the

exact concentration of 15-AcDON as well as DON.

We compared NAT-267 VHH in terms of affinity and specificity with the other

„hapten-specific‟ VHH fragments that have been isolated from both naïve (Sheedy et al.,

2006; Yau et al., 2003b) and hyper-immunized llama libraries (Alvarez-Rueda et al.,

2007; Frenken et al., 2000; Ladenson et al., 2006; Spinelli et al., 2000). For example,

the indoleacetic acid (IAA)-binding VHH CSF2A, isolated from a naïve llama library, had

a KD of 20 µM for free IAA (Sheedy et al., 2006). Unlike NAT-267, CSF2A cross-

reacted to structurally-similar analogues. Furthermore, a picloram-specific VHH, 3-ID2,

isolated from a naïve ribosome-display llama library had an IC50 of 800 µM for the

hapten picloram (Yau et al., 2003b). In both cases, these naïve-library-based VHHs had

much lower affinities than did NAT-267.

Unlike the hapten-specific VHHs isolated from naïve libraries (Sheedy et al., 2006;

Yau et al., 2003b), the VHH sequences isolated from hyper-immunized llama libraries

(e.g., Alvarez-Rueda et al., 2007; Ladenson et al., 2006; Spinelli et al., 2000) were in

agreement with our work in that each VHH isolated was found to be hapten-specific with

minimum cross-reactivity to structurally-similar analogues and their IC50 values as

determined by competition with free hapten were in the µM range. For example, a

caffeine-specific VHH, VSA2, had an IC50 of ca. 154 µM with minimum cross-reactivity

Page 140: PhD Fusarium

117

to structurally-similar haptens (Ladenson et al., 2006). Similarly, seven methotrexate

(MTX)-specific VHHs isolated from a hyper-immunized llama library (Alvarez-Rueda et

al., 2007) had nanomolar affinities (KD = 29 - 515 nM) for MTX-BSA; however, we

estimate the affinity for the free hapten, which was not determined by the authors, to

range from 10 and 70 µM based on IC50 values (Alvarez-Rueda et al., 2007). Finally, the

KD for an azo dye (Reactive-Red-6; M.W. = 728 Da)-specific VHH, known as RR6-R2,

was determined to be 22 nM. Although the affinity of this VHH is much higher than

NAT-267, the unusually high affinity of RR6-RR2 was attributed to a co-ordination bond

between the copper of the dye and a histidine moiety in the VHH, rather than to a normal

VHH-hapten interaction (Spinelli et al., 2000). Therefore, based upon the results of these

authors (Alvarez-Rueda et al., 2007; Ladenson et al., 2006; Sheedy et al., 2006; Spinelli

et al., 2000; Yau et al., 2003b), NAT-267 has the lowest hapten-based IC50 of any hapten-

specific VHH yet discussed in the literature.

The four scaffold FR residues within the NAT-267 VHH sequence had over 80%

similarity with the VHH gene sequences of CSF2A (Sheedy et al., 2006), VSA2

(Ladenson et al., 2006), and RR6-R2 (Spinelli et al., 2000). Based on nucleotide acid

identity, NAT-267 VHH belongs to camelid VHH sub-family I (Harmsen et al., 2000).

Furthermore, each of the three CDRs of „NAT-267‟ has unique amino acid residues.

Contrary to published VHH sequences which have been shown to bind to large protein

antigens, the CDR3 of NAT-267 is not constrained by an inter-loop disulfide bond to

CDR1 since no cysteine residues were present within either region. Therefore, other than

for the disulfide bridge formed between FR Cys-22 and Cys-92 (Figure 4.2), it is unlikely

Page 141: PhD Fusarium

118

that other disulfide bonds are present within NAT-267. Based on these observations, we

concluded that the CDR1 to CDR3 inter-loop disulfide bonds present within many

published sequences of VHHs are important for facilitating VHH binding to large

antigenic proteins but not to low molecular weight haptens (i.e. < 600 Da). Therefore, we

hypothesize that the highly-specific binding of NAT-267 VHH to the haptens 15-AcDON

and DON-fluorescein as well as to the protein conjugates 15-DON-OVA and 15-DON-

HRP is likely conveyed by novel sequences within the non-crosslinked CDR residues.

Furthermore, since the VHH paratope is not regarded to be a flat surface (Spinelli et al.,

1996), we further hypothesized that high affinity interaction of NAT-267 VHH with the

two haptens and coating conjugates occurs within flexible grooves or cavities between the

three CDR regions (Spinelli et al., 1996; 2000; 2001).

4.6 Conclusions

This research further confirms that hapten-specific recombinant VHH fragments can

be selected from a hyper-immunized llama VHH library. Both monomer and pentamer

forms of the NAT-267 dominant clone selected by four rounds of panning was specific to

the mycotoxin 15-AcDON as determined by competitive binding assay, and there was no

cross-reactivity to closely related moieties of DON toxin. Although the dominant clone

was both novel and highly specific to an acetylated moiety of the target mycotoxin, the

nucleotide sequences for all four framework regions were similar to previously-isolated

hapten-specific VHH sequences. Further research to assess different panning strategies

and various affinity maturation strategies as a means of improving cross reactivity and

overall binding affinity are now being pursued. Furthermore, since 15-AcDON has been

Page 142: PhD Fusarium

119

shown to have a similar eukaryotic cytotoxicity as DON (Binder et al., 1997;

Poppenberger et al., 2003), we are focusing on expression of NAT-267 VHH in

eukaryotic test systems such as yeast and Arabidopsis thaliana to determine whether the

negative effects of the toxin may be reduced or eliminated.

4.7 Acknowledgements

The authors of this paper would like to thank Tomoko Hirama (NRCC, Ottawa,

Canada) for conducting the SPR analysis and Hong Tong-Sevinc (NRCC, Ottawa,

Canada) for expression and purification of the VHH clones. This research was supported

by the Natural Sciences and Engineering Research Council (NSERC) of Canada, the

Canada Research Chairs Program, the National Research Council (NRC) of Canada, and

the Ontario Ministry of Agriculture and Food (OMAF).

Page 143: PhD Fusarium

120

5 RESEARCH CHAPTER 2: INTRACELLULAR EXPRESSION OF A

SINGLE-DOMAIN ANTIBODY REDUCES CYTOTOXICITY OF 15-

ACETYL-DEOXYNIVALENOL IN YEAST.

5.1 Abstract

15-Acetyl-Deoxynivalenol (15-AcDON) is a low molecular weight sesquiterpenoid

trichothecene mycotoxin associated with Fusarium Ear Rot of maize and Fusarium Head

Blight of small grain cereals. The accumulation of mycotoxins such as deoxynivalenol

(DON) and 15-AcDON within harvested grain is subject to stringent regulation as both

toxins pose dietary health risks to humans and animals. These toxins inhibit peptidyl

transferase activity which in turn limits eukaryotic protein synthesis. To assess the ability

of intracellular antibodies (intrabodies) to modulate mycotoxin-specific cytotoxocity, a

gene encoding a camelid single-domain antibody fragment (VHH) with specificity and

affinity for 15-AcDON was expressed in the methylotropic yeast Pichia pastoris.

Cytotoxicity and VHH immunomodulation were assessed by continuous measurement of

cellular growth. At equivalent doses, 15-AcDON was significantly more toxic to wild-

type P. pastoris than was DON. In turn, DON was orders of magnitude more toxic than

3-AcDON. Intracellular expression of a mycotoxin-specific VHH within P. pastoris

conveyed significant (p = 0.01) resistance to 15-AcDON cytotoxicity at doses ranging

from 20 to 100 µg∙mL-1

. We also documented a biochemical transformation of DON to

15-AcDON to account for the attenuation of DON cytotoxicity at 100 and 200 µg∙mL-1

.

The proof of concept established within this eukaryotic system suggests that in planta

VHH expression may lead to enhanced tolerance to mycotoxins and thereby limit

Fusarium infection of commercial agricultural crops.

Page 144: PhD Fusarium

121

5.2 Introduction

Fusarium head blight (FHB) of cereals and Fusarium Ear Rot of maize are caused

by morphologically similar species (F. graminearum, F. culmorum, etc.) common

throughout global agricultural regions. With few exceptions, Fusarium epidemics are

characterized by cyclical and highly aggressive infection of commercial crops with

economic impacts on food and feed industries that are immediate and far reaching. For

example, losses associated with the most recent Fusarium outbreak in North America in

the 1990s were estimated to range from $1.3 to 3.0 billion USD (Demcey Johnson et al.,

2003).

A toxin class commonly found within agricultural commodities infected by

Fusarium are trichothecene mycotoxins. Trichothecenes represent a highly diverse group

of over 180 sesquiterpenoid low molecular weight (typically 200-500 Da) mycotoxins

characterized by a tricyclic ring structure containing a double bond at C-9,10 and an

epoxide group at C-13 (Desjardins, 2006). Regardless of size and structural composition,

trichothecenes are potent inhibitors of eukaryotic protein synthesis with specific activity

on ribosomal protein L3 within the 60S subunit resulting in inhibition of peptidyl

transferase activity (Carter and Cannon, 1977; Rocha et al., 2005). Although the

capacity to inhibit protein synthesis is regarded as central to trichothecene cytotoxicity

(Rotter et al., 1996; Ueno, 1984), adverse effects on eukaryotic cells may actually be

attributed to dysregulation of cellular signaling and alterations in downstream gene

expression (Pestka, 2008). As a result, trichothecenes such as deoxynivalenol (DON),

15-acetyl-deoxynivalenol (15-AcDON), and 3-acetyl-deoxynivalenol (3-AcDON) (Figure

Page 145: PhD Fusarium

122

5.1) are considered to be inherently hazardous food and feed borne contaminants

(Desjardins, 2006; D‟Mello et al., 1999).

Trichothecene MW

a

(Da)

Oxygenation and Esterification at Position

C-3 C-15

3-Acetyl-DON 338 O-CO-CH3 OH

15-Acetyl-DON 338 OH O-CO-CH3

Deoxynivalenol 296 OH OH a MW = Molecular Weight

Figure 5.1 Structure, composition, and molecular weight of trichothecene mycotoxins

used within this study (Desjardins, 2006).

Numerous studies have demonstrated a correlation between in planta DON

accumulation and Fusarium virulence in susceptible cultivars of wheat (Mesterházy,

2002) and maize (Harris et al., 1999a). Based on these findings, mechanisms which

convey innate and acquired host plant resistance to DON and other trichothecene toxins

have received considerable attention. To date, in planta trichothecene resistance has been

achieved through mechanisms which alter targeted proteins within host cell ribosomes

(e.g., Harris and Gleddie, 2001; Mitterbauer et al., 2004), promote metabolic

Page 146: PhD Fusarium

123

transformation to less toxic forms: e.g., DON-glucosyl conjugate (Poppenberger et al.,

2003) or to 3-AcDON (Alexander, 2008), and/or reduce intracellular concentrations to

effectively limit mycotoxin exposure to sensitive cellular targets. Collectively, such

research can be applied to impart novel mechanisms of trichothecene resistance in higher

order plants.

Yeast is well suited as a eukaryotic model organism to identify and validate

mechanisms involved in host plant resistance to mycotoxins (Mitterbauer and Adam,

2002; Mitterbauer et al., 2004; Poppenberger et al., 2003). Test systems based on yeast

offer cost-effective convenience and flexibility as one can validate a wide range of novel

detoxification mechanisms within a short period of time at a minimal cost using common

/ non-specialized lab equipment. Assessment of mycotoxin resistance mechanisms is

likewise straightforward as reproducible treatment-specific effects can be precisely

determined based on simple measurements of cellular growth and function over time.

Single-domain heavy chain antibody fragments (i.e., VHH) from the camelidae

heavy chain IgG subfamily, are among the smallest functional recombinant antibody

(rAb) fragments at 14-15 kDa. VHH fragments exhibit the same exquisite specificity as

larger immunoglobulins, with added biochemical advantages of high solubility, stability,

and robust expression in various recombinant systems (Holliger and Hudson, 2005;

Muyldermans, 2001). VHH fragments have been generated against low molecular weight

ligands (haptens) and toxins (e.g., Doyle et al., 2008). Based on favorable physico-

chemical properties and efficacy against a wide range of antigens, single domain rAb

fragments have been developed and tested as immuno-therapeutic reagents with

Page 147: PhD Fusarium

124

applications ranging from pharmacology (Holliger and Hudson, 2005; Saerens et al.,

2008) to plant science (Jobling et al., 2003).

This paper demonstrates that intrabody expression of a VHH fragment isolated from

a hyper-immunized phagemid library with affinity for 15-AcDON (Doyle et al., 2008) can

impart real-time immunomodulation of mycotoxin-specific cytotoxicity within a model

eukaryotic system. Pichia pastoris was selected as the host organism based on an

expected high level expression of functional VHH intrabody fragments and anticipated

sensitivity to 15-AcDON. This system was established as a „proof of concept‟ to

demonstrate that intrabody expression of recombinant VHH fragments could impart a

novel means of mycotoxin-specific resistance.

5.3 Experimental Procedures

5.3.1 VHH Genes

NAT-267 VHH DNA sequence (GenBank: EU676170.1) (Doyle et al., 2008) was

used to design a P. pastoris codon optimized version of the gene to which was added 3‟

HA and 6His epitope tags, as well as EcoRI and XbaI cloning sites at the respective 5‟

and 3‟ ends (GeneArtTM

, Toronto, ON, Canada). A non-specific VHH gene (B-24)

isolated from a hyper-immunized phagemid llama library with confirmed non-specificity

for 15-AcDON, or any other trichothecene mycotoxin, was used as a VHH intrabody

control. Both VHH fragments were of the same immunoglobulin family and had similar

molecular weights and isoelectric points. Like NAT-267, B-24 VHH DNA was PCR

amplified to include 3‟ HA- and 6His- epitope tags and restriction cloning sites EcoRI

and XbaI with the following gene-specific primers: VHH-B24For (5‟-GGAATTCCATG

Page 148: PhD Fusarium

125

CAGGTAAAGCTGGAGGAG-5‟) and VHH-B24Rev (5‟-GGGTCTAGACCCTATGCT

CGGCCGGAACCGTAG-3‟). A homology chart of nucleotide and peptide sequences of

NAT-267 and B-24 VHHs is provided to show respective framework (FR) and

complementarity determining regions (CDR), endonuclease sites and epitope tags (Fig.

S1 in Supplementary Information). VHH DNA inserts (~ 0.46 kb) were ligated into

pPICZB vector (Invitrogen Inc., Carlsbad, CA, USA). Constructs were sequenced by

StemCore Labs (Ottawa, ON, Canada) using pPICZB-specific primers: 5‟ AOX1For (5 ‟-

GACTGGTTCCAATTGACAAGC-3‟) and 3‟ AOX1Rev (5‟-GCAAATGGCATTCTGA

CATCC-3‟).

5.3.2 Transformation

Expression constructs (Figure 5.2) were linearized with SacI and electroporated into

P. pastoris strain KM71H (Invitrogen Inc., Carlsbad, CA, USA). Transformants

Figure 5.2 Diagram of pPICZB expression vector (3745 bp) with VHH gene (NAT-

267 = 387 bp, B-24 = 390 bp). Legend in the box shows schematic

representation for each component of transgenic DNA for expression in P.

pastoris.

were plated onto yeast extract, peptone, dextrose, sorbitol (YPDS) agar containing 100

µg∙mL-1

zeocin and incubated for 3 days at 30°C until colonies formed. Ten colonies

5’ 3’ EcoRI XbaI

5’AOX1

Promoter

VHH DNA HA-Tag

6His-Tag 3’AOX1

Terminator

Zeocin Resistance

Gene

Page 149: PhD Fusarium

126

from each transformant were re-streaked on yeast extract, peptone and dextrose (YPD)

agar plates with 100 µg∙mL-1

zeocin to ensure pure clonal isolates.

5.3.3 Induction of VHH Expression

Single colonies of VHH and pPICZB control transformants were used to inoculate 5

mL YPD media. Cultures were grown overnight (30°C, 300 rpm). One mL of each

culture was used to inoculate 100 mL of minimal glycerol medium with histidine

(MGYH) and cultured in 1-L baffled flasks (30°C, 250-300 rpm) for 1 day. Cells were

harvested with centrifugation (3,000g x 5 min.) at room temp. To induce VHH

expression, Pichia cells were resuspended in 20 mL of minimal methanol medium with

histidine (MMH) and transferred to 125-mL baffled flasks and incubated at 30°C with

300 rpm shaking. Methanol (100%) was added to final conc. of 0.5% (% v/v) every 24

hours. Western blot analysis of ten VHH (NAT-267 and B-24) transformants from 0, 24,

48, 72, 96, and 120 hours post-induction was used to select clones and induction time

points corresponding to the highest overall protein expression.

5.3.4 Preparation of Soluble VHH Extracts and Western Blot Analysis

Cell pellets were thawed on ice and resuspended in 100 µL lysis buffer (50 mM

Tris, pH 7.5, 1mM EDTA, 0.5% Triton-X 100, 1 mM DTT, 1X Roche‟s Complete

Protease Inhibitor Cocktail). Equal volumes of acid-washed 0.5-mm glass beads were

added to each resuspended pellet, followed by successive 30-second cycles of vortexing

and incubation on ice for a total of ten cycles. Cell lysate samples were clarified by

centrifugation (14,000 rpm x 10 min.) at 4°C. One-hundred microliters of 2X-SDS

loading buffer (BioRad Laboratories, Mississauga, ON, Canada) was added to the

Page 150: PhD Fusarium

127

supernatant followed by boiling (95°C x 5 min.) and electrophoresis on 12% SDS-PAGE

gels. Samples were transferred to PVDF membrane (BioRad, Mississauga, ON Canada)

and blocked overnight in 1% blocking reagent (Roche, Laval, QC, Canada) in TBS at

4°C. VHH intrabody fragments were detected by probing for 1 h with rabbit anti-HA IgG

primary antibody (Sigma, St. Louis, MO) diluted 1:1000 in 0.5% blocking buffer

followed by 2 washes with TBS + 0.1% Tween (TBST) and 2 washes with TBS + 0.5%

blocking reagent. Secondary antibody (mouse anti-rabbit IgG horseradish peroxidase

conjugate; Jackson, Immuno-Researcher Labs, West Grove, PA, USA), diluted 1:50,000

in 0.5% blocking buffer, was used to probe the membrane for 1 h, followed by 4

consecutive 15 min washes with TBST. VHH proteins were visualized with ECL Plus

Western Blotting Detection System (GE Healthcare, Baie d‟Urfé, QC, Canada).

Transformants with the highest VHH intrabody expression levels were used in subsequent

in vivo cytotoxicity assays.

5.3.5 Cytotoxicity Assays

Cells were induced as previously described. Forty-eight hours post induction, P.

pastoris cells were diluted to an OD600 of 0.25 in YPD and incubated for 4 hours (30°C,

300 rpm). After this “recovery period”, cells were diluted to an OD600 of 0.1 in YPD, and

immediately transferred (250 µL·well-1

) to an F96 microwell assay plate (Nalge Nunc

Inc., Naperville, IL, USA). Pre-calibrated concentrations of the four ribotoxin treatments

(15-AcDON, DON, 3-AcDON, or cycloheximide) prepared in DMSO and a DMSO only

control were added to P. pastoris cells contained in the wells of assay plates. All

treatments, respective controls, and cell-free wells containing only YPD media (i.e. blank

Page 151: PhD Fusarium

128

wells) were established in triplicate on each plate. Plates were sealed with Progene

pressure-sensitive optical sealing film (Ultident, St.-Laurent, QC, Canada) prior to

initiation of cytotoxicity assays. Cellular growth (30°C, 300 rpm) measured in real time

based on measurement of OD620 values at 25 min intervals, following 5 minutes of

shaking (600 rpm) in a Polarstar Optima Microplate Reader (BMG Labtech, GMbH,

Offenberg, Germany). Assay results were considered valid if similar and reproducible

effects were observed in three separate experiments, conducted in triplicate under the

same test parameters.

5.3.6 Data Analysis

Results were presented as P. pastoris growth curves over a 24-h time period. Mean

(n = 3) OD620 values for VHH and pPICZB (empty vector) transformants used in ribotoxin

and control treatments, minus OD620 values of YPD media (blank) wells, were plotted

against time (i.e. 25-min intervals). Standard error values and paired t-tests (p = 0.05 and

0.01) were used to assess statistically significant differences at the end of each 25-min

time interval.

Time to doubling of initial P. pastoris OD620 cellular growth values were calculated

from cytotoxicity assay data to assess relative toxicity of each ribotoxin treatment. A

comparative ranking of various ribotoxin treatments on P. pastoris growth was calculated

based on time required to double OD620 values of t = 0.0h pPICZB empty vector

transformants.

Differential area under curve (ΔAUC) values were calculated to quantify relative

differences in P. pastoris growth over the full time course of each cytotoxicity assay.

Page 152: PhD Fusarium

129

Relative differences in cellular growth were established by subtracting mean OD620

values of pPICZB (empty vector) wells from corresponding values for VHH-

transformants. ΔAUC values for each toxophore treatment were calculated based on

addition of differences in cellular growth between VHH and control transformants at each

25 min time interval across the full time course of each assay.

5.3.7 Quantitative Western Blot Analysis

20-mL cultures were centrifuged (1500g, 5 min, 4°C) 48 hours after methanol

induction. Culture pellets were lysed and prepared for SDS-PAGE as described above.

VHH lysate samples (500 µL) were diluted 1:5; 1:25; and 1:50 in non-reducing SDS

sample buffer (BioRad, Mississauga, ON, Canada). pPICZB vector only (control)

samples were not diluted. A reference VHH rAb fragment of a precisely-defined

concentration, and of similar size to NAT-267 and B-24 VHH with C-terminal HA and 6-

His epitope tags, was used to establish a standard dilution series at final protein

concentrations of: 300.0; 150; 75; 37.5; and 18.75 ng·lane-1

. To decrease the bias of

Western blot band intensities, VHH standard dilution series was prepared in P. pastoris

cell lysate from pPICZB empty-vector sample.

Samples were loaded onto 12.5% SDS-PAGE gels and electrophoresed followed by

transfer to a PVDF membrane (Millipore, Billerica, MA, USA) followed by a 1-h

blocking with 3% non-fat skimmed milk in PBST. Mouse anti-6His IgG primary

antibody (GE Healthcare, Piscataway, NJ, USA) diluted 1:2,500 in 3% milk was added to

membranes for 1 h followed by 1 h of blocking and 3 consecutive washes with PBST.

Alkaline phosphatase labeled goat-anti-mouse IgG conjugate (Cedarlane Laboratories,

Page 153: PhD Fusarium

130

Burlington, ON, Canada), diluted 1:3000 in blocking buffer, was used to probe the

membrane for 1 h followed by 3 consecutive washes with PBST. VHH proteins were

visualized using an alkaline phosphatase conjugate substrate kit (Bio-Rad Laboratories,

Hercules, CA, USA).

Quantitative Western blots were dried and photographed with an AlphaImager 3400

(Alpha Innotech Corporation, San Leandro, CA). Analysis was performed using the

AlphaEaseFc software package (Version 7.0.1, Alpha Innotech Corp.). Densometric

(Denso) values for a standard five data-point VHH dilution series (18.75 - 300 ng VHH ·

lane-1

) were used to produce regression equations. Denso values measured for 1:5; 1:25;

and 1:50 dilutions of NAT-267 or B-24 VHH samples were expressed as: „VHH mass per

µL of P. pastoris cell pellet (ng·µL-1

), based on: VHH quantity (ng) (from standard

regression equation) divided by cell pellet lysis volume loaded into each well (µL).

Cellular densities for each cytotoxicity assay were determined by multiplication of OD600

of 1.0 mL culture by 5 x 107 cells per mL culture (constant). VHH concentration (fg·cell

-

1) was calculated as VHH mass per µL cell pellet (ng·µL

-1) (above) divided by cell density

(number of cells·µL-1

culture). VHH intrabody concentration expressed as attoMole

VHH per cell (aMol·cell-1

) was calculated based on: VHH mass per cell (fg·cell-1

) divided

by molecular weight of NAT-267 or B-24 VHH proteins (16637 and 16976 Da,

respectively). Finally, intracellular VHH molarity (µMoles VHH ·L-1

) was calculated as

VHH concentration (aMol·cell-1

) divided by cell volume. P. pastoris cell volume was

estimated to be 29 femtoliters per cell (fL·cell-1

) based on average of independent

measurements of generic yeast-cell volume: 42, 37, 70, and 83 fL (Jorgensen et al., 2002;

Page 154: PhD Fusarium

131

Tyson et al., 1979; Sherman, 2000; and Tamaki et al., 2005; respectively) multiplied by

0.5 to account for an estimate that the cytosol represents 50% of total yeast cell volume

(Biswas et al., 2003).

Mean and standard error values of intracellular VHH expression values were

calculated based on five independent quantitative Western blot assays. Additional details

are summarized in supplemental information (Table 5.S1).

5.3.8 VHH Immunolocalization

Cultures of P. pastoris cells expressing either NAT-267 VHH or pPICZB (empty

vector) were induced with methanol (as described). Cells (20 OD600 units) were pelleted

and washed three times with PBS and resuspended in 500 µL PBS. Fifty-µL aliquots of

P. pastoris cells were incubated in 500 µL fixative solution (1:1 acetone:methanol) for 20

minutes at -20°C. Cells were pelleted (1000g x 1 min), washed three times with PBS and

resuspended in 500 µL PBS supplemented with Lyticase (5 U·µL-1

). After incubation

(30°C, 15 min.), cells were pelleted (as described) and incubated in 500 µL

permeabilization solution (3% BSA (bovine serum albumin), 0.5% Triton X-100 in PBS)

for 30 minutes at room temp. After washing with incubation solution (1% BSA, 0.5%

Tween 20), cells were probed with a 1:100 dilution of monoclonal anti-HA-FITC

antibody (Sigma-Aldrich, Oakville, ON, Canada) for 1-2 h at room temp. Cells were

mounted in PBS supplemented with DAPI (4′,6-diamidino-2-phenylindole dihydro-

chloride) stain (Sigma-Aldrich, Oakville, ON, Canada) at a final concentration of 1.3

µg·ml-1

. Cells were observed after 5 minutes using a Carl Zeiss LSM 510 Meta confocal

microscope equipped with Diode 405 nm and Argon/2 line 488 nm lasers for excitation.

Page 155: PhD Fusarium

132

Emission images were taken 420-480 nm and 505-565 nm for DAPI and FITC,

respectively.

5.3.9 Mycotoxin Biotransformation Assays

After 48 hours of induction (see above), cultures were diluted to an OD600 of 0.25 in

YPD and incubated for 4 h (30°C, 300 rpm). Cultures were diluted to OD600 0.1 in YPD,

and 2.5-mL aliquots of each culture were transferred to 50-mL tubes. Highly purified

DON was added to respective samples to a final concentration of 100 µg·mL-1

followed

by incubation (30°C, 300 rpm). Culture samples (0.5 mL) were taken at intervals of 30

min., 16 and 24 h after mycotoxin addition and used to measure OD600 and culture pH.

Samples were pelleted (3500g, 5 minutes). Supernatant was removed and transferred to

2.0-mL tubes. Samples were frozen in liquid nitrogen and stored at -80°C. Lysate

samples were prepared from cell pellets (as described above). All samples were split to

enable concurrent anti-HA Western blot analysis (as described above) and mycotoxin bio-

transformation assays.

Supernatant, cell lysate, and pellet of selected samples were washed with dd H2O

and passed through 0.8 µm filter (Millipore). Filtrates were passed through Chromosep

C18 columns (C18 Sep-Park Cartridge, Waters Corp., Millford, MA), pre-washed with 10

mL of 100 % (w/v) HPLC grade methanol and 10 mL of dd H2O. After washing with dd

water (10 mL), mycotoxin fractions were eluted with 10 ml of 100 % (v/v) HPLC grade

methanol and dried under a stream of N2. Selected culture samples (as described in Table

5.S2) were diluted in 0.5 mL HPLC grade methanol (100%) and 50.0-µL subsamples

were used for HPLC analysis.

Page 156: PhD Fusarium

133

Trichothecenes were separated on a Shimadzu HPLC system equipped with a 150 x

4.6 mm C18 column (5 µm packing size) and a Hichrome UV flow cell. Standards and

samples were eluted at a flow rate of 1.0 mL∙min

-1 with a 20 min linear gradient of 95%

solvent A (degassed dd H2O): 5% solvent B (acetonitrile) to 40% solvent A: 60% solvent

B. The column was washed from 20-25 min by ramping the gradient to 100% solvent B,

holding at 100% B for 5 min and then returning to the starting conditions over 2 min.

Assessment of DON bio-transformation was based on assessment of retention times of

sample peaks relative to trichothecene standard peaks.

GC/MS analysis, based on a standard Canadian Food Inspection Agency approved

method (Schwadorf and Müller, 1991), was used to further characterize and validate

HPLC samples for structural composition of trichothecene analytes. Biotransformation

samples from HPLC analysis were derivatized with 1.5 mg·mL-1

dimethylaminopyridine

(DMAP) in toluene:acetonitrile (95:5) solution and trifluoroacetic anhydride (TFAA).

This solution was heated at 60°C for 30 minutes, and neutralized twice with 5% KH2PO4.

Derivatized samples were analyzed on a Saturn 2000 GC/Ion-trap mass spectrometer

equipped for chemical ionization using acetonitrile.

1H NMR spectra of pooled P. pastoris lysate samples and subsequent HPLC

fractions of those samples were obtained on a Bruker AM 500 NMR spectrometer in

CDCl3. Chemical shifts are referenced to residual CHCl3 at 7.24 ppm for 1H spectra and

reported (δ) relative to TMS.

Page 157: PhD Fusarium

134

5.4 Results

5.4.1 Sensitivity to Ribotoxin Treatments

The structure, composition, and molecular weight of the mycotoxins used in these

experiments are shown in Figure 5.1. The sensitivity of wild-type P. pastoris to

trichothecene and cycloheximide (control) ribotoxin treatments, was governed by dose

and chemical structure tested. Treatments based on 15-AcDON resulted in the most

immediate and largest overall reduction of cellular growth (Table 5.1).

Table 5.1 Sensitivity of P. pastoris (KM71H) pPICZB transformants to various

ribotoxin treatments tested. Standard deviations (σ) for each mean value

shown in brackets.

Dose

(µg∙mL-1

)

Time to doubling of initial OD620 cellular growth (minutes)

15-AcDON DON 3-AcDON Cycloheximide L.S.D. ††

0† 205 (2.2) 221 (14.4) 219 (21.7) 221 (14.4) 14.5

20 322 (4.4) n.d. n.d. n.d. -

30 384 (4.4) n.d. n.d. n.d. -

40 439 (6.6) n.d. n.d. n.d. -

50 494 (8.8) 308 (7.2) 190 (124) 283 (7.2) 71.7

100 642 (138) 417 (47.3) 194 (119) 310 (20.1) 99.3

200 996 (312) 590 (58.1) 217 (13.0) 341 (7.2) 166.6

L.S.D. 117.2 40.2 91.3 14.0

n.d. not determined

† DMSO, ribotoxin-free media.

††

Least Significant Difference (p=0.05)

P. pastoris was comparatively less sensitive to equivalent doses of DON and

cycloheximide. It was not possible to establish a dose-specific sensitivity to 3-AcDON

Page 158: PhD Fusarium

135

relative to toxin-free DMSO media as cellular growth was not inhibited at the highest

concentration (200 µg·mL-1

) (Table 5.1).

5.4.2 Transformation and VHH Intrabody Expression

P. pastoris KM71H cells were successfully transformed with linearized NAT-267

(treatment) or B-24 (control) VHH DNA ligated into expression vector pPICZB (Figure

5.2). Lysate fractions of methanol-induced transformants were assessed by anti-HA

epitope Western blot analysis and robust intracellular VHH expression was confirmed by

the presence of a 17 kDa band at 48 and 72 hours post methanol-induction (data not

shown).

5.4.3 Quantitative Western Blot Assay

Mean levels of soluble intracellular VHH expression (t = 0h) transformants tested

within cytotoxicity assays were determined by quantitative western blot analysis (Figure

5.3). The initial concentration of NAT-267 VHH in P. pastoris cells within cytotoxicity

assays of 15-AcDON (Figure 5.4) was 4.01 ± 0.31 aMol VHH·cell-1

(Table 5.2).

Figure 5.3 Quantitative Western blot analysis of VHH intrabody expression within

cytotoxicity assays. Blot 1 is VHH standard dilution series from 300.0 to

18.75 ng·well-1

. Blot 2 is NAT-267 VHH expression for cytotoxicity assay

#1 (Figure 5.4). Blot 3 is NAT-267 VHH expression for cytotoxicity assay

#2 (Figure 5.5). Blot 4 is B-24 (control) VHH expression for cytotoxicity

assay #3 (Figure 5.6). BioRad Molecular weight marker (MW) indicated

on the left. Protein dilutions are shown along bottom of blots 2 to 4.

VHH Standard (ng)

300 150

75

37.5

18.8

MW

15 kDa

20 kDa

NAT-267

Assay #1

1/ 5 1/ 25 1/50

NAT-267

Assay #2

1/ 5 1/ 25 1/50

B-24

Assay #3

1/ 5 1/ 25 1/50

Page 159: PhD Fusarium

136

Accounting for an estimated mean P. pastoris cytosol volume of 29 fL·cell-1

(as

previously described), initial expression of NAT-267 VHH was equivalent to an

intracellular concentration of 138 ± 10.8 µMoles VHH·L-1

. Likewise, mean NAT-267

intrabody concentration within cells used in cytotoxicity assays of DON, 3-AcDON, and

cycloheximide (Figure 5.5) was 3.18 ± 0.42 aMol·cell-1

or 110 ± 14.4 µMol·L-1

(Table

5.2). The average initial concentration of B-24 control VHH intrabody in cytotoxicity

assays of 15-AcDON and cycloheximide (Figure 5.6) was 1.93 ± 0.14 aMol·cell-1

or 66.6

± 4.3 µMol·L-1

(Table 5.2).

Table 5.2 Mean VHH intrabody concentration (t = 0 h) within P. pastoris transformants

used in cytotoxicity assays (Figures: 5.4, 5.5, 5.6). Values derived from data

generated from quantitative Western blot assays (Figure 5.3) as summarized

in Table 5.S1. VHH concentration values expressed in femtogram and

attoMol VHH per cell, as well as µMol·L-1

VHH equivalent within P.

pastoris cytosol. Standard Error for all means (n = 9) is shown in

parenthesis.

VHH

Gene

Cytotoxicity

Assay

VHH Intrabody Concentration

fg·cell-1

aMol·cell-1

µmol·L-1

NAT-267 15-AcDON

(Figure 5.4) 66.8 (± 5.2) 4.01 (± 0.31) 138 (± 10.8)

NAT-267

DON,

3-AcDON

Cycloheximide

(Figure 5.5)

53.0 (± 7.0) 3.18 (± 0.42) 110 (± 14.4)

B-24 15-AcDON

Cycloheximide

(Figure 5.6)

32.8 (± 2.3) 1.93 (± 0.14) 66.6 (± 4.3)

Page 160: PhD Fusarium

137

5.4.4 Cytotoxicity Assays

Intrabody VHH expression did not influence the cellular growth of transformed P.

pastoris cultures in mycotoxin-free DMSO media, as OD620 values and overall growth

rate of NAT-267 and B-24 VHH transformants were similar to pPICZB empty-vector

control cells (Figures: 5.4, 5.5, 5.6).

5.4.4.1 15-AcDON

Significant differences (p = 0.01) in cellular growth were observed between NAT-

267 VHH and pPICZB control transformants grown in media spiked with 20-100 µg·mL-1

15-AcDON (Figure 5.4). The impact of NAT-267 VHH intrabody expression on cellular

growth was dependent on the concentration of 15-AcDON used. At the lowest dose (20

µg·mL-1

), growth of NAT-267 VHH expressing cells was similar to cell lines grown in

DMSO (control) media.

Time to significantly improved (p=0.01) growth of NAT-267 VHH intrabody

transformants was dose-dependent. Significant (p=0.01) differences in OD620 values

were observed after 7.5 hours of growth for cells cultured in medium supplemented with

15-AcDON at both 20 and 50 µg·mL-1

(Figure 5.4). Assays spiked with 30, 40, and 100

µg·mL-1

15-AcDON had significant differences in OD620 values after 4.6, 2.9, and 11.3

hours of growth, respectively. No differences in OD620 values were observed between

NAT-267 VHH and control transformants at 200 µg·mL-1

15-AcDON (Figure 5.4).

Differential area under curve (ΔAUC) analysis was used to quantify relative

differences in cellular growth between NAT-267 and B-24 (control) VHH transformants.

Page 161: PhD Fusarium

138

Figure 5.4 Cytotoxicity assays showing the effect of various concentrations (20 to 200 µg·mL

-

1) of 15-AcDON on the growth of P. pastoris pPICZB and NAT-267 VHH

transformants as measured by optical density at 620 nm (OD620). Differential Area

Under Curve (ΔAUC) values for NAT-267 VHH minus pPICZB empty vector are

shown on each graph. At all concentration of 15-AcDON (except 200 µg·mL-1

)

where blue versus red symbols are used, there was a significant difference (p=0.01)

at each time between NAT-267 and pPICZB transformants.

NAT-267, DMSO

pPICZB, DMSO pPICZB, 15-AcDON

NAT-267, 15-AcDON

pPICZB, 15-AcDON (p=0.01)

NAT-267, 15-AcDON (p=0.01)

0.000

0.200

0.400

0.600

0.800

1.000

1.200

0 2 3 5 7 8 10 12 13 15 17 18 20 22 23

0.000

0.200

0.400

0.600

0.800

1.000

1.200

0 2 3 5 7 8 10 12 13 15 17 18 20 22 23

20 µg·mL-1

30 µg·mL-1

OD620 OD620

0.000

0.200

0.400

0.600

0.800

1.000

1.200

0 2 3 5 7 8 10 12 13 15 17 18 20 22 23

40 µg·mL-1

0.000

0.200

0.400

0.600

0.800

1.000

1.200

0 2 3 5 7 8 10 12 13 15 17 18 20 22 23

0.000

0.200

0.400

0.600

0.800

1.000

1.200

0 2 3 5 7 8 10 12 13 15 17 18 20 22 23

100 µg·mL-1

200 µg·mL-1

Time (hours) Time (hours)

ΔAUC = 4.48

ΔAUC = 5.25

ΔAUC = 4.50

ΔAUC = 1.20

ΔAUC = 0.00

0.000

0.200

0.400

0.600

0.800

1.000

1.200

0 2 3 5 7 8 10 12 13 15 17 18 20 22 23

50 µg·mL-1

ΔAUC = 4.45

Page 162: PhD Fusarium

139

The largest ΔAUC value (5.25) corresponded to NAT-267 VHH expressing P. pastoris

grown with 15-AcDON at 30 µg·mL-1

. Cytotoxicity assays based on 15-AcDON at 20,

40, and 50 µg·mL-1

had similar ΔAUC values (4.48; 4.50; and 4.45, respectively) (Figure

5.4). Accordingly, assays at 100 µg·mL-1

produced a substantially lower ΔAUC value

(1.20) while NAT-267 VHH intrabody expression conferred no immunomodulation when

spiked with 200 µg·mL-1

15-AcDON (Figure 5.4).

5.4.4.2 DON, 3-AcDON, Cycloheximide

NAT-267 VHH intrabody expression resulted in a dose-dependent response to DON.

Although no beneficial impact on cellular growth was observed in cultures supplemented

with 50 µg·mL-1

DON, NAT-267 VHH resulted in significantly-improved cellular growth

at 100 and 200 µg·mL-1

DON relative to pPICZB control transformants. Comparative

differences in growth were greater for yeast spiked with 100 relative to 200 µg·mL-1

DON (ΔAUC values = 2.11 and 1.68, respectively). In addition, time to significant

differences in OD620 values was several hours later for the higher dose (Figure 5.5A).

Differences in cellular growth between NAT-267 VHH and pPICZB control P.

pastoris cells could not be established for cytotoxicity assays spiked with 3-AcDON

because this trichothecene had little or no adverse effect on P. pastoris growth (Table

5.1). Assays based on cycloheximide (control ribotoxin) exhibited a dose-dependent

effect on cellular growth; however, no differences were observed between P. pastoris

NAT-267 VHH and pPICZB transformants spiked with doses of 50, 100, and 200 µg·mL-1

(Figure 5.5B).

Page 163: PhD Fusarium

140

Figure 5.5 Cytotoxicity assays showing the effect of various concentrations (50 to 200

µg·mL-1

) of DON (A) and cycloheximide (B) on the growth of P. pastoris

pPICZB and NAT-267 VHH transformants as measured by optical density at

620 nm (OD620). Differential Area Under Curve (ΔAUC) values for NAT-

267 VHH minus pPICZB empty vector are shown for each concentration of

DON. ΔAUC values for cycloheximide treatments were nil. At all

concentrations of DON where green versus red symbols are used, there was

a significant difference (p=0.01) at each time between the NAT-267 and

pPICZB transformants; this was not the case for all cycloheximide

treatments.

A

0.000

0.200

0.400

0.600

0.800

1.000

1.200

0 2 3 5 7 8 10 12 13 15 17 18 20 22 23

Time (hours)

OD620

ΔAUC = 1.20

ΔAUC = 2.11

ΔAUC = 1.68

Cycloheximide

Time (hours)

OD620

pPICZB, DMSO

NAT-267, DMSO

.

pPICZB, Cycloheximide

+ NAT-267, Cycloheximide

pPICZB, DMSO

NAT-267, DMSO

NAT-267, DON

pPICZB, DON

pPICZB, DON (p=0.01)

NAT-267, DON (p=0.01)

200 µg·mL-1

100 µg·mL-1

50 µg·mL-1

200 µg·mL-1

100 µg·mL-1

50 µg·mL-1

0.000

0.200

0.400

0.600

0.800

1.000

1.200

0 2 3 5 7 8 10 12 13 15 17 18 20 22 23

B

Deoxynivalenol

Page 164: PhD Fusarium

141

5.4.4.3 B-24 VHH Control

Expression of B-24 control VHH intrabody had no effect on cellular growth rate

relative to pPICZB control transformants in DMSO or in media supplemented with either

15-AcDON or Cycloheximide (Figure 5.6). These results suggest that trichothecene-

specific immunomodulation imparted by NAT-267 VHH intrabody expression was

trichothecene-specific, as Pichia cells expressing a non-specific VHH intrabody were

equally as sensitive as pPICZB control transformants.

5.4.5 VHH Immunolocalization

NAT-267 VHH intrabody expression and distribution within P. pastoris

transformants was further validated with fluorescent confocal microscopy (Figure 5.7).

Detection of the HA-tag on NAT-267 VHH intrabody fragments within transformants

grown in YPD media to OD600 = 20 further confirmed robust intrabody expression within

our test system.

5.4.6 Mycotoxin Biotransformation Assays

Given that NAT-267 VHH was confirmed previously to have binding affinity

limited to 15-AcDON (Doyle et al., 2008), we were surprised to observe a dose-

dependent amelioration to DON cytotoxicity within NAT-267 VHH transformants (Figure

5.5A). To validate this observation and re-confirm in vivo antigen specificity of NAT-

267 VHH intrabody expression, we assessed the potential biotransformation of DON

within our P. pastoris test system by spiking with DON. HPLC analysis of 16-h and 24-h

cultures grown in DON-supplemented YPD media consistently yielded another peak in

addition to the DON peak at ~ 5.85 min (Table 5.S2). This peak appeared at ~

Page 165: PhD Fusarium

142

Figure 5.6 Cytotoxicity assays showing the effect of various concentrations (50 to 200

µg·mL-1

) of 15-AcDON (A) and cycloheximide (B) on the growth of P.

pastoris pPICZB and non-specific B-24 VHH transformants as measured by

optical density at 620 nm (OD620). Differential Area Under Curve (ΔAUC)

values for B-24 VHH minus pPICZB empty vector for both15-AcDON and

cycloheximide are nil. At all concentrations of DON and cycloheximide

there was no significant difference (p=0.01) at each time between the B-24

and pPICZB transformants.

OD620

100 µg·mL-1

200 µg·mL-1

pPICZB, DMSO

B-24, DMSO

. pPICZB, Cycloheximide

+ B-24, Cycloheximide

0.000

0.200

0.400

0.600

0.800

1.000

1 3 5 6 8 10 11 13 15 16 18 20 21 23

Cycloheximide

Time (hours)

200 µg·mL-1

50 µg·mL-1

100 µg·mL-1

OD620

0.000

0.200

0.400

0.600

0.800

1.000

1 3 5 6 8 10 11 13 15 16 18 20 21 23

15-AcDON

B-24, 15-AcDON

pPICZB, 15-AcDON

pPICZB, DMSO

B-24, DMSO

Time (hours)

A

B

Page 166: PhD Fusarium

143

Figure 5.7 Representative confocal microscopy photographs of P. pastoris (KM71H)

cells isolated, washed and immuno-probed with anti-HA epitope mAb-FITC

conjugate and DAPI stain after growing in YPD media to OD600 = 20. A =

Control, pPICZB empty vector (control). B = P. pastoris transformants

expressing NAT-267 VHH intrabody shown by fluorescent green cells.

9.5 min and matched the retention time of 15-AcDON (Table 5.S2). HPLC

chromatograms of cell lysate and supernatant samples of pPICZB and NAT-267 VHH

transformants provided clear evidence of a metabolic conversion of DON to 15-AcDON.

No trichothecenes were found within cell pellet fractions (Table 5.S2). HPLC analysis

revealed no evidence of DON-glucosyl metabolites or any structurally-similar compounds

within samples tested. GC/MS analysis confirmed the presence of 15-AcDON within

pooled cell lysate samples (data not shown). The GC retention time and mass spectrum

of the peak confirmed the presence of 15-AcDON and absence of 3-AcDON within

samples tested (data not shown). Finally, the 1H NMR spectra of the total filtrate extract

Page 167: PhD Fusarium

144

was dominated by DON and other impurities from the medium. However, the presence

of 15-AcDON was confirmed in the NMR spectrum of the HPLC fraction isolated at ~9.5

min from the pooled lysate samples (Figure 5.8).

Figure 5.8 Representative 500 mHz 1H NMR spectrum of A = 15-AcDON

Standard. B = HPLC fraction of P. pastoris cell lysate sample (24 hrs).

Unique chemical shifts of 15-AcDON are labeled due to H-11 (4.87

ppm), H-7 (4.81 ppm), and H-15 (4.21 ppm) protons which confirm the

presence of 15-AcDON within cell lysate.

Page 168: PhD Fusarium

145

5.5 Discussion

Various strategies have been used to develop plants with enhanced resistance to

trichothecene cytotoxicity and associated Fusarium pathogenesis. These approaches tend

to be based on mechanisms which alter host cellular targets (Harris and Gleddie, 2001;

Mitterbauer et al., 2004), reduce mycotoxin cytotoxicity (Alexander, 2008; Poppenberger

et al., 2003), or enhance innate host resistance through crop breeding techniques

(Snijders, 2004). Our goal was to evaluate the ability of a mycotoxin-specific VHH

intrabody to immuno-modulate, or attenuate, the cytotoxic effects of 15-AcDON using a

yeast model.

Aside from application as a bioassay-indicator organism (Abolmaali et al., 2008;

Binder, 1999), several species of yeast have been used to evaluate the in situ function of

various trichothecene-specific genes (Alexander et al., 1999; Poppenberger et al., 2003)

or assess efficacy of mycotoxin-specific transgenes prior to in planta application

(Mitterbauer and Adam, 2002; Mitterbauer et al., 2004; Poppenberger et al., 2003). The

methylotropic yeast P. pastoris (strain KM71H) was selected as our eukaryotic model

based on expected sensitivity to target ribotoxins and known capacity to express high

levels of functional heterologous proteins (Cereghino and Cregg, 2000). We

demonstrated that it was possible to transform, assess, and validate P. pastoris cells

within a matter of weeks, thus creating an ideal platform for screening mycotoxin-specific

constructs before in planta evaluation.

Trichothecene cytotoxicity is determined by the C-12,13 epoxide group common to

this class of mycotoxins (Carter and Cannon, 1977; Rocha et al., 2005; Rotter et al.,

Page 169: PhD Fusarium

146

1996). However, the number and position of hydroxyl and acetyl-ester groups on the

trichothecene structure can also influence the mechanism of protein synthesis inhibition

and relative toxicities within eukaryotic cells (Betina, 1989; Desjardins et al., 2007;

Ehrlich and Daigle, 1987). In this regard, we found that the position of an acetyl-ester

group on either carbon 3 or 15 of DON (Figure 5.1) had a profound effect on their

cytotoxicity to P. pastoris (Table 5.1). Our findings align with results established in other

yeast species where 15-AcDON was significantly more toxic than DON (Mitterbauer et

al., 2004; Poppenberger et al., 2003), which was more toxic than 3-AcDON (Binder et al.,

1997). These results also support findings of structure-activity studies established in

planta that demonstrated 15-AcDON is more toxic than DON (Desjardins et al., 2007;

Poppenberger et al., 2003) and 3-AcDON (Alexander, 2008; Desjardins, 2006). Our

observation of no measurable cytotoxicity of 3-AcDON is in agreement with previous

studies which demonstrated that addition of an acetyl group at C-3 (Figure 5.1) serves to

eliminate cytotoxicity as part of a metabolic defence mechanism during trichothecene

synthesis (Kimura et al., 1998a; Kimura et al., 2006).

Our results also quantify relative differences in trichothecene cytotoxicity over time,

not at a specific assay endpoint. Our P. pastoris model system enables real-time

evaluation of cellular effects during the initial phases of ribotoxin exposure. We propose

that this biological system is very sensitive (i.e. compared to whole plant systems) and

shows clear effects on cellular growth within the first hours of exposure thereby

indicating the potency of trichothecene-mediated cytotoxicity.

Page 170: PhD Fusarium

147

A sequential two-step process was required before commencement of each

cytotoxicity assay. First, transformants were induced with methanol for 48h. After

induction of VHH intrabody expression, P. pastoris cells were transferred to YPD media

for a 4-h „recovery period‟ to re-stimulate cellular growth. Mycotoxin or cycloheximide

(control) treatments were then added (at t = 0h) to initiate each cytotoxicity assay. Given

the well-established negative impact on eukaryotic protein synthesis, it was an obvious

necessity to induce VHH expression prior to addition of ribotoxin treatments. The 4-h

recovery in YPD media was required because induction conditions did not support a

optimal cellular division and growth over time. We also noted that, by virtue of its

design, our test system ensured a fixed ratio of VHH antibody to ribotoxin hapten

throughout the course of each cytotoxicity assay. We recognized the potential

disadvantage of culturing induced cells in YPD as leading to a continuous dilution of

VHH concentration within the cytosol of rapidly dividing cells. However, we sought to

maintain a fixed ratio of Ab:Ag within each microtiter well to ensure unbiased

measurements of overall cellular growth.

Soluble NAT-267 and B-24 control VHH intrabody fragments were well expressed

within P. pastoris. Average protein expression levels (Table 5.2) were found to be much

higher than previously reported in P. pastoris (Gurkan et al., 2004; Omidfar et al., 2007;

Rahbarizadeh et al., 2006). Furthermore, images generated by confocal immuno-

microscopy confirmed that NAT-267 VHH fragments were well distributed within the

cytosol of P. pastoris transformants even after growth to a very high OD600 (Figure 5.7).

Page 171: PhD Fusarium

148

Our principal hypothesis was that the expression of mycotoxin-specific VHH

intrabody fragments would effectively sequester (i.e., neutralize) bio-available 15-

AcDON concentrations within the cell and thereby limit cellular toxicity, thereby

resulting in enhanced growth of P. pastoris. VHH-mediated mycotoxin binding was

assessed by time-to-significant immunomodulation and ΔAUC values between VHH

transformants and pPICZB (control) cell lines for each ribotoxin treatment tested (Figures

5.4 – 5.6). The initial doses of 15-AcDON tested are generally equivalent to those found

during Fusarium infection of agricultural crops. For example, intracellular levels of

DON have been reported at concentrations ranging from 13.3 to 88.7 µg·g-1

within F.

graminearum inoculated cereal tissue (Del Ponte et al., 2007; Mudge et al., 2006).

Attenuation of 15-AcDON activity clearly demonstrates that NAT-267 VHH intrabody

expression conveys significant (p = 0.01) resistance to mycotoxin-specific cytotoxicity

(Figure 5.4) by regulating the availability of free and VHH-bound 15-AcDON.

Consequently, results were dose-dependent as time to immumodulation was optimal for

30 and 40 µg·mL-1

15-AcDON, when compared to the lower (20 µg·mL-1

) and higher

concentrations (i.e., 50 and 100 µg·mL-1

) of the toxophore. Sequestration of 15-AcDON

was substantially reduced and delayed at concentrations of 100 µg·mL-1

while at 200

µg·mL-1

there was no benefit associated with VHH expression (Figure 5.4).

NAT-267 VHH intrabody immuno-modulation was trichothecene-specific as no

attenuation of 15-AcDON toxicity was observed in assays supplemented with control

ribotoxin (cycloheximide) (Figure 5.5). It was not possible to assess the effect of NAT-

267 VHH on the cytotoxicity of 3-AcDON because all doses tested were not toxic to P.

Page 172: PhD Fusarium

149

pastoris cells (Table 5.1). Further confirmation of NAT-267 VHH specificity was shown

with cytotoxicity assays using P. pastoris transformants expressing a control VHH

intrabody, i.e., B-24. Furthermore, there was no immunomodulation of either 15-

AcDON- or cycloheximide-specific cytotoxicity when pPICZB control cells were used

(Figure 5.6).

The attenuation of DON cytotoxicity at 100 and 200 µg·mL-1

(Figure 5.5) was an

unexpected result since we previously-reported that NAT-267 VHH has no affinity for

DON (Doyle et al., 2008). In vivo sequestration of DON was explained by confirmation

of bio-transformation of DON to 15-AcDON in P. pastoris cell lysate and culture

supernatant samples of NAT-267 and pPICZB transformants (Table 5.S2; Figure 5.8).

Data mining of the genome of NRRL Y-11430 P. pastoris (the parent strain of KM71H)

by Integrated Genomics Inc. (Chicago, IL, USA) revealed no similarities or related

homologs to a previously characterized fungal gene responsible for acetylation of DON to

15-AcDON (McCormick et al., 1996). We therefore attributed this biochemical

conversion to a previously-uncharacterized acetyl-transferase gene within the P. pastoris

genome.

To predict the impact of NAT-267 VHH intrabody binding to reduce effective 15-

AcDON toxin concentrations within the cytosol, we adopted a previously described

(Almquist et al., 2004) mass-balance model based on the following equation.

[Bound Toxin] =

[VHH] [Total Toxin] x

KD [VHH] + ( )

Page 173: PhD Fusarium

150

We assumed linear, dose-independent binding of VHH to 15-AcDON within an

aqueous environment where 1 g = 1 mL. The dissociation constant (KD) of NAT-267

VHH was taken as1.24 µM (Doyle et al., 2008) with 1:1 stoichiometry of Ab:Ag binding.

NAT-267 ([VHH]) concentration within the P. pastoris cytosol was estimated to be 138

µmol·L-1

(Tables 5.2, 5.S1). We assumed optimal disulfide bond formation and post-

translational VHH folding within the endoplasmic reticulum and cytosol (Ellgaard, 2004)

with no NAT-267 VHH intrabody leakage from the P. pastoris cell membrane. The

concentration of 15-AcDON within the cytosol was taken as equivalent to culture media,

and we also assumed that there were no trichothecene targets outside of the cells.

Based on total 15-AcDON concentration of 50 µg·mL-1

(or 148 µM) (Figure 5.4), this

model predicts (138 µM x 148 µM) ÷ (138 µM + 1.24 µM) = 147 µM of 15-AcDON (or

> 99.9%) toxin was bound and ~ 1.3µM (or < 0.1%) of 15-AcDON was free. This simple

model indicates that 15-AcDON cytotoxicity at 50 µg·mL-1

should be completely

eliminated by expression of NAT-267 VHH; however, although the effects of 15-AcDON

were significantly ameliorated by VHH expression at this concentration, they were not

completely eliminated (Figure 5.4).

A more complete assessment of NAT-267 VHH immunomodulation should also

account for in situ competition effects and binding affinity between 15-AcDON and its

major cellular target, i.e., ribosome binding. Binding affinity is also an important

consideration because NAT-267 VHH has a relatively low affinity (KD = 1.24 µM) for 15-

AcDON that is mediated by weak, non-covalent interactions (e.g., van der Waals forces,

hydrogen bonding, etc.) (Maynard and Georgiou, 2000). Due to the rapid dissociation rate

Page 174: PhD Fusarium

151

constant for NAT-267, we hypothesize that 15-AcDON is subject to continuous turnover

in terms of mycotoxin binding to the VHH. In other words, the efficiency of NAT-267

VHH in terms of limiting the cytotoxicity of 15-AcDON is limited by the short “residence

time” of hapten binding to the VHH (Copeland et al., 2006). Further evidence for this

hypothesis resides in the fact that even when the VHH intrabody is expressed in excess

(138 µmol·L-1

, Table 5.2) compared to 15-AcDON, i.e. at 89 μM (30 µg mL-1

), NAT-267

cannot fully immunomodulate the cytotoxic effects of 15-AcDON when compared to

controls (Figure 5.4). Thus, at higher doses ≥30 µg·mL-1

of 15-AcDON, we postulate

that the VHH simply cannot compete with a stronger and quite possibly longer residence

time of 15-AcDON on the 60S ribosomal protein subunit L3 (Rpl3) of P. pastoris.

We also assert that, at very high doses, 15-AcDON may cause other potentially

irreversible effects such as membrane disruption, inhibited RNA and DNA synthesis, and

various other apoptotic effects (reviewed in Rocha et al., 2005; Rotter et al., 1996; Ueno,

1984) which may also severely limit cellular growth. Thus, to accurately determine the

efficacy of NAT-267 VHH intrabody in P. pastoris cells one must account for the

dynamic nature of mycotoxin-mediated cytotoxicity and in vivo binding kinetics of 15-

AcDON for other cellular binding targets. Our assertion that attenuation of

trichothecene-specific cytotoxicity is not a simple process is in agreement with previous

work which demonstrated only partial remediation of DON cytotoxicity based on in

planta expression of an altered Rpl3 target protein (Harris and Gleddie, 2001;

Mitterbauer et al., 2004) or the expression of Tri101 (Alexander, 2008, Kimura et al.,

1998a).

Page 175: PhD Fusarium

152

Most phytopathogenic Fusarium species are believed to produce 15-AcDON or 3-

AcDON as acetylated precursors to DON (Mitterbauer and Adam, 2002). Production of

each acetylated compound has been described as geography-dependent; 3-AcDON

chemotypes dominate in Asia and Europe, while Fusarium species which produce 15-

AcDON are more prevalent in North America (Goswami and Kistler, 2004; Moss and

Thrane, 2004). Therefore, the accumulation of DON within plant tissue is believed to be

a metabolic process conferred by fungal and plant carboxyl-esterases which continuously

deacetylate either 15-AcDON or 3-AcDON within plant tissue (Mitterbauer and Adam,

2002). Confirmation of NAT-267 efficacy observed in this work is very significant since

15-AcDON was the most cytotoxic compound tested. If 15-AcDON is as toxic to plants

as it is to yeast then expressing NAT-267 VHH within the cytosol before trichothecene

accumulation may help limit in vivo pathogenesis and metabolism to DON during

Fusarium infection of plants such as corn and wheat.

Future experiments will focus on the development of an anti-15-AcDON VHH with

an improved dissociation constant (KD) to ensure a stronger association between the VHH

and target ligand for improved in vivo efficacy (Copeland et al., 2006). It would also be

of great interest to develop and test novel VHH fragments with affinity for various other

trichothecenes (e.g., neosolaniol, diacetoxyscripenol, T-2 toxin, etc.) and various other

mycotoxin classes (e.g., fumonisins, aflatoxins, etc.) using this test system. A logical

subsequent application would be constitutive in planta expression of optimized

mycotoxin-specific VHH fragments, possibly with catalytic activity, to bind and

deactivate/degrade mycotoxins during critical initial periods of plant pathogenesis.

Page 176: PhD Fusarium

153

5.6 Acknowledgements

We would like to express our sincere appreciation to Dr. Barbara Blackwell at

Agriculture and Agri-Food Canada Ottawa for assistance, insights, and expertise in

HPLC, GC/MS, and NMR assays to characterize mycotoxin biotransformation samples.

We are also indebted to colleagues at the Canadian Food Inspection Agency for GC/MS

analysis of biotransformation samples. This research was supported by grants to JCH

from the Natural Sciences and Engineering Research Council of Canada, the Canada

Research Chairs Program, and the Ontario Ministry of Agriculture, Food, and Rural

Affairs.

Page 177: PhD Fusarium

154

5.7 Supplemental

EcoRI

NAT-

267

AAGCTTGAATTCGGTACCATGCAAGTCCAATTGGAAGAATCCGGTGGTGGTTTAGTTCAA 60

M__Q__V__Q__L__E__E__S__G__G__G__L__V__Q_

B-24 A__Q__V__K__L__E__E__F__G__G__R__L__V__Q_

AAGCTTGAATTCGGTACCGCCCAGGTAAAGCTGGAGGAGTTTGGGGGAAGATTGGTGCAG 60

EcoRI

NAT-

267

GCCGGTGGTTCCTTGAGATTGTCTTGTGCTGCTTCTGGTAGAACTTCCTCCAACACCTTC 120

_A__G__G__S__L__R__L__S__C__A__A__S__G__R__T__S__S__N__T__F_

B-24 _A__G__G__S__L__R__L__S__C__A__A__S__V__R__T__F__S__N__R__P_

GCTGGGGGCTCTCTGAGACTCTCCTGTGCAGCCTCTGTACGCACCTTCAGTAACAGACCC 120

NAT-

267

GTCGGTTGGTTTAGACAAGCCCCAGGTAAAGAAAGAGAATTTGTCGCCGCCATTAGAAGA 180

_V__G__W__F__R__Q__A__P__G__K__E__R__E__F__V__A__A__I__R__R_

B-24 _M__G__W__F__R__Q__T__P__G__N__E__R__E__F__V__T__A__I__S__S_

ATGGGCTGGTTCCGCCAAACTCCAGGGAATGAGCGTGAGTTTGTAACAGCTATTAGTTCG 180

NAT-

267

TCCGACGACAGAACTTACTACGCTGCCTCTGTCAGAGGAAGATTCACCATTTCCGGTGAT 240

_S__D__D__R__T__Y__Y__A__A__S__V__R__G__R__F__T__I__S__G__D_

B-24 _S__G__A__S__T__R__Y__A__D__S__V__K__G__R__F__T__I__S__R__D_

AGTGGTGCGAGCACACGCTATGCAGACTCCGTGAAGGGCCGATTCACCATCTCCAGAGAC 240

NAT-

267

TCCGCTAAGAACGTCGTTGCTTTGCAAATGTCCTCCTTAAGACCAGAAGATACCGCCGTT 300

_S__A__K__N__V__V__A__L__Q__M__S__S__L__R__P__E__D__T__A__V_

B-24 _N__A__R__N__T__V__Y__L__Q__M__N__S__L__K__P__E__D__T__A__V_

AACGCCAGGAACACGGTGTATCTGCAAATGAACAGCCTGAAACCTGAAGACACGGCCGTT 300

NAT-

267

TACTACTGTGCTGCTACCAGAACTTGGTTGGTTACCGGTCAATCCGATTACCCATACXXX 360

_Y__Y__C__A__A__T__R__T__W__L__V__T__G__Q__S__D__Y__P__Y__X_

B-24 _Y__Y__C__A__A__S__Y__Y__S__G__T__P__T__T__S__S__A__Y__D__Y_

TATTACTGTGCAGCCTCATACTATAGTGGTACTCCCACCACCAGCAGCGCGTATGACTAC 360

NAT-

267

TGGGGTCAAGGTACTCAAGTTACCGTTTCCTCTGGTCAAGCCGGTCAACATCATCACCAT 420

_W__G__Q__G__T__Q__V__T__V__S__S__G__Q__A__G__Q__H__H__H__H_

B-24 _W__G__Q__G__T__Q__V__T__V__S__S__G__Q__A__G__Q__H__H__H__H_ 420

TGGGGCCAGGGGACCCAGGTCACCGTCTCCTCAGGCCAGGCCGGCCAGCACCATCACCAT

NAT-

267

CACCACGTGTCCTCTGGTAGATACCCATACGATGTCCCAGATTACGGTTCTGGTAGAGCC

_H__H__V__S__S__G__R__Y__P__Y__D__V__P__D__Y__G__S__G__R__A_

B-24 _H__H__V__S__S__G__R__Y__P__Y__D__V__P__D__Y__G__S__G__R__A_ 480

CACCATGTCTCCAGCGGCCGCTACCCGTACGACGTTCCGGACTACGGTTCCGGCCGAGCA

XbaI

NAT-

267

TAAGTCGACTCTAGAGCTC 486

_*_

B-24 _*_

TAGGTCGACTCTAGAGCTC 486

XbaI

Figure 5.S1 Homology chart of NAT-267 and B-24 VHH fragments. Framework regions (FR)

are outlined in black while complementarity determining regions (CDR) 1, 2, and 3

are outlined in blue. Cysteine residues at C23 and C97 responsible for disulfide

bond VHH folding are shown in red. XXX denotes extra codon within CDR3 of

NAT-267 gene. Restriction endonuclease sites, epitope tags (6HIS = green, HA =

pink) and stop codons (*) are marked accordingly. NAT-267: Theoretical pI for

NAT-267 and B-24 VHH = 9.00 and 9.17, respectively (ExPASy).

Page 178: PhD Fusarium

155

Table 5.S1: Raw data and calculations of VHH intrabody expression in Pichia pastoris

transformants derived from quantitative Western blot analysis (Figure 6.3) and as

summarized in Table 6.2.

Sample

Dilution

Total VHH

Lysis* (ng)

VHH, 1 mL cell †

(µg·mL-1

)

VHH, 1 mL pellet††

(µg·mL-1

)

Calculated Intracellular VHH Expression

(fg·cell-1

) ††

(aMol·cell-1

) ‡‡

µMol·L-1☺

Cytotoxicity Assay-1: NAT-267 vs. 15-AcDON (Figure 6.4)

Blot 1

1 / 5 435.8 217.9 77.35 71.96 4.325 149.1

1 / 25 71.2 178.0 63.19 58.78 3.533 121.8

1 / 50 n.d. n.d. n.d. n.d. n.d. n.d.

Blot 2

1 / 5 322.2 161.1 57.19 53.20 3.198 110.3

1 / 25 122.5 281.1 99.80 92.84 5.580 192.4

1 / 50 48.40 242.0 85.91 79.92 4.803 165.6

Blot 3

1 / 5 192.3 96.15 34.13 31.75 1.908 65.8

1 / 25 60.90 152.3 54.05 50.28 3.022 104.2

1 / 50 36.60 183.0 64.97 60.43 3.632 125.3

Blot 4

1 / 5 305.30 152.65 54.19 50.41 3.030 104.5

1 / 25 114.11 285.27 101.27 94.20 5.662 195.2

1 / 50 53.84 269.19 95.56 88.90 5.343 184.2

Blot 5

1 / 5 272.60 136.30 48.39 45.01 2.705 93.3

1 / 25 88.57 221.43 78.61 73.12 4.395 151.6

1 / 50 50.67 253.33 89.93 83.66 5.028 173.4

Mean (n=14) 66.75 4.01 138

Standard Error (n=14) 5.23 0.31 10.8

Cytotoxicity Assay-2: NAT-267 vs. DON, 3-AcDON, Cycloheximide (Figure 6.5)

Blot 1

1 / 5 127.9 63.94 22.70 19.48 1.171 40.4

1 / 25 25.11 62.78 22.29 19.13 1.150 39.6

1 / 50 n.d. n.d. n.d. n.d. n.d. n.d.

Blot 2

1 / 5 238.9 119.5 42.42 36.41 2.188 75.5

1 / 25 107.5 268.8 95.41 81.89 4.922 169.7

1 / 50 69.19 346.0 122.8 105.42 6.336 218.5

Blot 3

1 / 5 148.6 74.3 26.38 22.64 1.361 46.9

1 / 25 85.36 213.4 75.75 65.03 3.908 134.8

1 / 50 31.07 155.4 55.15 47.34 2.845 98.1

Blot 4

1 / 5 287.32 143.66 51.00 43.78 2.631 90.7

1 / 25 79.42 198.54 70.48 60.50 3.636 125.4

1 / 50 54.85 274.26 97.36 83.57 5.023 173.2

Blot 5

1 / 5 233.25 116.62 41.40 35.54 2.136 73.7

1 / 25 88.57 221.43 78.61 67.47 4.055 139.8

1 / 50 35.04 175.20 62.20 53.39 3.209 110.7

Mean (n=14) 52.97 3.18 109.8

Standard Deviation (n=14) 6.96 0.42 14.4

Page 179: PhD Fusarium

156

Table 5.S1, Continued: Raw data and calculations of VHH intrabody expression in Pichia

pastoris transformants derived from quantitative Western blot analysis (Figure

6.3) and as summarized in Table 6.2.

Sample

Dilution

Total VHH

Lysis* (ng)

VHH, 1 mL cell †

(µg·mL-1

)

VHH, 1 mL pellet††

(µg·mL-1

)

Calculated Intracellular VHH Expression

(fg·cell-1

) ‡

(aMol·cell-1

) ‡‡

µMol·L-1☺

Cytotoxicity Assay-3: B-24 vs. 15-AcDON and Cycloheximide (Figure 6.6)

Blot 1

1 / 5 137.9 68.95 24.48 26.46 1.559 53.8

1 / 25 n.d. n.d. n.d. n.d. n.d. n.d.

1 / 50 n.d. n.d. n.d. n.d. n.d. n.d.

Blot 2

1 / 5 179.8 89.9 31.91 34.50 2.032 70.1

1 / 25 35.24 88.1 31.28 33.81 1.992 68.7

1 / 50 24.29 121.5 43.11 46.61 2.746 94.7

Blot 3

1 / 5 137.5 68.75 24.41 26.39 1.554 53.6

1 / 25 33.21 83.03 29.47 31.86 1.877 64.7

1 / 50 15.54 77.70 27.58 29.82 1.757 60.6

Blot 4

1 / 5 187.29 93.65 33.24 35.94 2.117 73.0

1 / 25 51.31 128.27 45.53 49.23 2.900 100.0

1 / 50 n.d. n.d. n.d. n.d. n.d. n.d.

Blot 5

1 / 5 129.08 64.54 22.91 24.77 1.459 50.3

1 / 25 28.10 70.24 24.94 26.96 1.588 54.8

1 / 50 13.92 69.59 24.70 26.71 1.573 54.3

Mean (n=12) 32.76 1.93 66.6

Standard Deviation (n=12) 2.30 0.14 4.3

Calculations, Table 5.S1:

* Total VHH Lysis (ng) of diluted VHH samples from each assay derived from regression equation analysis

based on a five point dilution series (300, 150, 75.0, 37.5, 18.75 ng·lane-1

) of a reference VHH fragment

(as described in EXPERIMENTAL PROCEDURES).

x = Calculated mass of VHH in cell lysate (ng).

y = Measured densometric value of unknown sample (densometric units).

Blot 1: x = [(y - 3.292) ÷ 0.3387], R2 = 0.9555

Blot 2: x = [(y + 1.8174) ÷ 0.0913], R2 = 0.9815

Blot 3: x = [(y + 3.700) ÷ 0.5600], R2 = 0.9902

Blot 4: x = [(y + 17.561) ÷ 0.3949], R2 =0.9856

Blot 5: x = [(y + 2.9104) ÷ 0.3456], R2 =0.9975

† VHH from 1 mL Cell Lysis (µg · mL

-1) = Est. VHH Quantity (ng) . x 1,000 (µL·mL

-1)

Total Vol. Cell Pellet Loaded (µL) 1,000 (ng·µg-1

)

†† VHH from 1 mL Cell Pellet (µg · mL

-1) = VHH from 1 mL Cell Lysis (µg) .

Lysis Volume per 1 mL Cell Culture (µL)

Page 180: PhD Fusarium

157

Calculations, Table 5.S1 (Continued):

‡ VHH per cell (fg·cell

-1) = VHH from 1 mL Cell Pellet (µg·mL

-1) . x 1.0 x 10

9 fg

P. pastoris cell density (# cells· 1mL culture-1

) 1.0 µg

‡‡

VHH per cell (aMol·cell-1

) = Intracellular VHH per cell (fg·cell-1

) x 1.0 x 103 aMol

VHH MW (fg·fMol-1

) 1.0 fMol

☺ VHH Concentration within P. pastoris cytosol = VHH per cell (a Mol · cell

-1) ÷ 29 femto L · cell

-1

Constants, Table 5.S1:

- Total Volume of Cell Pellet Loaded for 1 / 5; 1/25 and 1/50 dilutions = 2.0; 0.4 and 0.2 µL, resp.

- Lysis Volume per 1.0 mL Cell Culture = 355 µL

- Mean OD600 of 1.0 mL P. pastoris culture for Cytotoxicity Assay-1 (Figure 5.4) = 21.5

- Mean OD600 of 1.0 mL P. pastoris culture for Cytotoxicity Assay-2 (Figure 5.5) = 23.3

- Mean OD600 of 1.0 mL P. pastoris culture for Cytotoxicity Assay-3 (Figure 5.6) = 18.5

- For Pichia pastoris, 1 OD600 = 5 x 107 cells·mL

-1

- For Pichia pastoris, Volume of one cell = 29 femtoliters (2.9 x 10-14

Liters)

Therefore,

- Mean P. pastoris cell density for Cytotoxicity Assay-1 = (5 x 107 cells · mL

-1) X 21.5 = 1.075 x 10

9

- Mean P. pastoris cell density for Cytotoxicity Assay-2 = (5 x 107 cells · mL

-1) X 23.3 = 1.165 x 10

9

- Mean P. pastoris cell density for Cytotoxicity Assay-3 = (5 x 107 cells · mL

-1) X 18.5 = 9.250 x 10

8

Molecular Weights (www.expasy.ch/)

- NAT-267 (treatment) VHH = 16637.3 Da (fg · fMol-1

)

- B-24 (control) VHH = 16975.6 Da (fg · fMol-1

)

Page 181: PhD Fusarium

158

Table 5.S2: HPLC retention times of trichothecene metabolites evaluated after 24 h of growth

of P. pastoris transformants cultured in YPD media supplemented with 100

µg·mL-1

of highly-purified DON. Transformation of DON (5.750 minutes) to 15-

AcDON (9.625 minutes) within supernatant (S/N), cell lysate (LYS), and cell

pellet debris (DEBS) shown in 30-minute, 16-h and duplicate 24-h samples of

NAT-267 VHH and pPICZB empty vector transformants.

Sample Description Peaks (Retention time, min)

DON 15-AcDON

Supernatant (S/N) Fraction

NAT-267 VHH, S/N, 30min. 5.850 H 9.542 NP

NAT-267 VHH, S/N, 16h 5.825 H 9.517 H

NAT-267 VHH, S/N, 24h, Rep 1 5.833 VH 9.533 VH

NAT-267 VHH, S/N, 24h, Rep 2 5.808 H 9.550 H

pPICZB (empty vector), S/N, 30min. 5.975 H 9.542 L

pPICZB (empty vector), S/N, 16h 5.817 VH 9.525 VH

pPICZB (empty vector), S/N, 24h, Rep 1 5.825 VH 9.542 H

pPICZB (empty vector), S/N, 24h, Rep 2 5.825 VH 9.533 H

Cell Lysate (LYS) Fraction

NAT-267 VHH, LYS, 24h, Rep 1 5.858 M 9.575 M

NAT-267 VHH, LYS, 24h, Rep 2 5.875 H 9.567 M

pPICZB (empty vector), LYS, 24h, Rep 1 5.817 M 9.542 M

pPICZB (empty vector), LYS, 24h, Rep 2 5.817 M 9.525 L

Cell Debris (DEBS) Fraction

NAT-267 VHH, DEBS, 24h 5.550 NP 9.567 NP

pPICZB (empty vector), DEBS, 24h 5.855 NP 9.525 NP

- No peak (NP) observed

- Low (L) = very small or no peak observed.

- Medium (M) = medium or measurable peak observed.

- High (H) = large peak observed.

- Very High (VH) = very large peak observed.

Page 182: PhD Fusarium

159

6 CONCLUSIONS

The research and results summarized within this thesis reaffirms the vast potential

of recombinant antibody engineering as a discipline with wide ranging applications. Our

specific goal was to develop a rAb fragment with binding affinity to DON, a key

mycotoxin produced during FHB pathogenesis. This was a proof of concept to apply a

novel technology platform commonly used in pharmaceutical research toward an untested

discipline, plant pathology. Our hypothesis was based on the notion that expression of an

engineered rAb fragment within a model eukaryotic test system could bind a target

mycotoxin and inherently limit in situ cytotoxicity to provide enhanced tolerance to FHB

pathogenesis. Although the results of this dissertation are well summarized within

Chapters # 4 and # 5, the following provides an overall summary of key conclusions and

outlines future research interests and potential technological applications.

Immunization of a llama with 15-DON-BSA conjugate resulted in a robust

polyclonal response to the hapten complex (i.e., DON, 15-AcDON, 3-AcDON). A hyper-

immunized, phage-display VHH library was successfully created from peripheral blood

lymphocytes based on standard procedures (Ghahroudi et al., 1997). A single dominant

VHH clone known as „NAT-267‟ was selected after four rounds of library panning against

15-DON-OVA conjugate. Both NAT-267 VHH monomer and pentamer demonstrated

absolute binding specificity to 15-AcDON. NAT-267 VHH nucleotide sequence results

(GenBank: EU676170.1 http://lifesciencedb.jp/ddbj/ff_view.cgi?accession=EU676170)

confirmed that Ag specificity was generated by somatic hypermutations within the CDR

regions. The NAT-267 VHH fragment was highly specific to 15-AcDON as it did not

Page 183: PhD Fusarium

160

recognize structurally similar trichothecenes, particularly DON and 3-AcDON. We

attributed the exquisite specificity of NAT-267 VHH to the fact that both the immunogen

and panning mycotoxin protein conjugates (BSA and OVA, respectively) were linked to

DON at carbon 15 using a common hemi-succinate linker.

The methylotropic yeast Pichia pastoris was selected as a model eukaryotic test

system based on expected sensitivity to 15-AcDON and well-established capacity to

produce well-characterized quantities of functional protein. The P. pastoris system was a

convenient and cost-effective system to generate reproducible data to assess the ability of

intracellular antibodies (intrabodies) to modulate mycotoxin-specific cytotoxocity as a

precursor to transformation of higher order plants. Mycotoxin-mediated cytotoxicity was

assessed by continuous measurement of cellular growth in real time which allowed for

precise measurements of cytotoxicity during initial exposure to each ribotoxin treatment.

At equivalent doses, 15-AcDON was significantly more toxic to wild-type P. pastoris

than was DON which, in turn, was more toxic than 3-AcDON. The order of

trichothecene toxicity established within our work aligned with results previously

published in other species of yeast and model plant systems. Our observation of no

measureable cytotoxicity of 3-AcDON was in agreement with previous studies which

demonstrated that the addition of an acetyl ester group at C-3 significantly limits DON

cytotoxicity. Intracellular expression of toxin-specific VHH within P. pastoris conveyed

significant (p = 0.01) resistance to 15-AcDON cytotoxicity at doses ranging from 20 to

100 µg·mL-1

. A key limitation of NAT-267 VHH efficacy was that rAb binding to 15-

AcDON was of low affinity. The specific limitation was that the Ab-Ag complex most

Page 184: PhD Fusarium

161

likely had a short „residence time‟ which limited VHH efficacy, especially at higher doses

of 15-AcDON. Furthermore, we documented a biochemical transformation of DON to

15-AcDON which explained significant attenuation to the phytotoxic effects of DON at

100 and 200 µg·mL-1

. This “proof of concept” model established in this work suggests

that in planta VHH expression may lead to enhanced tolerance to mycotoxins and thereby

serve as a novel tool to help reduce the impact of Fusarium infection of commercial

agricultural crops. Furthermore, based on these results, we accept our hypothesis in

affirming that the expression of a recombinant VHH antibody fragment within a model

eukaryotic test system can limit mycotoxin-specific cytotoxicity. However, we reiterate

that applications of mycotoxin-specific rAb fragments would be most effectively applied

as a tool to complement (and hopefully enable) other means of enhanced FHB resistance

in higher order plants.

We recommend that future experiments and next steps be focused on the

development of VHH rAbs with improved dissociation constants (KD) to ensure a longer

residence time supported by stronger association between VHH and the target ligand for

improved efficacy (Copeland et al., 2006). It would also be of interest to develop and test

novel VHH fragments for various other trichothecenes (e.g., neosolaniol,

diacetoxyscripenol, T-2 toxin, etc.) and perhaps other classes of mycotoxin (e.g.,

fumonisins, aflatoxins, etc.). A logical subsequent application would be constitutive in

planta expression of optimized mycotoxin-specific VHH fragments, possibly with

catalytic activity, to bind and deactivate/degrade mycotoxins during critical initial periods

of Fusarium pathogenesis.

Page 185: PhD Fusarium

162

7 REFERENCES

Abdel-Salam, H.A., El-Khamissy, T., Enan, G.A., Hollenberg, C.P. 2001. Expression of

mouse anticreatine kinase (MAK33) monoclonal antibody in the yeast Hansenula

polymorpha. Appl. Micro. Biotech. 56: 157-64.

Abolmaali, S., Mitterbauer, R., Spadiut, O., Peruci, M., Weindorfer, H., Lucyshyn, D.,

Ellersdorfer, G., Lemmens, M., Moll, W.-D., Adam, G. 2008. Engineered bakers

yeast as a sensitive bioassay indicator organism for the trichothecene toxin

deoxynivalenol. J. Microbiol. Meth. 72: 306-312.

Abramson, D., Clear, R.M., Gaba, D., Smith, D.M., Patrick, S.K. Patrick, Saydak, D.

2001. Trichothecene and Moniliformin Production by Fusarium Species from

Western Canadian Wheat. J. Food Protec. 64: 1220-1225.

Adam, G.C., Lemmens, M. 1996. Involvement of ABC transporter proteins in

trichothecene resistance. 8th

Int. Congress Molecular Plant-Microbe Interactions.

Knoxville, TN, Abstract B-3.

Adam, G., Mitterbauer, R., Raditschniz, A., Weindorfer, H., Glössl, J. 2000.

Introduction of a modified ribosomal protein L3 gene as a strategy to increase

trichothecene toxin resistance in plants. pp. 1-3. In: [Proc.] 2000 National Fusarium

Head Blight Forum, 10-12 December 2000, Erlanger, KY.

Adams, G.P., Schier, R. 1999. Generating improved single-chain Fv molecules for

tumor targeting. J. Immun. Meth. 231: 249-260.

Alexander, N.J. 2008. The TRI101 story: engineering wheat and barley to resist

Fusarium head blight. World Mycotoxin Journal. 1: 31-37.

Alexander, N.J., McCormick, S.P., Hohn, T.M. 1999. TRI12, a trichothecene efflux

pump from Fusarium sporotrichioides: Gene isolation and expression in yeast. Mol.

Gen. Genet. 261: 977-984.

Almquist, K. C., McLean, M. D., Niu, Y., Byrne, G., Olea-Popelka, F.C., Murrant, C.,

Barclay, J., Hall, J.C. 2006. Expression of an anti-botulinum toxin A neutralizing

single-chain Fv recombinant Ab in transgenic tobacco. Vaccine. 24: 2079-2086.

Almquist, K.C., Niu, Y., McLean, M.D., Mena, F.L., Yau, K.Y.F., Brown, K., Brandle,

J.E., Hall, J.C. 2004. Immunomodulation confers herbicide resistance in plants.

Plant Biotech. J. 2: 189-197.

Alvarez-Rueda, N.G., Ferré, V., Pugnière, M., Roquet, F., Gastinel, L., Jacquot, C.,

Aubry, J., Baty, D., Barbet, J., Birklè, S. 2007. Generation of llama single-domain

antibodies against methotrexate, a prototypical hapten. Mol. Immun. 44: 1691-1701.

Page 186: PhD Fusarium

163

Anand, N. N., Dubuc, G., Phipps, J., MacKenzie, C.R., Sadowska, J., Young, N.M.,

Bundle, D.R., Narang, S.A. 1991. Synthesis and expression in Escherichia coli of

cistronic DNA encoding an antibody fragment specific for a Salmonella Serotype B

O-antigen. Gene. 100, 39-44.

Andersen, A.L., 1948. The development of Gibberella zeae headblight of wheat.

Phytopathol. 38: 595-611.

Andris, J.S., Abraham, S.R., Pascual, V., Pistillo, M.P., Mantero, S., Ferrara, G.B., Capra,

J.D. 1995. The human antibody repertoire: heavy and light chain variable region

gene usage in six alloantibodies specific for human HLA class I and class II

alloantigens. Mol. Immunol. 32: 1105-1122.

Arbabi-Ghahroudi, M., Tanha, J., MacKenzie, R. 2005. Prokaryotic expression of

antibodies. Can. Metastasis. Rev. 24: 501-519.

Artsaenko, O., Kettig, B., Fiedler, U., Conrad. U., During, K. 1998. Potato tubers as a

biofactory for recombinant antibodies. Mol. Breed. 4: 313-319.

Artsaenko, O., Peisker, M., zur Nieden, U., Fiedler, U., Weiler, E.W., Muntz, K., Conrad,

U. 1995. Expression of a single-chain Fv antibody against abscissic acid creates a

wilty phenotype in transgenic tobacco. Plant J. 8: 745-750.

Atanasoff, Z., Nakamura, C., Mori, N., Kaneda, C., Kato, H., Jin, Y.-Z., Koshizawa, T.,

Murai, K. 1994. Mycotoxin production and pathogenicity of Fusarium species and

wheat resistance to Fusarium head blight. Can. J. Bot. 72: 161-167.

Atwell, J.L., K.A. Breheney, Lawrence, L.J., McCoy, A.J., Kortt, A.A., Hudson, P.J.

1999. scFv multimers of the anti-neuraminidase antibody NC10: length of the linker

between VH and VL domains dictates precisely the transition between diabodies and

triabodies. Protein Eng. 1999; 12: 597-604.

Bai, G-H, Shaner, G.E. 1994. FHB of wheat: perspective and control. Plant Disease.

78. 760-766.

Bakan, B., Bily, A.C., Melcion, D., Cahagnier, B., Regnault-Roger, C., Phiologene,

B.J.R., Richard-Molard, D. 2003. Possible role of plant phenolics in the production

of trichothecenes by Fusarium graminearum strains on different fractions of maize

kernels. J. Agric. Food Chem. 51: 2826-2831.

Baláza, F., Bagi, F. 1997. Polygalacturonase and cellulase activity of different strains of

Fusarium graminearum. Cereal Res. Commun. 25: 725-726.

Balint, R.F., Larrick, J.W. 1993. Antibody engineering by parsimonious mutagenesis.

Gene. 137: 109-118.

Page 187: PhD Fusarium

164

Balzi, E., Wang, M., Leterme, S., Van Dyck, L., Goffeau, A. 1994. PDR5, novel yeast

multidrug resistance conferring transporter controlled by the transcription regulator

PDR1. J. Biol. Chem. 269: 2206-2214.

Barbas, C.F. III, Bain, J.D., Hoekstra, D.M., Lerner, R.A. 1992. Semisynthetic

combinatorial antibody libraries: a chemical solution to the diversity problem. Proc.

Natl. Acad. Sci. USA. 89: 4457-4461.

Bauminger, S., Wilchek, M., 1980. The use of carbodiimides in the preparation of

immunizing conjugates. Method Enzymol. 70, 151-159.

Benhar, I., Pastan, I. 1994. Cloning, expression, and characterization of the Fv fragments

of the anti-carbohydrate mAbs B1 and B5 as single chain immunotoxins. Prot. Eng.

7: 1509-1515.

Bergstrom, G. C. 1993. Scab (head blight). pgs. 83-93. In: Seed-borne Diseases and Seed

Health Testing of Wheat. S.B. Mathur and B.M. Cunfer (eds.). Jordbrugsforlaget.

Frederiksberg, Denmark.

Berthiller, F., Lemmens, M., Werner, U., Krska, R., Hauser, M.-T., Adam, G.,

Schumacher, R. 2007. Short review: Metabolism of the Fusarium mycotoxins

deoxynivalenol and zearalenone in plants. Mycotoxin. Res. 23: 68-72.

Betina, V. 1989. Structure-activity relationships among mycotoxins. Chem. Biol.

Interact. 7: 105-146.

Better, M., Chang, C.P., Robinson, R.R., Horwitz, A.H. 1988. Escherchia coli secretion

of an active chimeric antibody fragment. Science. 240: 1041-1043.

Binder, J. 1999. A yeast bioassay for trichothecenes. Natural Toxins. 7: 401-406.

Binder, J., Horvath, E.M., Heidegger, J., Ellend, N., Danner, H., Krska, R., Braun, R.,

1997. A bioassay for comparison of the toxicity of trichothecenes and their microbial

metabolites. Cereal Res. Comm. 25, 489-491.

Bird, R.E., Hardman, K.D., Jacobson, J.W., Johnson, S., Kaufman, B.M., Lee, S.M., Lee,

S.H., Pope, G.S., Riordan, G.S., Whitlow, M. 1989. Single-chain antigen-binding

proteins. Science. 242: 423-426.

Biswas, S.K., Yamaguchi, M., Naoe, N., Takashima, T., Takeo, K. 2003. Quantitative

three-dimensional structural analysis of Exophiala dermatitidis yeast cell by freeze-

substitution and serial ultrathin sectioning. J. Electron. Microsc. 52: 133-143.

Boder, E.T., Wittrup, K.D. 1997. Yeast surface display for screening combinatorial

polypeptide libraries. Nat. Biotech. 15: 553-557.

Borrebaeck, C.K., Ohlin, M. 2002. Antibody evolution beyond nature. Nat. Biotech. 20:

1189-1190.

Page 188: PhD Fusarium

165

Boutigny, A-L., Richard-Forget, F., Barreau, C. 2008. Natural mechanisms for cereal

resistance to the accumulation of Fusarium trichothecenes. Eur. J. Plant Pathol. 121:

411-423.

Brian, P.W., Dawkins, A.W., Grove, J.F., Hemming, H.G., Lowe, R. Morris, G.L.F.

1961. Phytotoxic compounds produced by Fusarium equiseti. J. Exp. Bot. 12: 1-12.

Buerstmayr, H., M. Lemmens, H. Grausgruber, and P. Ruckenbauer. 1996. Scab

resistance of international wheat germplasm. Cereal Res. Commun. 24: 195-202.

Burton, D.R., Barbas, C.F., Persson, M.A., Koenig, S., Chanock, R.M., Lerner, R.A.

1991. A large array of human monoclonal antibodies to type 1 human

immunodeficiency virus from combinatorial libraries of asymptomatic seropositive

individuals. Proc. Natl. Acad. Sci. USA., 88: 10134-10137.

Bushnell, W.R., Hazen, B.E., Pritsch, C. 2003. Histology and Physiology of Fusarium

Head Blight. pp. 44-83 In: Fusarium Head Blight of Wheat and Barley. K.J.

Leonard and W.R. Bushnell (eds.). Am. Phytopath Society. St. Paul, MN.

Bushnell, W.R., Seeland, T.M., Krueger, D.E. 2002. Light-dependent bleaching of

detached barley leaf tissue by deoxynivalenol. Phytopathol. 92: S11 (Abstr.).

Canady, R.A., Coker, R.D., Egan, S.K., Krska, R., Kuiper-Goodman, T., Olsen, M.,

Pestka, J., Resnik, S., Schlatter, J. 2001. Deoxynivalenol. IPCS INCHEM

http://www.inchem.org/documents/jecfa/jecmono/v47je05.htm

Carter, C.J., Cannon, M. 1977. Structural requirements for the inhibitory action of

12,13-epoxytrichothecenes on protein synthesis in eukaryotes. Biochem. J. 166: 399-

409.

Cereghino, J.L., Cregg, J.M., 2000. Heterologous protein expression in the

methylotrophic yeast Pichia pastoris. FEMS Microbiol. Rev. 24: 45–66.

Chames, P., Coulon, S., Baty, D. 1998. Improving the affinity and the fine specificity of

an anti-cortisol antibody by parsimonious mutagenesis and phage display. J.

Immunol. 161: 5421-5429.

Chelkowski, J., 1989. Formation of mycotoxins produced by Fusaria in heads of wheat,

triticale, and rye. pp. 63-84. In: Fusarium - Mycotoxins, Taxonomy, and

Pathogenicity. J. Chelkowski (ed.), Elsevier, Amsterdam, The Netherlands.

Chen, L-F.; Bai, G-H., Desjardins, A.E. 2000. Recent advances in wheat and head scab

research in China. pp. 258-273 in: [Proc.] International Symposium on Wheat

Improvement for Scab Resistance. Suzhou and Nanjing, China, 5-11 May, 2000.

(http://www.nal.usda.gov/pgdic/WHS/whsindex.html).

Choi, G.-H., Lee, D-.H., Min, W-.K., Cho, Y-.J., Kweon, D-.H., Son, D-.H., Park, K.,

Seo, J.-H. 2004. Cloning, expression and characterization of single-chain variable

Page 189: PhD Fusarium

166

fragment antibody against mycotoxin deoxynivalenol in recombinant Escherichia coli.

Prot. Exp. Purif. 35: 84-92.

Chowdhury, P.S., Pastan, I. 1999. Improving antibody affinity by mimicking somatic

hypermutation in vitro. Nat. Biotech. 17: 568-572.

Clackson, T., Hoogenboom, H.R., Griffiths, A.D., Winter, G. 1991. Making antibody

fragments using phage display libraries. Nature. 352: 624-628.

Coia, G., Hudson, P.J., Irving, R.A. 2001a. Protein affinity maturation in vivo using E.

coli mutator cells. J. Immun. Meth.; 251: 187-193.

Coia, G., Pontes-Braz, L., Nuttal, S.D., Hudson, P.J., Irving, R.A. 2001b. Panning and

selection of proteins using ribosome display. J. Immun. Meth.; 254: 191-197.

Conrad, U., Fiedler, U. 1998. Compartment-specific accumulation of recombinant

immuno-globulins in plant cells: an essential tool for antibody production and

immunomodulation of physiological functions and pathogen activity. Plant Mol.

Biol. 38: 101-109.

Cook, G.P., Tomlinson, I.M. 1995. The human immunoglobulin VH repertoire.

Immunol. Today. 16: 237-242.

Copeland, R.A., Pompliano, D.L., Meek, T.D. 2006. Drug-target residence time and its

implications for lead optimization. Nature. 5: 730-739.

Cossette, F., Miller, J.D. 1995. Phytotoxic effects of deoxynivalenol and Gibberella ear

rot resistance in corn. Natural Toxins. 3: 383-388.

Cregg, J.M., Vedvick, T.S., Easchke, W.C. 1993. Recent advantages in the expression of

foreign gene in Pichia pastoris. Bio/Technol. 11: 905-910.

Creppy, E.E. 2002. Update of survey, regulation and toxic effects of mycotoxins in

Europe. Toxicology Letters. 127: 19-28.

Cundliffe, E. 1989. How antibiotic-producing organisms avoid suicide. Annu. Rev.

Microbiol. 4: 207-233.

Cutler, H.G., Jarvis, B.B. 1985. Preliminary observations on the effects of macrocyclic

trichothecenes on plant growth. Env. Exp. Bot. 25: 115-128.

D‟Mello, J.P.F., MacDonald, A.M.C., Postel, D., Dijksma, T.P., Dujardin, A., Placinta,

C.M. 1998. Pesticide use and mycotoxin production in Fusarium and Aspergillus

phytopathogens. Eur. J. Plant Path., 104, 741–751.

D‟Mello, J.P.F., Placinta, C.M., Macdonald, A.M.C. 1999. Fusarium mycotoxins: a

review of global implications for animal health, welfare and productivity. An. Feed

Sci. Tech. 80: 183-205.

Page 190: PhD Fusarium

167

da Luz, W.C., Stockwell, C.A., Bergstrom, G.C. 2003. Biological Control of Fusarium

graminearum. pp. 381-394 In: Fusarium Head Blight of Wheat and Barley. K.J.

Leonard and W.R. Bushnell (eds.). Am. Phytopath Society. St. Paul, MN.

Daniell, H., Carmona-Sanchez, O., Burns, B. 2004. Chloroplast derived antibodies,

biopharmaceuticals and edible vaccines. pp. 113-133. In: Molecular Farming. R.

Fischer, S. Schillberg (eds). Weinheim: Wiley-VCH.

Daugherty, P.S., Chen, G., Olsen, M.J., Iverson, B.L., Georgiou, G. 1998. Antibody

affinity maturation using bacterial surface display. Prot. Eng. 11: 825-832.

Daugherty, P.S., Olsen, M.J., Iverson, B.L., Georgiou, G. 1999. Development of an

optimized expression system for the screening of antibody libraries displayed on the

Escherichia coli surface. Prot. Eng. 12: 613-621.

Davies, J., Riechmann, L. 1994. „Camelising‟ human antibody fragments: NMR studies

on VH domains. FEBS Lett. 339: 285-290.

Davies, J., Riechmann, L. 1996a. Single antibody domains as small recognition units:

design and in vitro antigen selection of camelized, human VH domains with improved

protein stability. Protein Eng. 9: 531-537.

Davies, J., Riechmann, L. 1996b. Affinity improvement of single antibody VH domains:

residues in all three hypervariable regions affect antigen binding. Immunotech. 2:

169-179.

de Bruin, K., Spelt, J., Mol, R. Koes, R., Quattrocchio, F. 1999. Selection of high-

affinity phage antibodies from phage display libraries. Nature Biotech. 17: 397-399.

de Kruif, J., Boel, E., Logtenberg, T. 1995. Selection and application of human single

chain Fv antibody fragments from a semi-synthetic phage antibody display library

with designed CDR3 regions. J Mol Biol. 248: 97-105.

Decottignies, A., Goffeau, A. 1997. Complete inventory of the yeast ABC proteins. Nat.

Genetic. 15: 137–145.

Del Ponte, E.M., Fernandes, J.M.C., Bergstrom, G.C. 2007. Influence of Growth Stage

on Fusarium Head Blight and Deoxynivalenol Production in Wheat. J. Phytopath.

155, 577-581

Demangel, C., Zhou, J., Choo, A.B.H., Shoebridge, G., Halliday, G.M., Britton, W.J.

2005. Single chain antibody fragments for the selective targeting of antigens to

dendritic cells. Molec. Immun. 42: 979-985.

Demcey Johnson, D., Flaskerud, G.K., Taylor, R.D., Satyanarayana, V. 2003.

Quantifying Economic Impacts of Fusarium Head Blight in Wheat. pp. 461- 483. In:

Fusarium Head Blight of Wheat and Barley. K.J. Leonard and W.R. Bushnell (eds.).

Am. Phytopath. Society. St. Paul, MN.

Page 191: PhD Fusarium

168

Demont, M., Devos, Y. 2008. Regulating coexistence of GM and non-GM crops without

jeopardizing economic incentives. Trends BioTech. 26: 353-358.

Desjardins, A.E. 2006. Trichothecenes. pp. 13-64 In: Fusarium Mycotoxins.

Chemistry, Genetics, and Biology. Am. Phytopath. Society. St. Paul, MN.

Desjardins, A.E., Bai, G.H., Plattner, R.D. 2000. Analysis of aberrant virulence of

Gibberella zeae following transformation of a trichothecene-deficient (Tri5) mutant.

Microbiology. 146. 2059-2068.

Desjardins, A.E., Hohn, T.M. 1997. Mycotoxins in Plant Pathogenesis. Mol. Plant

Microbe Interact. 10: 147-152.

Desjardins, A.E., McCormick, S.P., Appell, M. 2007. Structure-activity relationships of

trichothecene toxins in an Arabidopsis thaliana leaf assay. J. Agric. Food Chem. 55:

6487-6492.

Desjardins, A.E., Plattner, R.D., Spencer, G.F. 1988. Inhibition of trichothecene toxin

biosynthesis by naturally occurring shikimate aromatics. Phytochem. 27: 767-771.

Desjardins, A.E., Proctor, R.H., Bai, G., McCormick, S.P., Shaner, G., Buechley, G.,

Hohn, T.M. 1996. Reduced virulence of trichothecene-nonproducing mutant of

Gibberella zeae in wheat field tests. Mol. Plant Microbe Interact. 9: 775-781.

Desjardins, A.E., Spencer, G.F., Plattner, R.D., Beremand, M.N. 1989. Furanocoumarin

phytoalexins, trichothecene toxins and infection of Pastinaca sativa by Fusarium

sporotrichioides. Phytopathol. 79: 170-175.

Desjardins, A.E., T.M. Hohn, and S.P. McCormick. 1992. The effect of gene disruption

on trichodiene synthase (Tox5) on the virulence of Gibberella pulicaris. Mol. Plant

Microbe Interact. 5: 214-222.

Desmyter, A., Decanniere, K., Muyldermans, S., Wyns, L., 2001. Antigen specificity and

high affinity binding provided by one single loop of a camel single-domain antibody.

J. Biol. Chem. 276, 26285-26290.

Desmyter, A., Spinelli, S. Payan, F.; Lauwereys; M. Wyns, L., Muyldermans, S.,

Cambillau, C. 2002. Three camelid VHH domains in complex with porcine

pancreatic α-amylase. J. Biol. Chem. 277: 23645–23650.

Desmyter, A., Transue, T.R., Ghahroudi, M.A., Thi, M.H., Poortmans, F., Hamers, R.,

Muyldermans, S., Wyns, L. 1996. Crystal structure of a camel single-domain VH

antibody fragment in complex with lysozyme. Nat. Struct. Biol. 3: 803-811.

Desplancq, D., King, D.J., Lawson, A.D.G., Mountain, A. 1994. Multimerization

behaviour of single chain Fv variants for the tumor-binding antibody B72.3. Protein

Eng. 7: 1027-1033.

Page 192: PhD Fusarium

169

Diaz de Ackermann, M., Kohli, M.M. 1997a. Research on Fusarium head blight of

wheat in Uruguay. pp. 13-18. In: Fusarium Head Scab: Global Status and Future

Prospects. H.J. Dubin, L. Gilchrist, J. Reeves, and A. McNab (eds.). CIMMYT, DF,

Mexico.

Diaz de Ackermann, M., Kohli, M.M. 1997b. Screening for Fusarium head blight

resistance in wheat. Cereal Res. Commun. 25: 733-734.

Dickson, J.G. 1942. Scab of wheat and barley and its control. USDA Farmers Bull. No.

1599 (revised): 1-22.

Dill-Macky, R. 2003. Inoculation Methods and Evaluation of Fusarium Head Blight

Resistance in Wheat. pp. 184-210. In: Fusarium Head Blight of Wheat and Barley.

K.J. Leonard and W.R. Bushnell (eds.). Am. Phytopath. Society. St. Paul, MN.

Dill-Macky, R., Jones, R.K. 2000. The effect of previous crop residues and tillage on

Fusarium head blight of wheat. Plant Dis. 84: 71-76.

Dolk, E., van der Vaart, M., Hulsik, D.L., Vriend, G., de Haard, H., Spinelli, S.,

Cambillau, C., Frenken, L., Verrips, T. 2005. Isolation of llama antibody fragments

for prevention of dandruff by phage display in shampoo. App. Env. Microbiol. 71,

442-450.

Dong, W., Yang, L., Shen, K., Kim, B., Kleter, G.A., Marvin, H.J.P., Guo, R., Liang, W.,

Zhang, D. 2008. GMDD: a database of GMO detection methods. BMC

Bioinformatics. Vol. 9: Art. 260. 10 p.

Dörsam, H., Rohrbach, P., Kürschner, T., Kipriyanov, S., Renner, S., Braunagel, M.,

Welschof, M., Little, M. 1997. Antibodies to steroids from a small human naive IgM

library. FEBS Lett. 414: 7-13.

Doyle, P.J., Arbabi-Ghahroudi, M., Gaudette, N., Furzer, G., Savard, M.E., Gleddie, S.,

McLean, M.D., MacKenzie, C.R., Hall, J.C. 2008. Cloning, Expression, and

Characterization of a Single Domain Antibody Fragment with Affinity for 15-Acetyl-

Deoxynivalenol. Molec. Imm. 45: 3703 - 3713.

Düring, K., Hippe, S., Kreuzaler, F., Schell, J. 1990. Synthesis and self-assembly of a

functional monoclonal antibody in transgenic Nicotiana tabacum. Plant Molec. Biol.

15: 281-293.

Ehrlich, K.C., Daigle, K.W. 1987. Protein synthesis inhibition by 8-oxo-12, 13-

epoxytrichothecenes. Biochem. Biophys. Acta. 923: 206-213.

Ellgaard, L. 2004. Catalysis of disulfide bond formation in the endoplasmic reticulum.

Biochem. Soc. Trans. 32: 663-667.

Page 193: PhD Fusarium

170

Eudes, F., Comeau, A., Rioux, S., Collin, J. 2001. Impact of trichothecenes on Fusarium

head blight [Fusarium graminearum] development in spring wheat (Triticum

aestivum). Can. J. Plant Pathol. 23, 318-322.

Evans, C.K., Kolaczkowski, E.K., Mirocha, C.J., Dill-Macky, R. 1997. Trichothecenes

in florets of barley inoculated with Fusarium graminearum. Phytopathol. 87: S29

(Abstr.).

Feinberg, B., McLaughlin, C.S. 1989. Biochemical mechanism of action of trichothecene

mycotoxins. pp.27-35 In: Trichothecene Mycotoxicosis: Pathophysiologic Effects.

U.R. Beasley (ed.) CRC Press, Boca Raton, FL.

Fellouse, F.A., Esaki, K., Birtalan, S., Raptis, D., Cancasci, V.J., Koide, A., Jhurani, P.,

Vasser, M., Wiesmann, C., Kossiakoff, A.A., Koide, S., Sidhu, S.S. 2007. High-

throughput generation of synthetic antibodies from highly functional minimalist

phage-displayed libraries. J. Mol. Biol. 373: 924-940.

Fernández, L.E., Gómez, Pacheco, S., Arenas, I., Gilla, S.S., Bravo, A., Soberón. 2008.

Employing phage display to study the mode of action of Bacillus thuringiensis Cry

toxins. Peptides. 29: 324-329.

Fernandez, M.R., Fernandez, J.M., Sutton, J.C. 1993. Effects of fallow and of summer

and winter crops on survival of wheat pathogens in crop residues. Plant Dis. 77: 698-

703.

Fernandez-Lobato, M., Cannon, M.M., Mitlin, J.A., Mount, R.C. 1990. Characterization

of Saccharomyces cerevisiae strains displaying high-level or low-level resistance to

trichothecene antibiotics. Biochem. J. 267: 709-713.

Fernando, W.G.D., Paulitz, T.C., Seaman, W.L., Dutillleul, P., Miller, J.D. 1997. Head

blight gradients caused by Gibberella zeae from area sources of inoculum in wheat

field plots. Phytopathol. 87: 414-421.

Fischer, R., Vaquero-Martin C., Sack, M., Drossard, J., Emans, N. Commandeur, U.

1999. Towards molecular farming in the future: transient protein expression in

plants. Biotech. Appl. Biochem. 30: 113-116.

Filpula, D. 2007. Antibody engineering and modification technologies. Review.

Biomolec. Eng. 24: 201-215.

Foroud, N.A., Eudes, F. 2009. Trichothecenes in Cereal Grains. Review. Int. J. Mol.

Sci. 10: 147-173.

Forrer, P., Jung, S., Plückthun, A. 1999. Beyond binding: using phage display to select

for structure, folding and enzymatic activity in proteins. Curr. Opin. Struct. Biol. 9:

514-520.

Page 194: PhD Fusarium

171

Francisco, J.A., R. Campbell, B.L. Iverson, and G. Georgiou. 1993. Production and

fluorescence-activated cell sorting of Escherchia coli expressing a functional antibody

fragment on the external surface. Proc. Natl. Acad. Sci. USA. 90: 10444-10448.

Freeman, G.G. and R.I. Morrison. 1948. Trichothecin: An antifungal metabolic product

of Trichothecium roesum Link. Nature. 162: 30-31.

Frenken, L.G., Hessing, J.G., van den Hondel, C.A., Verrips, C.T. 1998. Recent

advances in the large-scale production of antibody fragments using lower eukaryotic

microorganisms. Res. Immun. 149: 589-599.

Frenken, L.G.J., van der Linden, R.H.J., Hermans, P.W.J., Bos, J.W., Ruuls, R.C., de

Geus, B., Verrips, C.T., 2000. Isolation of antigen specific llama VHH antibody

fragments and their high level secretion by Saccharomcyes cerevisiae. J. Biotech.

78, 11-21.

Freyre, F.M., Vazquez, J.E., Ayala, M., Canaan-Haden, L., Bell, H., Rodriguez, I.,

Gonzalez, A., Cintado, A., Gavilondo, V. 2000. Very high expression of an anti-

carcinoembryonic antigen single chain Fv antibody fragment in the yeast Pichia

pastoris. J. Biotech. 76: 157-163.

Fried, H.M., Warner, J.R. 1981. Cloning of yeast gene for trichodermin resistance and

ribosomal protein L3. Proc. Natl. Acad. Sci. USA. 78: 238-242.

Fuchs, E., Binder, E.M., Heidler, D., Krska, R. 2002. Structural characterization of

metabolites after microbial degradation of type A trichothecenes by the bacterial

strain BBSH 797. Food Addit. Contam. 19: 379-386.

Fuchs, P., Breitling, F., Dübel, S., Seehaus, T., Little, M. 1991. Targeting recombinant

antibodies to the surface of Escherchia coli: fusion to a peptidoglycan associated

lipoprotein. Bio/Tech. 1991; 9: 1369-1372.

Furzer, G.S., Veldhuis, L., Hall, J.C., 2006. Development and comparison of three

diagnostic immunoassay formats for the detection of Azoxystrobin. J. Agric. Food

Chem. 54, 688-693.

Gach, J.S., Maurer, M., Hahn, R., Gasser, B., Mattanovich, D., Katinger, H., Kunert, R.

2007. High level expression of a promising anti-idiotypic antibody fragment vaccine

against HIV-1 in Pichia pastoris. J. Biotech. 128: 735-746.

Gasser, B., Maurer, M., Gach, J., Kunert, R., Mattanovich, D. 2006. Engineering of

Pichia pastoris for improved production of antibody fragments. Biotech. Bioeng. 94:

353-361.

Georgiou, G., Poetschke, Stathopoulos, C., Francisco, J.A. 1993. Practical applications

of engineering gram-negative bacterial cell surfaces. Trend. Biotech. 11: 6-10.

Georgiou, G., Stathopoulous, C., Daugherty, P.S., Nayak, A.R., Iverson, B.L., Curtiss, R.

Page 195: PhD Fusarium

172

1997. Display of heterologous proteins on the surface of microorganisms: from the

screening of combinatorial libraries to live recombinant vaccines. Nat. Biotech. 15:

29-34.

Ghahroudi, M., Desmyter, A., Wyns, L., Hamers, R., Muyldermans, S. 1997. Selection

and identification of single domain antibody fragments from camel heavy-chain

antibodies. FEBS Lett. 414: 521-526.

Gilbert, J.; A. Tekauz; E. Mueller; and U. Kromer. 1994. Occurrence of Fusarium head

blight in Manitoba in 1993. Can. Plant Disease Survey: 77-78.

Gilchrist, L., Rajaram, S., van Ginkel, M., Kazi, M., Franco, J. 1997. Characterizing

Fusarium graminearum resistance of CIMMYT bred wheat germplasm. Cereal Res.

Commun. 25: 655-658.

Giritch, A., Marillonnet, S., Engler, C., van Eldik, G., Botterman, J., Klimyuk, V., Gleba,

Y. 2006. Rapid high-yield expression of full-size IgG antibodies in plants coinfected

with non-competing viral vectors. Proc.Natl.Acad.Sci.USA 103: 14701-14706.

Goldman, E.R., Anderson, G.P., Liu, J.L., Delehanty, J.B., Sherwood, L.J., Olson, L.E.,

Cummins, L.B., Hayhurst, A., 2006. Facile generation of heat-stable antiviral and

antitoxin single domain antibodies from a semisynthetic llama library. Anal. Chem.

78: 8245-8255.

Goswami, R.S., Kistler, H.C. 2004. Pathogen Profile. Heading for disaster: Fusarium

graminearum on cereal crops. Molec. Plant Pathol. 5: 515-525.

Greenhalgh, R., Blackwell, B.A., Pare, J.R.J., Miller, J.D., Levandier, D., Meier, R.-M.,

Taylor, A., Ap-Simon; J.W. 1986. Isolation and characterization by mass

spectroscopy of secondary metabolites of some Fusarium species. pp. 137-152. In:

Mycotoxins and Phycotoxins. P.S. Steyn, R. Vleggaar (eds.), Elsevier, Amsterdam,

NL.

Griffiths, A.D., M. Malmqvist, J.D. Marks, J.M. Bye, M.J. Embelton, J. McCafferty, M.

Baier, K.P. Holliger, B.D. Gorick, N.C. Hughes-Jones, H.R. Hoogenboom, and G.

Winter. 1993. Human anti-self antibodies with high specificity from phage display

libraries. EMBO J. 12: 725-734.

Griffiths, A.D., Williams, S.C., Hartley, O., Tomlinson, I.M., Waterhouse, P., Crosby,

W.L., Kontermann, R.E., Jones, P.T., Low, N.M., Prospero, T.D., Hoogenboom,

H.R., Nissim, A., Cox, J.P.L., Harrison, J.L., Zaccolo, M., Gherardi, E., Winter, G.

1994. Isolation of high affinity human antibodies directly from large synthetic

repertoires. EMBO J. 13: 3245-3260.

Gunneriusson, E., Samuelson, P., Uhlen, M., Nygren, P.A., Stahl, S. 1996. Surface

display of a functional single-chain Fv antibody on staphylococci. J. Bacteriol. 178:

1341-1346.

Page 196: PhD Fusarium

173

Gurkan, C., Symeonides, S.N., Ellar, D.J. 2004. High level production of Pichia

pastoris of an anti-pI85HER-2

single-chain antibody fragment using an alternative

secretion expression vector. Biotechnol. Appl. Biochem. 39: 115–122.

Guttieri, M.C., Bookwalter, C., Schmaljohn, C. 2000. Expression of a human,

neutralizing monoclonal antibody specific to Puumala virus G2-protein in stably-

transformed insect cells. J. Immun. Meth. 246: 97-108.

Hamers-Casterman, C., Atarhouch, T., Muyldermans, S., Robinson, G., Hamers, C.,

Songa, E.B., Bendahman, N., Hamers, R., 1993. Naturally-occurring antibodies

devoid of light-chains. Nature. 363: 446-448.

Hanes, J., Plückthun, A. 1997. In vitro selection and evolution of functional proteins by

using ribosome display. Proc.Natl.Acad.Sci.USA. 94: 4937-4942.

Hanes, J., Jermutus, L., Weber-Bornhauser, S., Bosshard, H.R., Plückthun. A. 1998.

Ribosome display efficiently selects and evolves high-affinity antibodies in vitro from

immune libraries. Proc.Natl.Acad.Sci.USA. 95: 14130-14135.

Harding, S.E., Longman, E., Carrasco, B., Ortega, A., Garcia, J. 2004. Studying

Antibody Confirmations by Ultracentrifugation and Hydrodynamic Modeling. pp. 93-

113. In: Antibody Engineering. Methods and Protocols. B.K.C. Lo (ed). Humana

Press. Totowa, N.J., USA.

Harmsen, M.M., Ruuls, R.C., Nijman, I.J., Niewold, T.A., Frenken, L.G.J., de Geus, B.,

2000. Llama heavy chain V regions consist of at least four distinct subfamilies

revealing novel sequence features. Mol. Immun. 37: 579-590.

Harris, L.J., A.E. Desjardins, R.D. Plattner, P. Nicholson, G. Butler, J.C. Young, G.

Weston, R.H. Proctor, T.M. Hohn. 1999a. Possible role of trichothecene mycotoxins

in virulence of Fusarium graminearum on maize. Plant Disease. 83: 954-960.

Harris, L.J., Gleddie, S.C., Simmonds, J.A. 1999b. Trichothecene mycotoxin resistance

in plants through the modification of the ribosomal protein L3 gene. Can. Patent 130:

194485.

Harris, L.J., Gleddie, S.C. 2000. Developing plant tolerance to trichothecene mycotoxins

using a transgenic approach. Phytopathology. 90 (suppl): 596.

Harris, L.J., Gleddie, S.C. 2001. A modified Rpl3 gene from rice confers tolerance of

the Fusarium graminearum mycotoxin deoxynivalenol to transgenic tobacco. Phys.

Molec. Plant Path. 58: 173-181.

He, M., Taussig, M.J. 1997. Antibody-ribosome-mRNA (ARM) complexes as efficient

selection particles for in vitro display and evolution of antibody combining sites.

Nucleic Acids Res. 25: 5132-5134.

Hellwig, S., Emde, F., Raven, N.P.G., Henke, M., van der Logt, P., Fischer, R. 2001.

Page 197: PhD Fusarium

174

Analysis of single-chain antibody production in Pichia pastoris using on-line

methanol control in fed-batch and mixed-feed fermentations. Biotech. Bioeng. 74:

344-352.

Hiatt, A., Ma, J.K. 1992. Monoclonal antibody engineering in plants. FEBS Lett. 307:

71-75.

Hiatt, A., Cafferkey, R., Bowdish, K. 1989. Production of antibodies in transgenic plants.

Nature. 342: 76-78.

Hobden, A.N., Cundliffe, E. 1980. Ribosomal resistance to the 12,13-

epoxytrichothecene antibiotics in the producing organism Myrothecium verrucaria.

Biochem. J. 190: 765-770.

Holliger, P., Hudson, P.J. 2005. Engineered antibody fragments and the rise of the single

domains. Nat. Biotech. 23: 1126-1136.

Homdork, S., Fehrmann, H., Beck, R. 2000. Effect of field application of tebuconazole

on yield, yield components and the mycotoxin content of Fusarium-infected wheat

grain. J. Phytopathol. 148: 1-6.

Hoogenboom, H.R. 1997. Designing and optimizing library selection strategies for

generating high-affinity antibodies. Trends Biotech. 15: 62-70.

Hoogenboom, H.R. 2005. Selecting and screening recombinant antibody libraries. Nat.

Biotech. 23: 1105-1116.

Hoogenboom, H.R., Chames, P. 2000. Natural and designer binding sites made by phage

display technology. Immunol. Today. 21: 371-378.

Hoogenboom, H.R., de Bruine, A.P. Hufon, S.E., Hoet, R.M., Arends, J.W., Roovers,

R.C. 1998. Antibody phage display technology and its applications. Immunotech. 4:

1-20.

Hoogenboom, H.R., Marks, J.D., Griffiths, A.D., Winter, G. 1992. Building antibodies

from their genes. Immunol. Rev. 130: 41-68.

Horwitz, A.H., Chang, C.P., Better, M., Hellstrom, K.E., Robinson, R.R. 1988.

Secretion of functional antibody and Fab fragment from yeast cells. Proc. Natl. Acad.

Sci. USA. 85: 8678-8682.

Hu, Z-Q., Li, H-P., Zhang, J-B., Glinka, E., Liao, Y-C. 2008. Review: Antibody-

mediated prevention of Fusarium mycotoxins in the field. Int. J. Mol. Sci. 9: 1915-

1926.

Huang, C-H., Li, J-H., Zhang, Y-Q., Wang, M., Liu, S. 1993. A study of the control

effect of Bacillus-HCH on scab of wheat. J. Huazhong Agric. Univ. 12: 566-569.

Huang, D., Shusta, E.V. 2006. A yeast platform for the production of single-chain

Page 198: PhD Fusarium

175

antibody-green fluorescent protein fusions. Appl. Env. Micro. 72: 7748-7759.

Hudson, P.J. 1998. Recombinant antibody fragments. Curr. Opin. Biotechnol. 9: 395-

402.

Hudson, P.J. 1999. Recombinant antibody constructs in cancer therapy. Curr. Opin.

Immunol. 11: 548-557.

Huse, W.D., Sastry, L., Iverson, S.A., Kang, A.S., Alting-Mees, M., Burton, D.R.,

Benkovic, S.J., Lerner, R.A. 1989. Generation of a large combinatorial library of the

immunoglobulin repertoire in phage lambda. Science. 246: 1275-1281.

Huston, J.S., George, A.J.T., Tai, M.-S., McCartney, J.E., Jin, D., Segal, M., Keck, P.,

Oppermann, H. 1995. Single-chain Fv design and production by preparative folding.

pp. 185-227 In: Antibody Engineering. C.A. Borrebaeck (ed). Oxford University

Press. New York, NY.

Huston, J.S., Levinson, D., Mudgett-Hunter, M., Tai, M.S., Novotny, J., Margolies, M.N.,

Ridge, R.J., Bruccoleri, R.E., Haber, E., Crea, R., Opperman, H. 1988. Protein

engineering of antibody binding sites: recovery of specific activity in an anti-digoxin

single-chain Fv analogue produced in Escherichia coli. Proc.Natl.Acad.Sci. USA.

85: 5879-5883.

Huston, J.S., Margolies, M.N., Haber, E. 1996. Antibody binding sites. Adv. Prot.

Chem. 49: 329-450.

Huston, J.S., Mudgett-Hunter, M., Tai, M.-S., McCartney, Warren, F., Haber, E.,

Oppermann, H. 1991. Protein engineering of single-chain Fv analogs and fusion

proteins. Meth. Enzymol. 203: 46-48.

Irving, R.A., Coia, G., Raicevic, A., Hudson, P.J. 2002. Use of Escherichia coli mutator

cells to mature antibodies. Meth.Mol.Biol. 178: 295-302.

Irving, R.A., Kortt, A.A., Hudson, P.J. 1996. Affinity maturation of recombinant

antibodies using E. coli mutator cells. Immunotech. 2: 127-143.

Ismaili, A., Jalali-Javaran, M., Rasaee, M.J., Rahbarizadeh, F., Forouzandeh-Moghadam,

M., Memari, H.R. 2007. Production and characterization of anti-(mucin MUC1)

single-domain antibody in tobacco (Nicotiana tabacum cultivar Xanthi). Biotech.

Appl. Biochem. 47: 11-19.

Jacobsen, B.J., Bowen, K.L., Shelby, R.A., Diener, U.L., Kemppainen, B.W., Floyd, J.

1993. Mycotoxins and Mycotoxicoses. Circular ANR-767 (02/93) Alabama

Cooperative Extension System. Alabama A&M and Auburn Universities.

Jain, M., Kamal, N., Batra, K. 2007. Engineering antibodies for clinical applications.

Trends Biotech. 25: 307-316.

Jeffrey, P.D., Strong, R.K., Sieker, L.C., Chang, C.Y.Y., Campbell, R.L., Petsko, G.A.,

Page 199: PhD Fusarium

176

Haber, E., Margolies, M.N., Sherfiff, S. 1993. 26-10 Fab-digoxin complex: affinity

and specificity due to surface complementarity. Proc.Natl.Acad.Sci. USA. 90: 10310-

10314.

Jennings, P. 2002. Control of the fungus with fungicides. pp. 22-25. In: Food safety of

cereals: A chain-wide approach to reduce Fusarium mycotoxins. O.E. Scholten; P.

Ruckenbauer; A. Visconti; W.A. van Osenbruggen; A.P.M. den Nijs (eds).

Mycotochain. http://www.mycotochain.org

Jennings, P., Turner J.A., Nicholson, P. 2000. Overview of Fusarium ear blight in the

UK-Effect of fungicide treatment on disease control and mycotoxin production. pp.

707-712. In: Pests & Diseases. Vol. 2, London: BCPC Publications.

Jobling, S.A., Jarman, C., Teh, M-.M., Holmberg, N., Blake, C. Verhoeyen, M.E. 2003.

Immunomodulation of enzyme function in plants by single-domain antibody

fragments. Nat. Biotech. 21: 77-80.

Joosten, V., Lokman, C., van den Hondel, A.M.J.J., Punt, P.J. 2003. The production of

antibody fragments and antibody fusion proteins by yeasts and filamentous fungi.

Microbial Cell Factories. 2: 1-15.

Jorgensen, P., Nishikawa, J.L., Breitkreutz, B.-J., Tyers, M. 2002. Systematic

identification of pathways that couple cell growth and division in yeast. Science.

297: 395-400.

Kabat, E., Wu, H.M., Perry, T.T., Gottesman, K.S., Foeller, C. 1991. Sequence of

Proteins of Immunological Interest. US Department of Health and Human Services,

U.S. Public Health Service, NIH Bethesda, MD. Publication No. 91-3242.

Kang, A.S., Jones, T.M., Burton, D.R. 1991. Antibody redesign by chain shuffling from

random combinatorial immunoglobulin libraries. Proc.Natl.Acad.Sci. USA. 1991;

88: 11120-11123.

Kang, Z., Buchenauer, H. 1999. Immunocytochemical localization of Fusarium toxins in

infected what spikes by Fusarium culmorum. Physiol. Molec. Plant Pathol. 55. 275-

288.

Kang, Z. Buchenauer, H. 2000. Cytology and ultrastructure of the infection of wheat

spikes by Fusarium culmorum. Mycol. Res. 104: 1083-1093.

Kempf, H.J. Wolf, G. 1989. Erwina herbicola as a biocontrol agent of Fusarium

culmorum and Puccinia recondita f. sp. Tritici on wheat. Phytopathol, 79: 990–994.

Khonga, E.B., Sutton, J.C. 1986. Survival and inoculum production by Gibberella zeae

in wheat and corn residues. Can. J. Plant Pathol. 8: 351. (Abstr.).

Khonga, E.B., Sutton, J.C. 1988. Inoculum production and survival of Gibberella zeae

in maize and wheat residues. Can. J. Plant Pathol. 10: 232-239.

Page 200: PhD Fusarium

177

Khoudi, H., Laberge, S., Ferullo, J.M., Bazin, R., Darveau, A., Castonguay, Y., Allard,

G., Lemieux, R., Vézina, L.-P. 1999. Production of a diagnostic monoclonal

antibody. In perennial alfalfa plants. Biotech. Bioeng. 64: 135-143.

Kieke, M.C., Cho, B.K., Boder, E.T., Kranz, D.M., Wittrup, K.D. 1997. Isolation of

anti-T cell receptor scFv mutants by yeast surface display. Prot. Eng. 10: 1303-1310.

Kimura, M., Kaneko, I., Komiyama, M., Takatsuki, Koshino, H., Yoneyama, K.,

Yamaguchi, I. 1998a. Trichothecene 3-O-acetyltransferase protects both the

producing organism and transformed yeast from related mycotoxins. J. Biol. Chem.

273: 1654-1661.

Kimura, M., Matsumoto, G., Shingu, Y., Yoneyama, K., Yamguchi, I. 1998b. The

mystery of the trichothecene 3-O-acetyltransferase gene. Analysis of the region

around Tri101 and characterization of its homolouge from Fusarium sporotrichioides.

FEBS Lett. 435: 163-168.

Kimura, M., Takahashi-Ando, N., Nishiuchi, T., Ohsato, S., Tokai, T., Ochiai, N.,

Fujimura, M., Kudo, T., Hamamoto, H., Yamaguchi, I. 2006. Molecular biology and

biotechnology for reduction of Fusarium mycotoxin contamination. Minireview.

Pest. Biochem. Physiol. 86: 117-123.

Knappik, A., Plückthun, A. 1995. Engineered turns of a recombinant antibody improve

is in vivo folding. Protein Eng. 8. 81-89.

Knappik, A., Ge, L., Honegger, A., Pack, P., Fischer, M., Wellnhofer, G., Hoess, A.,

Wölle, J., Plückthun, A., Virnekäs, B. 2000. Fully synthetic human combinatorial

antibody libraries (HuCAL) based on modular consensus frameworks and CDRs

randomized with trinucleotides. J. Mol. Biol. 296: 57-86.

Ko, K., Koprowski, H. 2005. Plant biopharming of monoclonal antibodies. Virus Res.

111: 93-100.

Köhler, G., Milstein, C. 1975. Continuous cultures of fused cells secreting antibody of

predefined specificity. Nature. 256: 495-497.

Komori, T., Imayama, T., Kato, N., Ishida, Y., Ueki, J., Komari, T. 2007. Current status

of binary vectors and superbinary vectors. Plant Physiol. 145: 1155-1160.

Korpimäki, T., Rosenberg, J., Virtanen, P., Lamminmäki, U., Tuomola, M., Saviranta, P.

2003. Further improvement of broad specificity hapten recognition with protein

engineering. Protein Eng. 16: 37-46.

Krebs, B., Rauchenberger, R., Reiffert, S., Rothe, C., Tesar, M., Thomassen, E., Cao, M.,

Dreier, T., Fischer, D., Höss, A., Inge, L., Knappik, A., Marget, M., Pack, P., Meng,

X.Q., Schier, R., Söhlemann, P., Winter, J., Wölle, J., Kretzschmar, T. 2001. High-

throughput generation and engineering of recombinant human antibodies. J. Immun.

Meth. 254: 67-84.

Page 201: PhD Fusarium

178

Kuiper-Goodman. T. 1994. Prevention of human mycotoxicoses through risk assessment

of risk management. pp. 439-469. In: Mycotoxins in Grain. Compounds other than

Aflatoxin. J.D. Miller and H.L. Trenholm. (eds.) Eagan Press, St. Paul, MN.

Ladenson, R.C., Crimmins, D.L., Landt, Y., Ladenson, J.H. 2006. Isolation and

characterization of a thermally stable recombinant anti-caffeine heavy-chain antibody

fragment. Anal. Chem. 2006; 78: 4501-4508.

Lamminmäki, U., Paupério, S., Westerlund-Karlsson, A., Karvinen, J., Virtanen, P.L.,

Lövgren, T., Saviranta, P. 1999. Expanding the conformational diversity by random

insertions to CDRH2 results in improved anti-estradiol antibodies. J. Mol. Biol. 291:

1999; 589-602.

Larrick, J.W., Danielsson, L., Brenner, C.A., Abrahamson, M., K.E. Fry, Borrebaeck,

C.A.K. 1989. Rapid cloning of rearranged immunoglobulin genes from human

hybridoma cells using mixed primers and the polymerase chain reaction. Biochem.

Biophys. Res. Comm. 1989; 160: 1250-1256.

Lewandowski, S. Bushnell, W.R. 2001. Development of Fusarium graminearum on

floret surfaces of field-grown barley. p. 128 in [Proc.] 2001 National Fusarium Head

Blight Forum, 8-10 December, 2001, Erlanger, KY.

Li, M. 2000. Applications of display technology in protein analysis. Nat. Biotech. 18:

1251-1256.

Li, Y., Lipschultz, C.A., Mohan, S., Smith-Gill, S.J. 2001. Mutations of an epitope hot-

spot residue alter rate limiting steps of antigen-antibody protein-protein associations.

Biochem. 40: 2011-2022.

Lin, H-.Y., Chiang, J-.W., Shih, D.Y-.C. 2001. Detection of genetically modified

soybeans by PCR method and immunoassay kits. J. Food Drug Anal. 9: 160-166.

Little, M., Breitling, F., Dübel, S., Fuchs, P., Braunagel, M., Seehaus, T., Klewinghaus, I.

1993. Universal antibody libraries on phage and bacteria. Year Immunol. 7: 50-55.

Little, M., Kipriyanov, S.M., Le Gall, F., Moldenhauer, G. 2000. Of mice and men:

hybridoma and recombinant antibodies. Immunol. Today. 21: 364-370.

Lo, B.K.C. 2004. Antibody Engineering. Methods and Protocols. Humana Press.

Totowa, N.J. 561 pp.

Ma, J.K-C., Hiat, A., Hein, M.B., Vine, N., Wang, F., Stabila, P., van Dollewerd, C.,

Mostov, K., Lehner, T. 1995. Generation and assembly of secretary antibodies in

plants. Science. 268: 716-719.

Ma, B.L., Subedi, K., Evenson, L., Stewart, G. 2005. Evaluation of detection methods

for genetically modified traits in genotypes resistant to European Corn Borer and

Herbicides. J Environ. Sci. Health B. 40: 633-644.

Page 202: PhD Fusarium

179

MacKenzie, C.R., Sharma, V., Brummell, D., Bilous, D., Dubuc, G., Sadowska, J.,

Young, N.M., Bundle, D.R., Narang, S.A. 1994. Effect of C lambda-C kappa domain

switching on Fab activity and yield in Escherichia coli: Synthesis and expression of

genes encoding two anti-carbohydrate Fabs. Bio/Tech. 12, 390-395.

Magan, N., Hope, R., Colleate, A., Baxter, E.S. 2002. Relationship between growth and

mycotoxin production by Fusarium species, biocides and environment. Eur. J. Plant

Pathol. 108: 685-690.

Makvandi-Nejad, S., McLean, M.D., Hirama, T., Almquist, K.C., MacKenzie, C.R., Hall,

J.C. 2005. Transgenic tobacco plants expressing a dimeric single-chain variable

fragment (scFv) antibody against Salmonella enterica serotype Paratyphi B.

Transgen. Res. 14: 785-792.

Maragos, C.M., Jolley, M.E., Nasir, M.S. 2002. Fluorescence polarization as a tool for

the determination of deoxynivalenol in wheat. Food Add. Cont. 19, 400-407.

Maragos, C.M., McCormick, S.P. 2000. Monoclonal antibodies for the mycotoxins

Deoxynivalenol and 3-Acetyl-Deoxynivalenol. Food Agric. Immun. 12, 181-192.

Marks, J.D., Hoogenboom, H.R., Bonnert, T.P., McCafferty, J., Griffiths, A.D., Winter,

G. 1991. By-passing immunization. Human antibodies from V-gene libraries

displayed on phage. J.Mol. Biol. 222: 581-597.

Mattanovich, D., Gasser, B., Hohenblum, H., Sauer, M. 2004. Stress in recombinant

protein producing yeasts. J. Immun. Meth. 2004; 113: 121-135.

Maynard, J., Georgiou, G., 2000. Antibody engineering. Annu. Rev. Biomed. Eng. 2,

339–376.

McCafferty, J., Griffiths, A.D., Winter, G., Chiswell, D.J. 1990. Phage antibodies:

filamentous phage displaying antibody variable domains. Nature. 348: 552-554.

McCormick, S.P. 2003. The Role of DON in Pathogenicity. pp. 165-183. In: Fusarium

Head Blight of Wheat and Barley. K.J. Leonard and W.R. Bushnell (eds.). Am.

Phytopath Society. St. Paul, MN.

McCormick, S.P. Alexander, N.J. 2002. Fusarium Tri8 Encodes a Trichothecene C-3

Esterase. App. Env. Micro. 68: 2959-2964.

McCormick, S.P., Alexander, N.J., Trapp, S.E., Hohn, T.M. 1999. Disruption of

TRI101, the gene encoding trichothecene 3-O-acetyltransferase, from Fusarium

sporotrichioides. App. Env. Micro. 65: 5252–5256.

McCormick, S.P., Hohn, T.M., Desjardins, A.E. 1996. Isolation and characterization of

Tri3, a gene encoding 15-O-Acetyltransferase from Fusarium sporotrichiodies. App.

Env. Micro. 62: 353-359.

Page 203: PhD Fusarium

180

McLaughlin, C.S., Vaughn, M.H., Campbell, J.M., Wei, C.M., Stafford, M.E., Hansin,

B.S. 1977. Inhibition of protein synthesis by trichothecenes. pp. 263-273. In:

Mycotoxins in Human and Animal Health. H.V. Rodricks, C.W. Hesseltine, M.A.

Mehlman (eds.). Pathotoxin Publishers, Park Forest South, IL, USA.

McLean, M.D., Almquist, K.C., Niu, Y., Kimmel, R., Lai, Z., Schreiber, J.R., Hall, J.C.

2007. A human anti-Pseudomonas aeruginosa serotype 06ad immunoglobulin G1

expressed in transgenic tobacco is capable of recruiting immune system effector

function in vitro. Antimicrobial Agents and Chemo. 51: 3322-3328.

McMullen, M.P. 2003. Impacts of Fusarium Head Blight on the North American

Agricultural Community: The Power of One Disease to Catapult Change. pp. 484-

503. In: Fusarium Head Blight of Wheat and Barley. K.J. Leonard and W.R.

Bushnell (eds.). Am. Phytopath Society. St. Paul, MN.

McMullen, M.P., Jones, R., Gallenberg, D. 1997. Scab of wheat and barley: a re-

emerging disease of devastating impact. Plant Disease. 81: 1340-1348.

Meier, A., Birzele, B., Oerke, E.C., Dehne, H.W. 2000. Impact of growth conditions on

the occurrence of Fusarium spp. and the mycotoxin content of wheat. Mycotoxin

Res. 16A: 12-15.

Menzies, J., Gilbert, J. 2003. Diseases of Wheat. pp. 94-128. In.: Diseases of Field

Crops in Canada. K.L. Bailey, B.D. Gossen, R.K. Gugel, R.A.A. Morall (eds). Can.

Phytopath. Soc. Univ. Exten. Press. Univ. of Sask.

Mesterházy, Á. 1995. Types and components of resistance to Fusarium head blight of

wheat. Plant Breeding. 114:377–386.

Mesterházy, Á. 1997. Methodology of resistance testing and breeding against Fusarium

head blight in wheat and results of this selection. Cereal Res. Comm. 25: 631-637.

Mesterházy, Á. 2002. Role of deoxynivalenol in aggressiveness of Fusarium

graminearum and F. culmorum and in resistance of Fusarium head blight. Eur. J.

Plant Pathol. 108: 675-684.

Mesterházy, Á. 2003a. Breeding Wheat for Fusarium Head Blight Resistance in Europe.

pp. 211-240 In: Fusarium Head Blight of Wheat and Barley. K.J. Leonard and W.R.

Bushnell (eds.). Am. Phytopath Society. St. Paul, MN.

Mesterházy, Á. 2003b. Control of Fusarium Head Blight of Wheat by Fungicides. pp.

363-380 In: Fusarium Head Blight of Wheat and Barley. K.J. Leonard and W.R.

Bushnell (eds.). Am. Phytopath Society. St. Paul, MN.

Miedaner, T. 2002. Breeding for resistance to Fusarium spp. in wheat. pp. 7-16 In:

Food safety of cereals: A chain-wide approach to reduce Fusarium mycotoxins. O.E.

Scholten; P. Ruckenbauer; A. Visconti; W.A. van Osenbruggen; A.P.M. den Nijs

(eds.). Mycotochain. http://www.mycotochain.org

Page 204: PhD Fusarium

181

Miller, J.D. 1994. Epidemiology of Fusarium diseases in cereals. pp. 19-36 In;

Mycotoxins in Grain – Compounds Other than Aflatoxin. J.D. Miller and H.L.

Trenholm (eds.). Eagan Press, St. Paul, MN.

Miller, J.D. 2008. Review: Mycotoxins in small grains and maize: Old problems, new

challenges. Food Addit. Cont. 25: 219-230.

Miller, J.D., Arnison, P.G. 1986. Degradation of deoxynivalenol by suspension cultures

of a Fusarium head blight resistant wheat cultivar Frontana. Can. J. Plant Path. 8.

147-150.

Miller, J.D., Young, J.C., Sampson, D.R. 1985. Deoxynivalenol and Fusarium head

blight resistance in spring cereals. Phytopathol. Z. 133: 359-367.

Milus, E.A., Parsons, C.E.. 1994. Evaluation of Foliar Fungicides for Controlling

Fusarium Head Blight of Wheat. Plant Disease. 78. 697-699.

Minshull, J., Stemmer, W.P.C. 1999. Protein evolution by molecular breeding. Curr.

Opin. Chem. Biol. 3: 284-290.

Mirocha, C.J. 1983. Effects of trichothecene mycotoxins on farm animals. pp. 177-194.

In: Trichothecenes – Chemical, Biological and Toxicological Aspects. Y. Ueno

(ed.). Elsevier Science Publishing Company, Inc., New York, NY.

Mirocha, C.J., Xie, W., Filho, E.R. 2003. Chemistry and Detection of Fusarium

Mycotoxins. pp. 144-164. In: Fusarium Head Blight of Wheat and Barley. K.J.

Leonard and W.R. Bushnell (eds.). Am. Phytopath Society. St. Paul, MN

Mirocha, C.J., Yu, H., Evans, C.K., Kolaczkowski, E.K., Dill-Macky, R. 1997.

Chemistry and physiology of deoxynivalenol pathogenesis. Cereal Res. Commun.

25: 309-314.

Mitterbauer, R., G. Adam. 2002. Saccharomyces cerevisae and Arabidopsis thaliana:

Useful model systems for the identification of molecular mechanisms involved in

resistance of plants to toxins. Eur. J. Plant Pathol. 108: 699-703.

Mitterbauer, R., Poppenberger, B., Raditsching, A., Lucyshyn, D., Lemmens, M., Glössl,

J., Adam, G. 2004. Toxin-dependent utilization of engineered ribosomal protein L3

limits trichothecene resistance in transgenic plants. Plant Biotech. J. 2: 329-340.

Miyazaki, C., Iba, Y., Yamada, Y., Takahashi, H., Sawada, J.-I., Kurosawa, Y. 1999.

Changes in the specificity of antibodies by site-specific mutagenesis followed by

random mutagenesis. Prot. Eng. 12: 407-415.

Moss, M.O., Thrane, U. 2004. Fusarium taxonomy with relation to trichothecene

formation. Toxicol. Lett. 153: 23-28

Mudge, A.M., Dill-Macky, R., Dong, Y., Gardiner, D.M., White, R.G., Manners, J.M.

2006. A role for the mycotoxin deoxynivalenol in stem colonization during crown rot

Page 205: PhD Fusarium

182

disease of wheat caused by Fusarium graminearum and Fusarium

pseudograminearum. Physiol. Molec. Plant Path. 69: 73-85.

Muehlbauer, G.J., Bushnell, W.R. 2003. Transgenic Approaches to Fusarium Head

Blight Resistance. pp. 318-362. In: Fusarium Head Blight of Wheat and Barley.

K.J. Leonard and W.R. Bushnell (eds.). Am. Phytopath Society. St. Paul, MN

Muhitch, M.J., McCormick, S.P., Alexander, N.J. Hohn, T.M. 2000. Transgenic

expression of Tri101 and PDR5 genes increases resistance of tobacco to the

phytotoxic effects of trichothecenes during seed germination. Plant Sci. 157: 201-

207.

Muyldermans, S., 2001. Single domain camel antibodies: Current Status. Rev. Mol.

Biotech. 74, 277-302.

Ness, J.E., Kim, S., Gottman, A., Pak, R., Krebber, A., Borchert, T.V., Govindarajan, S.,

Mundorff, E.C., Minshull, J. 2002. Synthetic shuffling expands functional protein

diversity by allowing amino acids to recombine independently. Nat. Biotech. 20:

1251-1255.

Ness, J.E., Welch, M., Giver, L., Bueno, M., Cherry, J.R., Borchert, T.V., Stemmer,

W.P., Minshull, J. 1999. DNA shuffling of subgenomic sequences of subtilisin. Nat.

Biotech. 17: 893-896.

Nguyen, V.K., Hamers, R., Wyns, L., Muyldermans, S. 1999. Loss of splice consensus

signal is responsible for the removal of the entire CH1 domain of the functional camel

IgG2 A heavy-chain antibodies. Mol. Immun. 36: 515-524.

Nguyen, V.K., Hamers, R., Wyns, L., Muyldermans, S. 2000. Camel heavy-chain

antibodies: Diverse germline VHH and specific mechanisms enlarge the antigen-

binding repertoire. EMBO J. 19: 921-930.

Nicholson, P., Simpson, D.R., Weston, G., Rezanoor, H.N., Lees, A.K., Parry, D.W.

Joyce, D. 1998. Detection and quantification of Fusarium culmorum and Fusarium

graminearum in cereals using PCR assays. Physiol. Mol. Plant Pathol. 53: 17-37.

Nicol, M.J., Lauren, D.R., Miles, C.O., Jones, W.T. 1993. Production of a Monoclonal

Antibody with Specificity for Deoxynivalenol, 3-Acetyldeoxynivalenol and 15-

Acetyldeoxynivalenol. Food Agric. Immun. 5: 199-209.

Nieba, L., Honegger, A., Krebber, C., Plückthun, A. 1997. Disrupting the hydrophobic

paths at the antibody variable/constant domain interface: improved in vivo folding

and physical characterization of an engineered scFv fragment. Prot. Eng. 1997; 10:

435-444.

Nishiuchi, T., Masuda, D., Nakashita, H., Ichimura, K., Shinozaki, K., Yoshida, S.,

Kimura, M., Yamaguchi, I., Yamaguchi, K. 2006. Fusarium phytotoxin

trichothecenes have an elicitor-like activity in Arabidopsis thaliana, but the activity

Page 206: PhD Fusarium

183

differed significantly among their molecular species. Mol. Plant Microbe. Interact.

19: 512-520.

Nissim, A., Hoogenboom, H.R., Jones, P.T., Winter, G. 1994. Antibody fragments from

a „single pot‟ phage display library as immunochemical reagents. EMBO J. 1994; 13:

692-698.

Nölke, G., Fischer, R., Schillberg, S. 2006. Review: Antibody-based metabolic

engineering in plants. J. Biotechnol. 124: 271-283.

Nyyssönen, E., Keränen, S. 1995. V Multiple roles of the cellulase CBHI in enhancing

production of fusion antibodies by the filamentous fungus Trichoderma reesei. Curr.

Genet. 28: 71-79.

Nyyssönen, E., Penttilä, M., Harkki, A., Saloheimo, A., Knowles, J.K.C., Keränen, S.

1993. Efficient production of antibody fragments by the filamentous fungus

Trichoderma reesei. Bio/Tech. 11: 591-595.

Ohtsubo, K. 1983. Chronic toxicity of trichothecenes. pp. 171-176. In: Trichothecenes

– Chemical, Biological and Toxicological Aspects. Y. Ueno (ed.). Elsevier Science

Publishing Company, Inc., New York, NY.

Olea-Popelka, F. C., McLean, M. D., Horsman, J., Almquist, K., Brandle, J. E., Hall, J.

C. 2005. Increasing expression of an anti-picloram single-chain variable fragment

(scFv) Ab and resistance to picloram in transgenic tobacco (Nicotiana tabacum). J.

Agric. Food Chem. 53: 6683-6690.

Omidfar, K., Rasaee, M.J., Kashanian, S., Paknejad, M., Bathaie, Z. 2007. Studies of

thermostability in Camelus bactrianus (Bactrian camel) single-domain antibody

specific for the mutant epidermal-growth-factor receptor expressed by Pichia.

Biotechnol. Appl. Biochem. 46: 41-49.

Orlandi, R., Güssow, D.H., Jones, P.T., Winter, G. 1989. Cloning immunoglobulin

variable domains for expression by the polymerase chain reaction.

Proc.Natl.Acad.Sci. USA. 86: 3833-3837.

Orum H., Andersen, P.S., Oster, A., Johansen, L.K., Riise, E., Bjørnvad, M., Svendsen,

I., Engberg, J. 1993. Efficient method for constructing comprehensive murine Fab

antibody libraries displayed on phage. Nucleic Acids Res. 21(19): 4491-4498.

Otokawa, M. 1983. Immunological disorders. pp. 163-170. In: Trichothecenes –

Chemical, Biological and Toxicological Aspects. Y. Ueno (ed.). Elsevier Science

Publishing Company, Inc., New York, NY.

Outchkourov, N.S., Stiekema, W.J., Jongsma, Ma. 2002. Optimization of the expression

of equistatin in Pichia pastoris. Prot. Expr. Purif. 24: 18-24.

Page 207: PhD Fusarium

184

Owen, M., Gandecha, A., Cockburn, B., Whitelam, G. 1992. Synthesis of a functional

anti-phytochrome single-chain Fv protein in transgenic tobacco. Biotechnology 10:

790–794.

Packa, D. 1991. Cytogentic changes in plant cells as influenced by mycotoxins.

Mycotox. Res. 7: 150-155.

Parhami-Seren, B., Viswanathan, M., Margolies, M.N. 2002. Selection of high affinity

p-azophenylarsonate Fabs from heavy-chain CDR2 insertion libraries. J. Immun.

Meth. 259: 43-53.

Park, S.-G., Lee, J.-S., Je, E.-Y., Kim, I.-J., Chung, J.-H., Choi, I.-H. 2000. Affinity

maturation of natural antibody using a chain shuffling technique and the expression of

recombinant antibodies in Escherichia coli. Biochem. Biophys. Res. Comm. 275:

553-557.

Parry, D.W., Jenkinson, P, McLeod, L. 1995. Fusarium ear blight (scab) in small grain

cereals - a review. Plant Patholology 44: 207-238.

Paschke, M. 2006. Phage display systems and their applications. Appl. Microbiol.

Biotechnol. 70: 2-11.

Patten, P.A., Howard, R.J., Stemmer, W.P.C. 1997. Applications of DNA shuffling to

pharmaceuticals and vaccines. Curr. Opin. Biotech. 8: 724-733.

Paulitz, T.C., Fernando, W.G.D., Seaman, W.L. 1997. Susceptibility of wheat heads at

different growth stages when exposed to ascospores of Gibberella zeae. pp. 52-53 In:

[Proc.] 1997 National Fusarium Head Blight Forum, 10-12 November, 1997, St.

Paul, MN.

Peeters, K., De Wilde, C., De Jaeger, Angenon, G., Depicker, A. 2001. Production of

antibodies and antibody fragments in plants. Vaccine. 19: 2756-2761.

Peipp, M., Saul, D., Barbin, K., Bruenke, J., Zunino, S.J., Niederweis, M., Fey, G.H.

2004. Efficient eukaryotic expression of fluorescent scFv fusion proteins directed

against CD antigens for FACS applications. J. Immun. Meth. 285: 265-280.

Perkowski, J., Miedaner, T., Geiger, H.H., Müller, H.M., Chelkowski, J. 1995.

Occurrence of deoxynivalenol (DON), 3-acetyl-DON, zearalenone, and ergosterol in

winter rye inoculated with Fusarium culmorum. Cer. Chem. 72: 205-209.

Persson, M.A., Caothien, R.H., Burton, D.R. 1991. Generation of diverse high-affinity

human monoclonal antibodies by repertoire cloning. Proc.Natl.Acad.Sci. USA. 1991;

88: 2432-2436.

Peschen, D., Li, H.-P., Fischer, R., Kreuzaler, F., Liao, Y.-C. 2004. Fusion proteins

comprising a Fusarium-specific antibody linked to antifungal peptides protect plants

against a fungal pathogen. Nat. Biotech. 2004; 22: 732-738.

Page 208: PhD Fusarium

185

Pestka, J.J. 2008. Mechanisms of deoxynivalenol-induced gene expression and

apoptosis. Food Add. Cont. 25: 1128-1140.

Petrausch, U., Dernedde, J., Coelho, V., Panjideh, H., Frey, D., Fuchs, H., Thiel, E.,

Deckert, P.M. 2007. A33scFv-green fluorescent protein, a recombinant single-chain

fusion protein for tumor targeting. Protein Eng. Des. Sel. 20(12): 583-590.

Petruccelli, S., Otegui, M.S., Lareu, F., Tran Dinh, O., Fitchette, A.C., Circosta, A.,

Rumbo, M., Bardor, M., Carcamo, R., Gomord, V., Beachy, R.N. 2006. A KDEL-

tagged monoclonal antibody is efficiently retained in the endoplasmic reticulum in

leaves, but is both partially secreted and sorted to protein storage vacuoles in seeds.

Plant Biotech. J. 4: 511-27.

Plückthun, A., Skerra, A. 1989. Expression of functional antibody Fv and Fab fragments

in Escherchia coli. Methods Enzymol. 1989; 178: 497-515.

Poppenberger, B., Berthiller, F., Lucyshyn, D., Sieberer, T., Schuhmacher, R., Krska, R.,

Kuchler, K., Glössl, J., Luschnig, C., Adam, G. 2003. Detoxification of the

Fusarium mycotoxin deoxynivalenol by a UDP-gluco-syltransferase from

Arabidopsis thaliana. J. Biol. Chem. 278: 47905-47914.

Prelusky, D.B., Rotter, B.A., Rotter, R.G. 1994. Toxicology of mycotoxins. pp. 359-

403 In: Mycotoxins in Grains: Compounds Other than Aflatoxin. J.D. Miller and

H.L. Trenholm (eds.). Eagan Press, St. Paul. MN.

Pritsch, C., Muehlbauer, G.J., Bushnell, W.R., Somers, D.A., Vance, C.P. 2000. Fungal

development and induction of defense response genes during early infection of wheat

spikes by Fusarium graminearum. Mol. Plant Microbe Interact. 13: 159-169.

Proctor, R.H., Desjardins, A.E., McCormick, S.P., Plattner, R.D., Alexander, N.J.,

Brown, D.W. 2002. Genetic analysis of the role of trichothecene and fumonisin

mycotoxins in the virulence of Fusarium. Eur. J. Plant Pathol. 108: 691-698.

Proctor, R.H., Hohn, T.M., McCormick, S.P. 1995. Reduced virulence of Gibberella

zeae caused by disruption of a trichothecene toxin biosynthetic gene. Mol. Plant Mic.

Interac. 8: 593-601.

Proctor, R.H., Hohn, T.M., McCormick, S.P. 1997. Restoration of wild-type virulence to

Tri5 disruption mutants of Gibberella zeae via gene reversion and mutant

complementation. Microbiology. 143. 2583-2591.

Rahbarizadeh, F., Rasaee, M.J., Forouzandeh, M., Allameh, A.-A. 2006. Overexpression

of anti-MUC1 single-domain antibody fragments in the yeast Pichia pastoris. Molec.

Immun. 43: 426-443.

Ramirez, N., Rodriguez, M., Ayala, M., Cremata, J., Perez, M., Martinez, A., Linares, M.,

Hevia, Y., Paez, R., Valdes, R., Gavilondo, J.V., Selman-Housein, G. 2003.

Expression and characterization of an anti-(hepatitis B surface antigen) glycosylated

Page 209: PhD Fusarium

186

mouse antibody in transgenic tobacco (Nicotiana tabacum) plants and its use in the

immunopurification of its target antigen. Biotech. Appl. Biochem. 38: 223-230.

Reavy, B., Ziegler, A., Diplexcito, J., MacIntosh, S.R., Torrance, L., Mayo, M. 2000.

Expression of functional recombinant antibody molecules in insect cell expression

system. Prot. Exp. Purif. 18: 221-228.

Reiter, Y., Brinkman, U., Kreitman, R.J., Jung, S.H., Lee, B., Pastan, I. 1994.

Stabilization of the FV fragments in recombinant immunotoxins by disulfide bonds

engineered into conserved framework regions. Biochem. 33: 5451-5459.

Ribichich, K.F., Lopez, S.E., Vegetti, A.C. 2000. Histopathological spikelet changes

produced by Fusarium graminearum in susceptible and resistant wheat cultivars.

Plant Dis. 84: 794-802.

Roberts, R. W. 1999. Totally in vitro protein selection using mRNA-protein fusions and

ribosome display. Curr. Opin. Chem. Biol. 3: 268-273.

Roberts, S., Cheetham, J.C., Rees, A.R. 1987. Generation of an antibody with enhanced

affinity and specificity for its antigen by protein engineering. Nature 1987; 328: 731-

734.

Robinson, C.R., Sauer, S.T. 1998. Optimizing the stability of single-chain proteins by

linker length and composition mutagenesis. Proc. Natl. Acad. Sci. USA. 1998; 95:

5929-5934.

Rocha, O., Ansari, K., Doohan, F.M. 2005. Effects of trichothecene mycotoxins on

eukaryotic cells: a review. Food Addit. Contam. 22: 369-378.

Rotter, B.A., Thompson, B.K., Lessard, M., Trenholm, H.L., Tryphonas, H. 1994.

Influence of Low-Level Exposure to Fusarium Mycotoxins on Selected

Immunological and Hematological Parameters in Young Swine. Fund. Appl. Tox.

23: 117-124.

Rotter, B.A., Prelusky, D.B., Pestka, J.J. 1996. Invited Review: Toxicology of

deoxynivalenol (Vomitoxin). J. Tox. Env. Health. 48: 1-34.

Russel, M., Lowman, H.B., Clackson, T. 2004. Introduction to Phage Biology and Phage

Display. pp. 1-26. In: Phage Display A Practical Approach. Clackson, T., Lowman,

H.B., editors. Oxford Univ. Press. Oxford, UK.

Saalbach, I., Giersberg, M., Conrad, U. 2001. High-level expression of a single-chain Fv

fragment (scFv) antibody in transgenic pea seeds. J. Plant Physiol. 158: 529-533.

Saerens, D., Gassabeh, G.H., Muyldermans, S. 2008. Single-domain antibodies as

building blocks for novel therapeutics. Curr. Opin. Pharma. 8: 1-9.

Savard, M.E. 1991. Deoxynivalenol Fatty Acid and Glucoside Conjugates. J. Agric.

Food Chem. 39: 570-574.

Page 210: PhD Fusarium

187

Savard, M.E., Sinha, R.C., Seaman, W.L., Fedak, G. 2000. Sequential distribution of the

mycotoxin deoxynivalenol in wheat spikes after inoculation with Fusarium

graminearum. Canadian Journal of Plant Pathology. 22 280-285.

Schaafsma, A. W., Tamburic-Ilincic, L., Miller, J. D., Hooker, D. C. 2001. Agronomic

considerations for reducing deoxynivalenol in wheat grain. Can. J. Plant Pathol. 23:

279-285.

Schaffitzel, C., Hanes, J., Jermutus, L, Plückthun, A. 1999. Ribosome display: an in

vitro method for selection and evolution of antibodies from libraries. J. Immun.

Meth. 231: 119-135.

Schier, R., Marks, J.D., Wolf, E.J., Apell, G., Wong, C., McCartney, J.E., Bookman,

M.A., Huston, J.S., Houston, L.L., Weiner, L.M., Adams, G.P. 1995. In vitro and in

vivo characterization of a human anti-c-erbB-2 single-chain Fv isolated from a

filamentous phage antibody library. Immunotech. 1: 73-81.

Schier, R., Balint, R.F., McCall, A., Apell, G., Larrick, J.W., Marks, J.D. 1996a.

Identification of functional and structural amino-acid residues by parsimonious

mutagenesis. Gene. 169: 147-155.

Schier, R., Bye, J., Apell, G., McCall, A., Adams, G.P., Malmqvist, M., Weiner, L.M.,

Marks, J.D. 1996b. Isolation of high-affinity monomeric human anti-c-erbB-2 single

chain Fv using affinity-driven selection. J. Mol. Biol. 255: 28-43.

Schier, R., McCall, A., Adams, G.P., Marshall, K.W., Merritt, H., Yim, M., Crawford,

R.S., Weiner, L.M., Marks, C., Marks, J.D. 1996c. Isolation of picomolar affinity

anti-c-erbB-2 single-chain Fv by molecular evolution of the complementarity

determining regions in the center of the antibody binding site. J. Mol. Biol. 263:

551-567.

Schillberg, S., Fischer, R., Emans, N. 2003. Molecular farming of recombinant

antibodies in plants. Cell. Mol. Life Sci. 60: 433-445.

Schroeder, H.W., Christensen.J.J. 1963. Factors affecting resistance of wheat to scab

caused by Gibberella zeae. Phytopathol 53:831–838.

Schwadorf, K., Müller, H.M. 1991. Determination of trichothecenes in cereals by gas

chromatography with ion trap detection. Chromatographia. 32: 137-142.

Scott, P.M. 1991. Possibilities of reduction or elimination of mycotoxins present in

cereal grains. pp. 529-572. In: Cereal Grain Mycotoxins, Fungi and Quality in

Drying and Storage. J. Chelkowski (ed.). Elsevier, Amsterdam, NL.

Scott, P.M., Nelson, K., Kanhere, S.R., Karpinski, K.F., Hayward, S., Neish, G.A., Teich,

A.H. 1984. Decline in deoxynivalenol (vomitoxin) concentrations in 1983 Ontario

winter wheat before harvest. App. Env. Micro. 48: 884-886.

Page 211: PhD Fusarium

188

Seeland, T., Bushnell, W.R. 2001. Effects of deoxynivalenol on barley leaf

pigmentation. pp. 151-153. In: [Proc.] 2001 National Head Blight Forum. 8-10

December, 2001. Erlanger, KY.

Shaner, G. 2003. Epidemiology of Fusarium Head Blight of Small Grain Cereals in

North America. pp. 84-119 In: Fusarium Head Blight of Wheat and Barley. K.J.

Leonard and W.R. Bushnell (eds.). Am. Phytopath Society. St. Paul, MN.

Sheedy, C., MacKenzie, C.R., Hall, J.C. 2007. Isolation and affinity maturation of

hapten-specific antibodies. Biotech. Adv. 25: 333-352.

Sheedy, C., Yau, K.Y.F., Himara, T., MacKenzie, C.R., Hall, J.C., 2006. Selection,

characterization, and CDR shuffling of naïve Llama single-domain antibodies

selected against auxin and their cross-reactivity with auxinic herbicides from four

chemical families. J. Agric. Food Chem. 54, 3668-3678.

Sherman, F. 2000. Getting started with yeast. Methods Enzymol. 350: 3-41.

Shi, Z-X, Wang, D.-B. 1991. Inhibitory effect of Bacillus spp. as biocontrol agent for

Gibberella Zeae (Schw.) Petch. J. Huazhong Agric. Univ. 20. 112-113.

Simpson, D.R., Weston, G.E., Turner, J.A., Jennings, P., Nicholson, P. 2001.

Differential control of head blight pathogens of wheat by fungicides and

consequences for mycotoxin contamination of grain. Eur. J. Plant Path. 107, 421-

431.

Sinclair, G., Choy, F.Y. 2002. Synonymous codon usage bias and the expression of

human glucocerebrosidase in the methylotrophic yeast Pichia pastoris. Prot. Expr.

Purif. 26: 96-105.

Sinha, R.C., Savard, M.E. 1997. Concentration of deoxynivalenol in single kernels and

various tissues of wheat heads. Can. J. Plant Pathol. 19: 8-12.

Sinha, R.C., Savard, M.E., Lau, R., 1995. Production of monoclonal antibodies for the

specific detection of deoxynivalenol and 15-acetyldeoxynivalenol by ELISA. J. Agric.

Food Chem. 43, 1740-1744.

Skerra, A., Plückthun, A. 1988. Assembly of a functional immunoglobulin Fv fragment

in Escherichia coli. Science. 240: 1038-1041.

Smith, G.P. 1985. Filamentous fusion phage: novel expression vectors that display

cloned antigens on the virion surface. Science. 240: 1038-1041.

Smith, G.P. and V.A. Petrenko. 1997. Phage Display. Chem. Rev. 97: 391-410.

Snijders, C.H.A. 1990a. Fusarium head blight and mycotoxin contamination of wheat, a

review. Netherlands J. Plant Pathol. 96: 187-198.

Page 212: PhD Fusarium

189

Snijders, C.H.A. 1990b. Genetic variation for resistance to Fusarium head blight in

bread wheat. Euphytica. 50: 171-179.

Snijders, C.H.A. 2004. Resistance of wheat to Fusarium infection and trichothecene

formation. Toxicol. Lett. 153-37-46.

Snijders, C.H.A., Perkowski, J. 1990. Effects of head blight caused by Fusarium

culmorum on toxin content and weight of wheat kernels. Phytopathol. 80: 566-570.

Songsivilai S., Bye J.M., Marks J.D., Hughes-Jones N.C. 1990. Cloning and sequencing

of human lambda immunoglobulin genes by the polymerase chain reaction. Eur. J.

Immunol. 20: 2661-2666.

Spencer, G.F., Desjardins, A.E., Plattner, R.D. 1990. 5-(2-carboxyethyl)-6-hydroxy-7-

methoxybenzofuran, a fungal metabolite of xanthotoxin. Phytochem. 29: 2495-2497.

Spinelli, S., Frenken, L.G., Bourgeois, D., de Ron, L., Bos, W., Verrips, T., Anguille, C.,

Cambillau, C., Tegoni, M. 1996. The crystal structure of a llama heavy chain

variable domain. Nat. Struct. Biol. 9: 752-757.

Spinelli, S., Frenken, L., Hermans, P., Verrips, T., Brown, K., Tegoni, M., Cambillau, C.

2000. Camelid heavy-chain variable domains provide efficient combining sites to

haptens. Biochemistry. 39: 1217-1222.

Spinelli, S., Tegoni, M., Frenken, L., van Vliet, C., Cambillau, C. 2001. Lateral

Recognition of a dye hapten by a llama VHH domain. J. Mol. Biol. 311: 123-129.

Stave, J. 2002. Protein immunoassays methods for detection of biotech crops:

applications, limitations and practical considerations. J. AOAC Int. 85: 780-786.

Steffenson, B.J. 2003. Fusarium Head Blight of Barley: Impact, Epidemics,

Management, and Strategies for Identifying and Utilizing Genetic Resistance. pp.

241-295 In: Fusarium Head Blight of Wheat and Barley. K.J. Leonard and W.R.

Bushnell (eds.). Am. Phytopath Society. St. Paul, MN

Stemmer, W.P.C. 1994. DNA shuffling by random fragmentation and reassembly: in

vitro recombination for molecular evolution. Proc.Natl.Acad.Sci.USA 91: 10747-

10751.

Stemmer, W.P.C., Crameri, A., Ha, K.D., Brennan, T.M., Heyneker, H.L. 1995. Single-

step assembly of a gene and entire plasmid from large numbers of

oligodeoxyribonucleotides. Gene. 164: 49-53.

Stockwell, C.A., da Luz, W.C., Bergstrom, G.C. 2000. Identification of bioprotectants

for control of Gibberella zeae. pp. 114-117 In: [Proc.] 2000 National Fusarium Head

Blight Forum 10-12 December 2000, Erlanger, KY.

Stoger, E., Sack, M., Fischer, R., Christou, P. 2002. Plantibodies: applications,

advantages and bottlenecks. Curr. Opin. Biotech. 13: 161-166.

Page 213: PhD Fusarium

190

Stoger, E., Sack, M., Nicholson, L., Fischer, R., Christou, P. 2005. Recent progress in

plantibody technology. Curr. Pharm. Des. 11: 2439-2457.

Stoger, E., Vaquero, C., Torres, E., Sack, M., Nicholson, L., Drossard, J., Williams, S.,

Keen, D., Perrin, Y., Christou, P., Fischer, R. 2000. Cereal crops as viable

production and storage systems for pharmaceutical scFv antibodies. Plant Molec.

Biol. 42: 583-590.

Strachan, G., McElhiney, J., Drever, M.R., McIntosh, F., Lawton, L.A., Porter, A.J.R.

2002. Rapid selection of anti-hapten antibodies isolated from synthetic and semi-

synthetic antibody phage display libraries expressed in Escherichia coli. FEMS

Microbiol. Lett. 210: 257-261.

Sudakin, D.L. 2003. Trichothecenes in the environment: relevance to human health.

Tox. Lett. 143: 97-107.

Sudarshana, M.R., Roy, G., Falk, B.W. 2007. Methods for engineering resistance to

plant viruses. Meth. Mol. Biol. 354: 183-195.

Sutton, J.C. 1982. Epidemiology of wheat blight and maize ear rot caused by Fusarium

graminearum. Can. J. Plant Pathol. 4: 195-209.

Tamaki, H., Yun, C.-W., Mizutani, T., Tsuzuki, T., Takagi, Y., Shinozaki, M., Kodama,

Y., Shirahige, K., Kumagai, H. 2005. Glucose-dependent cell size is regulated by a

G protein-coupled receptor system in yeast Saccharomyces cerevisiae. Genes to

Cells. 10: 193-206.

Tan, W., Lam, P.H. 1999. Expression and purification of a secreted functional

mouse/human chimaeric antibody against bacterial endotoxin in baculovirus-infected

insect cells. Biotech. Appl. Biochem. 30: 59-64.

Tanha, J., Muruganandam, A., Stanimirovic, D. 2003. Phage display technology for

identifying specific antigens on brain endothelial cells. Meth. Mol. Med. 89: 435-449.

Tanha, J., Xu, P., Chen, Z., Ni, F., Kaplan, H., Narang, S.A., MacKenzie. 2001. Optimal

design features of camelized human single-domain antibody libraries. J. Biol. Chem.

276: 24774-24780.

Tavlodoraki, P., Benvenuto, E., Trinca, S., de Martinis, D., Galeffi, P. 1993. Transgenic

plants expressing a functional single-chain Fv antibody are specifically protected from

virus attack. Nature. 366: 469-472.

Teich, A.H., 1989. Epidemiology of wheat (Triticum aestivum L.) scab caused by

Fusarium spp. pp. 269-282. In: Fusarium - Mycotoxins, Taxonomy, and

Pathogenicity. J. Chelkowski (Ed.), Elsevier, Amsterdam, NL.

Tekauz, A.; McCallum, B., Gilbert, J. 2000. Review: Fusarium head blight of barley in

western Canada. Can. J. Plant Pathol. 22: 9-16.

Page 214: PhD Fusarium

191

Terao, K. 1983. Target organella of trichothecenes. pp. 147-162. In: Trichothecenes –

Chemical, Biological and Toxicological Aspects. Y. Ueno (ed.). Elsevier Science

Publishing Company, Inc., New York, NY.

Terpe, K. 2006. Overview of bacterial expression systems for heterologous protein

production: from molecular and biochemical fundamentals to commercial systems.

Appl Micro. Biotech. 72: 211-222.

Tomlinson, I.M., Walter, G., Marks, J.D., Llewelyn, M.B., Winter, G. 1992. The

repertoire of human germline VH sequences reveals about fifty groups of VH

segments with different hypervariable loops. J. Mol. Biol. 227: 776-798.

Tonegawa, S. 1983. Somatic generation of antibody diversity. Nature. 302: 575-581.

Trenholm, H.L., Charmley, L.L., Prelusky, D.B., Warner, R.M. 1991. Two physical

methods for the decontamination of four cereals contaminated with deoxynivalenol

and zearalenone. J. Agric. Food Chem. 39: 356-360.

Trenholm, H.L., Charmley, L.L., Prelusky, D.B., Warner, R.M. 1992. Washing

procedures using water or sodium carbonate for the decontamination of three cereals

contaminated with deoxynivalenol and zearalenone. J. Agric. Food Chem. 40: 2147-

2151.

Trenholm, H.L., Forster, B.C., Charmley, L.L., Thompson, B.K., Hartin, K.E., Coppock,

R.W., Albassam, M.A. 1994. Effects of feeding diets containing Fusarium

(naturally) contaminated wheat or pure deoxynivalenol (DON) in growing pigs. Can.

J. Anim. Sci. 74: 361-369.

Tripathi, L. 2005. Techniques for detecting genetically modified crops and products.

Afr. J. Biotech. 4: 1472-1479.

Turner, D.J., Ritter, M.A., George, A.J. 1997. Importance of the linker in expression of

single-chain Fv antibody fragments: optimisation of peptide sequence using phage

display technology. J.Immunol. Meth. 1997; 205: 43-54.

Tyson, C.B., Lord, P.G., Wheals, A.E. 1979. Dependency of size of Saccharomyces

cerevisiae cells on growth rate. J. Bacteriol. 138: 92-98.

Ueno, Y. 1983. General toxicology. pp. 135-146. In: Trichothecenes – Chemical,

Biological and Toxicological Aspects. Y. Ueno (ed.) Elsevier Science Pub. Co., Inc.,

New York, NY.

Ueno, Y. 1984. Toxicological features of T-2 toxin and related trichothecenes. Fundam.

Appl. Toxicol. 4: S124-S132.

Urban, M., S. Daniels, E. Mott, and K. Hammond-Kosack. 2002. Arabidopsis is

susceptible to the cereal ear blight fungal pathogens Fusarium graminearum and

Fusarium culmorum. The Plant Journal. 32: 961-973.

Page 215: PhD Fusarium

192

Urbanek, H. 1989. The role of cutinase and cell wall degrading enzymes produced by

Fusaria in pathogenesis. pp. 243-256 in: Fusarium: Mycotoxins; Taxonomy and

Pathogenicity. J. Chelkowski (ed.). Elsevier Sci. Pub., Amsterdam, NL.

Valjakka, J., Hemminki, A., Niemi, S., Soderlund, H., Takkinen, K., and Rouvinene, J.

2002. Crystal structure of an in vitro affinity- and specificity-matured anti-

testosterone Fab in complex with testosterone. J. Biol. Chem. 277: 44021-44027.

Van den Bulcke, M., De Schrijver, A., De Bernardi, D., Devos, Y., MbongoMbella, G.,

Casi, A.L., Moens, W., Sneyers, M. 2007. Detection of genetically modified plant

products by protein strip testing: an evaluation of real-life samples. Eur. Food Res.

Technol. 225: 49-57.

van der Linden, R., de Geus, B., Stok, W., Bos, W., van Wassenaar, D., Verrips, Th.,

Frenken, L., 2000. Induction of immune responses and molecular cloning of the

heavy chain antibody repertoire of Lama glama. J. Immun. Meth. 240, 185-195.

van der Linden, R.H.J., Frenken, L.G.J., De Geus, B., Harmsen, M.M., Ruuls, R.C.,

Stok, W., De Ron, L., Wilson, S., Davis, P., Verrips, C.T., 1999. Comparison of

physical properties of llama VHH antibody fragments and mouse monoclonal

antibodies. Biochim. Biophys. Acta. 1431, 37-46.

Vaughan, T.J., Williams, A.J., Pritchard, K., Osbourn, J.K., Pope, A.R., Earnshaw, J.C.,

McCafferty, J., Hodits, R.A., Wilton, J., Johnson, K.S. 1996. Human antibodies with

sub-nanomolar affinities isolated from a large non-immunized phage display library.

Nat Biotech. 14: 309-314.

Verch, T., Yusibov, V., Koprowski, H. 1998. Expression and assembly of a full-length

monoclonal antibody in plants using a plant virus vector. J. Immun. Meth. 220: 69-

75.

Verma, R., Boleti, E., George, A.J. 1998. Antibody engineering: comparision of

bacterial, yeast, insect and mammalian expression systems. J. Immun. Meth. 216:

165-181.

Visconti, A., De Girolamo, A. 2002. Fusarium mycotoxins in cereals: storage,

processing and decontamination. pp. 30-41. In: Food safety of cereals: A chain-wide

approach to reduce Fusarium mycotoxins. O.E. Scholten; P. Ruckenbauer; A.

Visconti; W.A. van Osenbruggen; and A.P.M. den Nijs. (eds.). Mycotochain.

http://www.mycotochain.org

Vu, K.B., Ghahroudi, M.A., Wyns, L., Muyldermans, S., 1997. Comparison of Llama

VH sequences from conventional and heavy chain antibodies. Mol. Immun. 34, 1121-

1131.

Wakulinski, W. 1989. Phytotoxicity of the secondary metabolites of fungi causing wheat

head fusariosis (head blight). Acta. Physiol. Plant. 11: 301-306.

Page 216: PhD Fusarium

193

Wang, S-.H., Du, X-.Y., Huang, Y-.M., Lin, D-.S., Hart, P.L., Wang, Z-.H. 2007.

Detection of deoxynivalenol based on a single-chain fragment variable of the

antideoxynivalenol antibody. FEMS Microbiol Lett. 272: 214-219.

Wang, S.-H., Du, X.-Y., Lin, L., Huang, Y.-M., Wang, Z.-H. 2008. Zearalenone (ZEA)

detection by a single chain fragment variable (scFv) antibody. World J. Microbiol.

Biotechnol. 24: 1681-1685.

Wang, Y-Z. 1997. Epidemiology and management of wheat scab in China. pp. 97-105.

in: Fusarium Head Scab: Global Status and Future Prospects. H.J. Dubin, L.

Gilchrist, J. Reeves, and A. McNab (eds.). CIMMYT, DF, Mexico.

Wang, Y.Z., Miller, J.D. 1987. Screening techniques and sources of resistance to

Fusarium head blight. pp. 239-250 in: Wheat Production Constraints in Tropical

Environments. D.A. Saunders (ed.) CIMMYT, Mexico.

Wang, Y.Z., Miller, J.D. 1988. Effect of Fusarium graminearum metabolites on wheat

tissue in relation to Fusarium head blight resistance. J. Phytopathol. 122: 118-125.

Weiss, Y., Shulman, A., Ben Shir, I., Keinan, E., Wolf, S. 2006. Herbicide-resistance

conferred by expression of a catalytic antibody in Arabidopsis thaliana. Nat. Biotech.

24: 713-717.

Whitlow, M., Bell, B.A., Feng, S.L., Filpula, D., Hardman, K.D., Hubert, S.L.,

Rolenence, M.L., Wood, J.F., Schott, M.E., Milenic, D.E. 1993. An improved linker

for single-chain FV with reduced aggregation and enhanced proteolytic stability. Prot.

Eng. 6: 989-995.

Wilcoxson, R.D., Kommedahl, T., Ozmon, E.A., Windels, C.E. 1988. Occurrence of

Fusarium species in scabby wheat from Minnesota and their pathogenicity to wheat.

Phytopathol. 78: 586-589.

Willats, W.G. 2002. Phage display: practicalities and prospects. Plant Mol. Biol. 50:

837-854.

Windels, C.E., 2000. Economic and social impacts of Fusarium head blight: Changing

farms and rural communities in the northern great plains. Phytopathol. 90: 17-21.

Winter, G. 1998. Synthetic human antibodies and a strategy for protein engineering.

FEBS Lett. 430: 92-94.

Winter, G., Griffiths, A.D., Hawkins, R.E., Hoogenboom, H.R. 1994. Making antibodies

by phage display technology. Annu. Rev. Immunol.; 12: 433-455.

Winter, G., Milstein, C. 1991. Man-made antibodies. Nature. 349: 293-299.

Woo, J.H., Liu, Y.Y., Mathias, A., Stavrou, S., Wang, Z., Thompson, J., Neville, Jr.,

D.M. 2002. Gene optimization is necessary to express a bivalent anti-human anti-T

cell immuotoxin in Pichia pastoris. Prot. Expr. Purif. 25: 270-282.

Page 217: PhD Fusarium

194

Wu, H., Beuerlein, G., Nie, Y., Smith, H., Lee, B.A., Hensler, M., Huse, W.D., Watkind,

J.D. 1998. Stepwise affinity maturation of vitaxin, an αvβ3–specific humanized mAb.

Proc. Natl. Acad. Sci.USA. 95: 6037-6042.

Wu, H., Nie, Y., Huse, W.D., Watkins, J.D. 1999. Humanization of a murine

monoclonal antibody by simultaneous optimization of framework and CDR residues.

J. Mol. Biol. 294: 151-162.

Yano, A., Maeda, F., Takekoshi, M. 2004. Transgenic tobacco cells producing the

human monoclonal antibody to Hepatitis B virus surface antigen. J. Med. Virol.

2004; 73: 208-215.

Yau, K.F.Y., Lee, H., Hall, J.C. 2003a. Emerging trends in the synthesis and

improvement of hapten-specific recombinant antibodies. Biotech. Advan. 21: 599-

637.

Yau, K.Y., Groves, M.A., Li, S., Sheedy, C., Lee, H., Tanha, J., MacKenzie, C.R.,

Jermutus, L., Hall, J.C. 2003b. Selection of hapten-specific single domain antibodies

from a non-immunized llama ribosome display library. J. Immun. Meth. 281, 161-

175.

Yuan, Q., Hu, W., Pestka, J.J., He, S.Y., Hart, L.P. 2000. Expression of a functional

antizearalenone single-chain Fv antibody in transgenic Arabidopsis plants. App. Env.

Micro. 66: 3499-3505.

Yuan, Q., Pestka, J.J., Hespenheide, B.M., Kuhn, L.A., Linz, J.E., Hart, L.P. 1999.

Identification of mimotope peptides which bind to the mycotoxin deoxynivalenol-

specific monoclonal antibody. App. Env. Micro. 65: 3279-3286.

Zhang, J., Tanha, J., Hirama, T., Khieu, N.H., To, R., Tong-Sevinc, H., Brisson, J.-R.,

MacKenzie, C.R., 2004. Pentamerization of single-domain antibodies from phage

libraries: A novel strategy for the rapid generation of high-avidity antibody reagents.

J. Mol. Biol. 335, 49-56.

Zhao, H., Giver, L., Shao, Z., Affholter, J.A., Arnold, F.H. 1998. Molecular evolution by

staggered extension process (StEP) in vitro recombination. Nat. Biotech. 16: 258-

261.

zu Putlitz, J., Kubasek, W.L., Duchêne, M., Marget, M., von Specht, B.U., Domdey, H.

1990. Antibody production in baculovirus-infected insect cells. Biotech. 8: 651-654.