231
CLONING, TRANSFORMATION AND EXPRESSION OF cDNA OF HUMAN scFv AGAINST HEPATOCELLULAR CARCINOMA IN INDICA RICE VARIETY (IR-64) A THESIS Submitted by AADARSH RS for the award of the degree of DOCTOR OF PHILOSOPHY DEPARTMENT OF INDUSTRIAL BIOTECHNOLOGY Dr.M.G.R EDUCATIONAL AND RESEARCH INSTITUTE UNIVERSITY (Declared u/s 3 of UGC Act.1956) CHENNAI 600 095 JUNE 2011

CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

CLONING, TRANSFORMATION AND EXPRESSION

OF cDNA OF HUMAN scFv AGAINST

HEPATOCELLULAR CARCINOMA IN INDICA RICE

VARIETY (IR-64)

A THESIS

Submitted by

AADARSH RS

for the award of the degree

of

DOCTOR OF PHILOSOPHY

DEPARTMENT OF INDUSTRIAL BIOTECHNOLOGY

Dr.M.G.R

EDUCATIONAL AND RESEARCH INSTITUTE

UNIVERSITY

(Declared u/s 3 of UGC Act.1956)

CHENNAI 600 095

JUNE 2011

Page 2: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ii

BONAFIDE CERTIFICATE

I certify that this thesis entitled “CLONING, TRANSFORMATION AND

EXPRESSION OF cDNA OF HUMAN scFv AGAINST HEPATOCELLULAR

CARCINOMA IN INDICA RICE VARIETY (IR-64)” is the bonafide work of

Mrs. AADARSH R S M.Sc., who carried out the research under my supervision.

Certified further, that to the best of my knowledge the work reported herein does not

form part of any other thesis or dissertation of the basis of which a degree or award

was conferred on an earlier occasion on this or any other candidate.

Dr. P. BALAKRISHNA MURTHY Ph.D., PDF, D.Sc.

Director-IIBAT

(Supervisor)

Dr. M. DEECARAMAN Ph.D., PDF

Dean of Industrial Biotechnology

Dr. M.G.R Educational and Research Institute

(Co- Supervisor)

Page 3: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

iii

AADARSH R S, M. Sc., Department of Biotechnology,

Research Scholar IIBAT,

Padappai – 601 301.

DECLARATION

This is to certify that the thesis titled, “CLONING, TRANSFORMATION AND

EXPRESSION OF cDNA OF HUMAN scFv AGAINST HEPATOCELLULAR

CARCINOMA IN INDICA RICE VARIETY (IR-64)” submitted by me to the Dr.

M.G.R. Educational and Research Institute University for the award of the degree of

Doctor of Philosophy is a Bonafide record of research work carried out by me under

the supervision of Dr. P. BALAKRISHNA MURTHY, Director, IIBAT and Dr. M.

DEECARAMAN, Dean, Department of Industrial Biotechnology, Dr. M.G.R

Educational and Research Institute, during the period from 2007 to 2011. The contents

of this thesis, in full or in parts, have not been submitted to any other Institute or

University for the award of any degree or diploma.

AADARSH RS

Page 4: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

iv

ABSTRACT

Page 5: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

v

ACKNOWLEGMENT

I thank the Almighty God, who has helped me to take another step in my life.

In every step of my life He has been and will be my rock, and my fortress, and my

deliverer, my rock, in whom I take refuge, my shield, and the horn of my salvation, my

stronghold.

My sincere respect and thanks to our Chancellor Thiru A.C. Shanmugan

B.A., B.L., and Pro-Chancellor Thiru A.C.S. Arunkumar, Dr. M.G.R. Educational and

Research Institute for providing me an opportunity to do research work. I also thank

Dr.P. Aravindan, Vice Chancellor and Dr. P.T. Manogaran, Vice President

(Academic) for sound advice and guidance.

It is difficult to overstate my sincere gratitude to my Ph.D. supervisor, Dr. P.

Balakrishna Murthy, Ph.D., PDF, D.Sc, Director, IIBAT, for providing facility and

support to carry out my research work. With his enthusiasm, his inspiration, and his

great efforts to explain things clearly and simply, he helped to make research work

exciting for me. Throughout my research period, he provided encouragement, sound

advice, good teaching, good company, and lots of good ideas. I would have been lost

without him.

Page 6: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

vi

I warmly thank my Co-Supervisor, Dr. M. Deecaraman Ph.D., PDF, Dean

of Industrial Biotechnology, Dr. M.G.R Educational and Research Institute for his

valuable advice and friendly help. His extensive discussions around my work and

interesting explorations on my research work have been very helpful.

I am deeply grateful to Dr. R. Shridar, Head of the Department of

Biotechnology, IIBAT, for his detailed and constructive comments, and for his

important support throughout my research work.

I wish to express my warm and sincere thanks to Dr. Osmat Azzam

Jefferson (CAMBIA, Australia) for providing me with plasmid pCAMBIA 1301 for

cloning.

I am thankful to Dr. Tomasz Pniewski, (Institute of Plant Genetics, Polish

Academy of Sciences, Poland) for providing me Agrobacterium tumefaciens EHA105

strain for transformation.

I am thankful to Dr. K. Veluthambi (Department of plant genetics, MKU

University, India) for providing me with E. coli DH5α strain for cloning and for his

extensive suggestion about my research work, which have been very helpful.

I express my sincere thanks to Dr. A. Ramesh, Head of the Department of

Analytical chemistry, IIBAT, for his constant encouragement and support.

I wish to thank Dr. Rama Vaidyanathan and Dr. M. Vijaya Lakshmi, Head

of Industrial Biotechnology, Dr. M.G.R Educational and Research Institute for the

support and encouragement.

Page 7: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

vii

I wish to thank my friends and colleagues, especially Mrs. Deepa

Surendirababu and Mrs. Sathya Diwakar, for helping me get through the difficult

times, and for all the emotional support, entertainment, and caring they provided.

I greatly express my sincere gratitude for co-operation and valuable support

from my beloved husband G.K. Prasanna, my sweet daughter K.P. Shreenidhi, my

father A. Radhakrishnan, my mother D. Sujatha and other family members without

whom I would not have achieved anything.

AADARSH R S

Page 8: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

viii

TABLE OF CONTENTS

CHAPTERS TITLE

PAGE

NO

ABSTRACT iv

LIST OF TABLES xii

LIST OF FIGURES xiv

LIST OF SYMBOLS & ABBREVIATIONS Xix

1. INTRODUCTION 1

2. LITERATURE REVIEW 4

3. AIM AND OBJECTIVE OF THE WORK 31

4. MATERIALS AND METHODS 34

4.1. GENE CLONING USING VECTOR NTI SOFTWARE 35

4.2. SYNTHESIS OF HCC-scFv GENE-GENEART 40

Page 9: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ix

4.3. 4.3. TRANSFORMATION OF PLASMID pCAMBIA 1301 AND

pMA-T INTO E. coli DH5α AND SCREENING OF

TRANSFORMED E. coli DH5α STRAIN

41

4.4. ISOLATION OF PLASMID pCAMBIA 1301 AND pMA-T

from TRANSFORMED E. coli DH5α AND CONFIRMATION

BY GEL ELECTROPHORESIS

47

4.5. CLONING OF HCC-scFv FROM pMA-T PLASMID INTO

pCAMBIA 301

56

4.5.1. RESTRICTION DIGESTION OF pCAMBIA 1301 AND

pMA-T for CLONING

57

4.5.2. LIGATION OF HCC-scFv AND pCAMBIA 1301 61

4.6. TRANSFORMATION OF pCAMBIA 1301-scFv FROM E. coli

DH5α INTO Agrobacterium tumefaciens EHA105

70

4.7. CALLUS INDUCTION OF IR64 RICE VARIETY FOR

TRANSFORMATION

76

4.8. TRANSFORMATION OF pCAMBIA 1301-HCC-scFv FROM

Agrobacterium tumefaciens EHA 105 INTO CALLUS BY

Agrobacterium- MEDIATED TRANSFORMATION

80

4.9. EXTRACTION OF TOTAL GENOMIC DNA FROM PLANT

TISSUES FOR MOLECULAR ANALYSIS

90

Page 10: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

x

4.10. MOLECULAR ANALYSIS OF PUTATIVE TRANSFORMED

PLANTS BY POLYMERASE CHAIN REACTION (PCR)

97

5. RESULTS AND DISCUSSIONS 103

5.1. GENE CLONING USING VECTOR NTI SOFTWARE 104

5.2. SYNTHESIS OF HCC-scFv GENE-GENEART 122

5.3. TRANSFORMATION OF PLASMID pCAMBIA 1301 AND

pMA-T INTO E. coli DH5α AND SCREENING OF

TRANSFORMED E. coli DH5α STRAIN

134

5.4. ISOLATION OF PLASMID pCAMBIA 1301 AND pMA-T

from TRANSFORMED E. coli DH5α AND CONFIRMATION

BY GEL ELECTROPHORESIS

141

5.5. CLONING OF HCC-scFv FROM pMA-T PLASMID INTO

pCAMBIA 301

144

5.5.1. RESTRICTION DIGESTION OF pCAMBIA 1301 AND

pMA-T for CLONING

144

5.5.2. LIGATION OF HCC-scFv AND pCAMBIA 1301

146

5.6. TRANSFORMATION OF pCAMBIA 1301-scFv FROM E. coli

DH5α INTO Agrobacterium tumefaciens EHA105 149

Page 11: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xi

5.7. CALLUS INDUCTION OF IR64 RICE VARIETY FOR

TRANSFORMATION

152

5.8. TRANSFORMATION OF pCAMBIA 1301-HCC-scFv FROM

Agrobacterium tumefaciens EHA 105 INTO CALLUS BY

Agrobacterium- MEDIATED TRANSFORMATION

154

5.9. EXTRACTION OF TOTAL GENOMIC DNA FROM PLANT

TISSUES FOR MOLECULAR ANALYSIS

156

5.10. MOLECULAR ANALYSIS OF PUTATIVE

TRANSFORMED PLANTS BY POLYMERASE CHAIN

REACTION (PCR)

157

6. SUMMARY 159

7. CONCLUSION AND SCOPE OF FUTURE WORK 162

APPENDICES 1-3

REFERENCES

LIST OF PUBLICATIONS

Page 12: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xii

LIST OF TABLES

TABLE

No

TITLE

PAGE

No

Table

2.5.1

Plant expression systems used for recombinant protein

production

10

Table

2.6.8.1

Antibodies expressed in plant expression system 26-27

Table

4.5.1.1

Restriction Digestion set up: pCAMBIA 1301: Vector

DNA 59

Table

4.5.1.2

Restriction Digestion set up: pMA-T (HCC-scFv): Insert

DNA 59

Table

4.5.2.1

Setup of Ligation reaction 66

Table

4.5.2.2

Controls for Ligation setup. 67

Table

4.5.2.3

Controls kept for the cloning experiment. 59

Page 13: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xiii

Table

4.7.1

Chemical composition of MS 2, 4-D medium 78

Table

4.8.1

Chemical composition of AAM Medium 84

Table

4.8.2

Chemical composition of N6 Medium 85

Table

4.10.1

Reaction mixture for PCR setup 101

Table

5.3.1

Screening of the transformed E. coli containing

pCAMBIA 1301 on LB medium containing kanamycin. 135

Table

5.3.2

Screening of the transformed E. coli containing

pCAMBIA 1301 on LB medium containing -

kanamycin+X-gal +IPTG.

137

Table

5.3.3

Screening of the transformed E. coli containing pMA-T-

HCC-scFv on LB medium containing ampicillin. 139

Page 14: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xiv

LIST OF FIGURES

FIGURE No TITLE PAGE

No

Figure 2.6.2.1

Schematic representation of full length antibody and

partial antibody used for therapeutic antibody

production

12

Figure 2.6.6.1

N-glycans structure in plants and mammals. 19

Figure 2.6.7.1

Glycoengineering/ Humanization of plantibodies 21

Figure 4.5.1

Illustration of cloning HCC-scFv gene into pCAMBIA

1301.

56

Figure 4.6.1

Illustration of transformation of pCAMBIA 1301-

HCC-scFv into A.tumefaciens EHA 105 by triparental

mating

71

Figure 4.8.1

Illustration of transformation of pCAMBIA 1301-

HCC-scFv into IR 64.

81

Page 15: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xv

Figure 5.1.1

Analysis of Gene sequence of HCC-scFv: AY686498

downloaded from the Gen Bank using vector NTI

software

106

Figure 5.1.2

Analysis of Gene Map of HCC-scFv: AY686498

downloaded from the Gen Bank using vector NTI

software

106

Figure 5.1.3

Modified Gene sequence of HCC-scFv: AY686498

downloaded from the Gen Bank using vector NTI

software

107

Figure 5.1.4

Modified Gene Map of HCC-scFv: AY686498

downloaded from the Gen Bank using vector NTI

software

107

Figure 5.1.5

Analysis of Gene sequence of pCAMBIA 1301

downloaded from the Gen Bank using vector NTI

software

113

Figure 5.1.6

Analysis of Gene Map of pCAMBIA 1301 downloaded

from the Gen Bank using vector NTI software

114

Figure 5.1.7

Gene Map: Cloning of pCAMBIA 1301 and HCC-scFv

to form pCAMBIA 1301-HCC-scFv using vector NTI

software

115

Page 16: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xvi

Figure 5.1.8

Gene Sequence: Cloning of pCAMBIA 1301 and HCC-

scFv to form pCAMBIA 1301-HCC-scFv using vector

NTI software

121

Figure 5.2.1

Plot showing G+C content 123

Figure 5.2.3

Gene sequenced from the synthesized gene. 129

`

Figure 5.2.4

Sequence identity of the synthesized gene sequence

and the given sequence.

131

Figure 5.2.5

Gene sequence of HCC-scFv gene cloned into plasmid

pMA-T (pMA-T-HCC-scFv) with NcoI and PmlI

restriction site.

132

Figure 5.2.6

Gene Map of pMA-T containing HCC-scFv with

various restriction sites.

133

Figure 5.3.1

Transformation of pCAMBIA1301 into E. coli DH5α

and screening of the transformed cells on LB medium

containing Kanamycin antibiotic.

136

Figure 5.3.2

Screening of the E. coli DH5α transformed with

pCAMBIA1301 on LB medium containing

Kanamycin, X-gal and IPTG.

138

Figure 5.3.3

Transformation of pMA-T-HCC-scFv into E. coli 140

Page 17: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xvii

DH5α and screening of the transformed cells on LB

medium containing ampicillin antibiotic.

Figure 5.4.1

Plasmid isolated from transformed E. coli DH5α

containing pCAMBIA 1301.

142

Figure 5.4.2

Plasmid isolated from transformed E. coli DH5α

(pCAMBIA 1301 and pMA-T-HCC-scFv).

143

Figure 5.5.1

Restriction digestion of pCAMBIA 1301 and pMA-T

(HCC-scFv) with NcoI and PmlI restriction enzymes.

145

Figure 5.5.2.1

Transformation of the ligated pCAMBIA 1301 and

pMA-T (HCC-scFv) into E. coli DH5α and screened on

LB medium containing Kanamycin.

147

Figure 5.5.2.2

Confirmation of the transformed E. coli DH5α

containing ligated pCAMBIA 1301 and pMA-T (HCC-

scFv)

148

Figure 5.6.1 Transformation of pCAMBIA 1301-HCC-scFv into

A.tumefaciens EHA 105 by triparental mating and

screening on AB minimal medium containing

Kanamycin antibiotic

150

Figure 5.6.2

Transformation of pCAMBIA 1301-HCC-scFv into

A.tumefaciens EHA 105 by triparental mating. 151

Figure 5.7.1

Callus induction of IR4 using 2, 4 D-MS medium 153

Page 18: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xviii

Figure 5.8.1 Transformation of pCAMBIA 1301-HCC-scFv from

A.tumefaciens EHA 105 into IR64 and selection of the

transformed callus on Hygromycin.

155

Figure 5.10.1

Molecular analysis of the transformed plant (CaMV

35 S) by polymerase chain reaction

158

Page 19: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xix

LIST OF SYMBOLS & ABBREVIATIONS

µg/ml- micro gram per millilitre

2,4D - Dichlorophenoxy acetic acid

BiP- Binding protein

bp- Base pair

CaMV35S - Cauliflower mosaic virus 35S promoter

cDNA- Complementary Deoxyribonucleic acid

CTAB - Cetyltrimethylammonium bromide

DMF - dimethyl formamide

DNA - Deoxyribonucleic acid

EDTA - Ethylenediaminetetraacetic acid

ER- Endoplasmic reticulum

F (ab)’ 2 – Bivalent antigen binding fragment

Fab- Antigen binding fragment

Fc-Fragment constant

Fv- Fragment variable

g/L- gram per litre

g/mol- grams per mole

GRAS- Generally regarded as safe

HAMA- Human anti mouse antibody

HBV- Hepatitis B virus

HCC- Hepatocellular carcinoma

HCV- Hepatitis C virus

Page 20: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xx

HDEL - His-Asp-Glu-Leu (ER retention signal)

HIV- Human immunodeficiency virus

HSV- Herpes-simplex virus

IgA- Immunoglobulin A

IgD - Immunoglobulin D

IgE- Immunoglobulin E

IgG- Immunoglobulin G

IgM- Immunoglobulin M

IPTG (Iso propyl thiogalactoside or isopropyl beta-D-thiogalactopyranoside

Kb –kilo base pairs

KDEL - Lys-Asp-Glu-Leu (ER retention signal)

Kg/ha- kilogram per hectare

mg/Kg – milligram per kilogram

mg/L –milligram per litre

mg/ml- milligram per millilitre.

mM- millimolar

NAA- Naphthalene acetic acid

ng – nanogram

nm- nanometers

PSV- Protein storage vacuole

PVP - Polyvinylpyrrolidone

RNAi - Ribonucleic acid interference

scFv- Single chain variable fragment

SDS- sodium dodecyl sulfate

SEKDEL - Ser-Glu-Lys-Asp-Glu-Leu (ER retention signal)

sIgA/G - Secretory immunoglobulin A/G

Tm - melting temperature

TSP- Total soluble protein

Page 21: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxi

VH- Heavy chain variable domain

VL- Light chain variable domain

X-gal (5-Bromo-4-chloro-3-indolyl-β-D-galactoside)

FLW- Fresh leaf weight

mg/g- milligram per gram

µg/g – microgram per gram

Page 22: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxii

CHAPTER – 1

INTRODUCTION

Page 23: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxiii

CHAPTER 1

INTRODUCTION

Recombinant monoclonal antibodies are used for various therapeutic and

diagnostic purposes. Genetic engineering of plants for the expression of monoclonal

antibodies has led to the production of “plantibodies” (antibodies produced by plants)

in large scale (Conrad 1998, Kilpatrick 1995, Ma 1996, Fischer 1999, Smith 2000,

Stoger 2002). Genetic manipulation of eukaryotic or prokaryotic DNA by cloning and

transformation, of the known or modified antibody genes in order to produce novel

therapeutic recombinant antibody is of major significance. Plant is an effective

antibody expression system than bacterial and animal expression systems, as they are

cost effective, comparatively safe and easier to scale up antibody production in tons

rather than in kilograms per year (Fischer, 2000). Expression of full-length antibody

and single chain fragment variable (scFv) in plant expression system for cancer

diagnosis and immunotherapy is significant. The antibody-mediated tumor

immunotherapy has become critical in biotherapy and it is used for diagnosis of

cancer, metastasis, fine staging and decisions regarding therapeutic approaches

(Schneebaum, 2000).

Page 24: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxiv

Hepatocellular carcinoma (HCC) is the third leading cause of cancer death

and the fifth most common malignancy occurring worldwide. Understanding the HCC

biology with recent advances in biotechnology has led to the development of novel

molecular agents targeted for this malignancy. In cancer therapy, antibody fragments

can be applied in the construction of immunotoxins, gene delivery and as anticancer

intrabodies. Antibody fragments directed towards cancer antigens can also be fused

with various toxins such as cytotoxic proteins (Pai, 1998), radionuclides (Liu, 1998) or

drugs (Chari, 1998). The resulting immunotoxins specifically deliver these agents to

cancer antigen presenting cells. These cancer cells are killed when immunotoxins are

internalized. Tumor-specific scFv fused to interleukin-2 has been used for T-cell

mediated eradication of tumors (Thor, 2001). Engineering antibody gene sequence

specific for HCC by cloning and transforming the gene into plant chromosome by

Agrobacterium-mediated transformation is the key tool for HCC-plantibody

production. Producing antibody fragments against HCC in plants to yield higher

amounts of plantibodies is a challenging task. The present research work focuses on

cloning, transforming and expressing a humanized monoclonal antibody (scFv)

specific for HCC in IR-64 rice variety, which will have the capacity to bind

specifically to the receptors of HCC.

Page 25: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxv

CHAPTER – 2

LITERATURE

REVIEW

Page 26: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxvi

CHAPTER 2

LITERATURE REVIEW

2.1. PLANTIBODIES

Plant expression system is a cost effective and potential alternative for

antibody production than transgenic animals (Morrow, 2001). Transgenic plant cells

are capable of synthesizing, maturing and assembling the light and heavy polypeptide

chains of the antibody similar to mammalian antibody. A functionally active mouse

antibody was first expressed in tobacco plant wherein the light and heavy chains of the

antibody are correctly folded and assembled in late 1980’s and early 1990’s (Hiatt

Page 27: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxvii

1989, During 1990). Since the production of the first functional antibody in plants,

many antibodies and antibody fragments have been produced for therapeutic and

diagnostic purposes. Recombinant proteins can be targeted into specific subcellular

compartments or organelles by attaching the signal peptide and KDEL retention signal

which plays a major role in protein accumulation and retention in plants. Glyco-

engineering and humanization of N-glycans can be carried out by mutating the gene

sequence, RNAi interference, retaining the protein in ER, addition of galactose and

sialic acid residues to produce non immunogenic N-glycans. Possibilities of

humanization and glyco-engineering of the plantibodies make plant expression system

as an excellent bioreactor for unlimited production of clinically important antibodies.

The plantibodies are further modified by fusing with enzymes for prodrug therapy,

toxins for cancer, viruses for gene therapy and to target the ligands to specific cellular

targets. It is evident that transgenic plants offers number of advantages for antibody

production over humans, animals, microbes and transfected animals. This review

focuses on plantibody production including glycosylation, glyco-engineering and its

advancements.

2.2. HEPATOCELLULAR CARCINOMA

The incidence of HCC in Asia and Africa is 40 times higher than any other

region in the world due to endemic hepatitis B virus (HBV) infection (Bosch 2004,

Parkin 2001 and Pisani 2002). The major risk factors for HCC development have been

identified as chronic hepatitis B virus (HBV) infection, hepatitis C virus (HCV)

infection, aflatoxin B1 uptake and metabolic disorders, such as hemo-chromatosis and

α1-antitripsin deficiency. In western countries, the incidence of HCC has showed a

sharp increase, mainly because of the high prevalence of hepatitis C virus (HCV)

infection (Fattovich 2004). HCC cases are diagnosed at an early stage and are treated

by surgery (resection or liver transplantation) and loco regional procedures

Page 28: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxviii

(radiofrequency ablations). Patients diagnosed with early HCC can survive for 5 years.

However after loco regional treatment patients have a poor prognosis due to

underlying liver disease and lack of effective systemic treatment. Application of

monoclonal antibodies against tumor antigens for cancer diagnosis and

immunotherapy is of great importance.

2.3. MONOCLONAL ANTIBODY

Monoclonal antibodies are used as integral components of diagnostic kits

for both screening of asymptomatic individuals, to characterize the stage of tumors and

assessing the tumour response. The property of the monoclonal antibody is their

discrete specificity for a single specific antigenic epitope. A number of monoclonal

antibodies have been produced that are specific for tumor cell antigens. These

antibodies are used in the detection assays to screen the abnormal gene products. The

antibody molecules conjugated with radiolabeled substances provide an alternative

diagnostic tool. Though monoclonal antibodies have extraordinary potential in

diagnoses and treatment, the major disadvantage of using mouse-derived antibody is

the generation of human anti mouse antibody (HAMA) response against mouse-

derived antibody (Osbourn 2003, Krauss 2003). The murine monoclonal antibodies

have short half-life (Kim, 2005) in humans and in cancer patients. Immunological

reactions (Berger, 2002) like allergy (Dass, 2006), serum sickness (Sparrow, 2007)

and renal impairment (Mendez, 1997) have been reported with antibodies of non-

human origin. Genetically engineered antibody such as chimeric antibody or

humanized antibody has shown increased therapeutic efficacy and reduced

immunogenicity during clinical applications.

2.4. FULL LENGTH AND PARTIAL LENGTH ANTIBODY

Page 29: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxix

The natural immunoglobulin molecule is composed of two heavy and two

light chains. Each chain consists either of one variable and one constant region (light

chain) or one variable and several constant regions (heavy chain). The antigen binding

sites are located in the region of variable protein sequences called the Fv fragment.

Each Fv fragment consists of a heavy chain variable domain (VH) and a light chain

variable domain (VL). These two domains are responsible for specific attachment to

an antigen. Recent advances in recombinant technology have facilitated gene

manipulation, cloning and expression of antibody-encoding genes in various hosts

(Ma, 1998). Recombinant antibodies can be cloned and expressed as intact antibodies,

monovalent antigen binding fragment (Fab), variable-region fragment (Fv) (Hiatt,

1990) and scFv. All of these antibodies and antibody derivatives retain their binding

specificity to the antigenic epitope. One of the most popular antibody forms is scFv

(Ma, 1994). An antibody in scFv format consists of variable regions of heavy and light

chains, which are fused together by a linker sequence. The scFv has been

demonstrated to be particularly useful since it can be encoded by a single gene (which

warrants simple manipulation) and is expressed as a single-function polypeptide chain.

The scFv fragments have the advantage of better penetration into tissues and rapid

clearance than whole immunoglobulins.

2.5. PLANT EXPRESSION SYSTEM -RICE (Oryza sativa)

Selection of a crop is based on a number of factors including overall yield,

in situ protein stability during storage and transport, ease of purification and the cost of

regulatory oversight for the production of plantibodies. While tobacco is favoured

because of its high yield, proteins produced in the tobacco leaf tends to be unstable,

extraction must be carried out immediately to avoid the necessity for post harvest

desiccation or freezing, and processing must also be taken into account for potential

toxic alkaloids. In contrast, cereal seeds provide the ideal environment in which

proteins can accumulate stably and from a regulatory perspective it is generally

Page 30: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxx

regarded as safe (GRAS) (Sparrow, 2007). Cereal seeds have evolved for protein

storage, allowing the stable accumulation of recombinant proteins in the endosperm.

The endosperm tissue provides the appropriate biochemical environment for protein

accumulation by creating specialized storage compartments such as protein bodies and

storage vacuoles, which are derived from the secretory pathway. Mature cereal seeds

are desiccated, which reduces the exposure of stored proteins to non-enzymatic

hydrolysis and protease degradation. It has been shown that antibodies expressed in

cereal seeds remain stable for several years at room temperature with no detectable

loss of activity (Stoger, 2000).

Rice used for recombinant protein production, shares many advantages with

maize such as the well-established gene transfer technology, its GRAS status, high

grain yield, agricultural and processing infrastructure. Rice is self-pollinating, reduces

gene flow unlike maize. Another difference is that, the endosperm tissue accounts for

more than 90% of the total seed weight (Takaiwa, 2007) and two protein storage

systems are used, namely PB-I and PB-II which facilitate the accumulation of

recombinant proteins (Yamagata, 1986). A comparison of rice with tobacco showed

that rice actually had the higher yield per unit of biomass, although tobacco had the

highest overall yield due to the multiple cropping cycles per year (Stoger, 2002b).

Other crops used as plant expression system are shown in Table 2.5.1. The choice

between rice and maize eventually comes down to local preferences, while rice likely

to dominate in Asia and maize in Africa, Europe and North America. Rice has the

marginal advantage because of its completed genome sequence, which makes it

simpler to identify favourable new regulatory elements and factors affecting yield and

protein stability (Goff, 2002).

Page 31: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxxi

Page 32: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxxii

2.6. LITERATURE SURVEY ON PLANTIBODIES

2.6.1. In vivum production of antibodies

Page 33: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxxiii

An ideal production system must be able to carry out co-and post-

translational modifications, including signal peptide cleavage, pro-peptide processing,

protein folding, disulfide bond formation and glycosylation (Gomord, 2004).

Currently, none of the microbial or mammalian expression system satisfies all these

requirements. The proteins produced in prokaryotes are not properly folded or

processed to provide the desired biological activity. However, for simple therapeutic

proteins such as insulin, interferon or human growth hormones which do not require

folding or extensive post translational processing to be biologically active, microbial

expression system is the choice (Walsh, 2006). Eukaryotic expression systems like

yeast and mammalian cell cultures suffer from many disadvantages in addition to high

operating costs, difficulties in scaling up and potential contamination by viruses and

prions.

Considering the limitations of the prokaryotic and eukaryotic expression

systems and increasing demand for clinical antibodies, plant expression system has

emerged as a suitable alternative expression system with high recombinant protein

production, reproducible quality, efficient and low production cost. It has been well

documented that plant expression system has the capacity to produce high quality

mammalian antibodies which are safe and biologically active (Walsh 2006, Twyman

2003 and Gomord 2004). The production capacity of recombinant antibodies in plant

expression system is unlimited. Recombinant protein production up to 20 kg/ha were

obtained in a plant bioreactor regardless of the plant material (Khoudi 1999, Austin

1994).The usage of transgenic plants could be a solution to meet the rapid increasing

demand for therapeutic antibodies, although expression level of therapeutic proteins

are relatively low (Hiatt 1989, Ma 1995).

2.6.2. Full length and partial antibody produced in plants

Page 34: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxxiv

Antibodies, produced by the B lymphocytes of the blood plasma cells are

protein molecules that recognize and bind to specific epitopes (termed antigen). The

role of the antibodies is to neutralize and eliminate the antigens. Various classes of

serum antibodies are IgG, IgA, IgM, IgD and IgE. Schematic representation of full-

length and partial antibody is shown in Figure 2.6.2.1

Figure 2.6.2.1: Schematic representation of full length antibody and partial

antibody used for therapeutic antibody production

Full-length antibody bind bivalent antigen and has constant region (Fc-

Fragment constant), which involves in secondary effector mechanisms. CaroRx, the

sIgA/G derived from Guy’s 13 was the first full length plantibody expressed in

tobacco and tested in clinical trials (Ma 1998). Full length antibody production has

been reported in plants by several other groups (Hiatt 1989, Hiatt 1990, During 1990,

Page 35: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxxv

Ma 1994, Van Engelen 1994). It is also possible to express antibody fragments or

partial antibody rather than the full length antibody and still retain the antigen-binding

specificity (Smith 2000, Chadd 2001 and Fischer 2003). The antigen binding fragment

Fab, F (ab)’ 2 and scFv (single chain variable fragment) are partial antibodies which

can be used for therapeutic antibody production. The Fab and F (ab)’2 fragments

contain only the sequences distal to the hinge region and bind monovalent and bivalent

antigen respectively. The scFv fragments contain variable regions of the heavy and

light chains joined by a flexible peptide chain and bind monovalent antigen. These

fragments can often be effectively used as therapeutic proteins, since they show

increased penetration of target tissues, reduced immunogenicity and are rapidly

removed from the tissues. The scFv antibody specific for Stolbur Phytoplasma has

been expressed in plants to resist against mollicutes, which are wall less bacteria

infecting human, animals and plants (Peeters, 2001). Other derivatives of antibody

include bispecific scFv’s, which contain the antigen recognition elements of two

different antibodies and can bind to two different antigens and scFv-fusions, in which

additional functions are linked to the recombinant proteins (Fischer, 2003). Successful

expression of whole IgGs and smaller antibody fragments, such as Fab, scFv, and

diabody has been demonstrated in plants (Vaquero 2002, Fischer 1999 and Giddings

2000). In tobacco leaves, full length antibodies (IgG) have been expressed at levels of

0.35–1.3% of the total soluble protein (TSP) and scFv at levels between 0.01% - 6.8%.

2.6.3. Antibody gene transformation method

The most commonly used method for the transformation of antibody gene

into plant cell is by Agrobacterium-mediated transformation using Ti-plasmid or by

particle bombardment with DNA-coated gold particles (Hiei 1994). High efficiency

gene transformation has been reported by Agrobacterium tumefaciens (soil borne

bacterium) mediated transformation for the production of fertile and heritable

Page 36: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxxvi

transgenic plants (Hellens, 2000). Agrobacterium-mediated transformation has several

advantages, which includes higher transformation efficiency, ability to transfer large

piece of DNA with minimal rearrangement, integrate relatively low number of

transgenic copies and stable gene integration into plant cells. Dicotyledonous plants,

like tobacco, are mainly transformed using Agrobacterium tumefaciens. T-DNA

vectors derived from the tumor inducing (Ti) plasmid of A. tumefaciens are used for

gene transformation under the control of viral plant promoter, mostly the constitutive

cauliflower mosaic virus (CaMV35S) promoter. The antibody gene is cloned into T-

DNA region which is flanked by two 25 base pair long imperfect repeats. The T-DNA

transfers the antibody gene from Agrobacterium to the plant nucleus by the action of

virulence (vir) genes in the T-DNA vector (same gene cassette or a separate plasmid)

and subsequently integrates into the plant genome by non-homologous recombination.

Transformed cells are selected by antibiotic resistant markers and complete transgenic

plants are regenerated from the transformed calli (Alamillo, 2006). Rapid transient

antibody production in plants has also been performed using modified plant viral

vectors (Porta 1996, Kikkert 2005) with production levels up to 0.7 % of TSP.

Monocotyledonous plants are mostly transfected by particle gun (Larrick 2001).

Particle bombardment allows the simultaneous introduction of multiple gene

constructs coated on the gold particles into the plant cell by biolistic gun, thereby

expecting the recovery of transgenic lines expressing multimeric antibodies like

secretory immunoglobulin A (sIgA) (Hein, 1991).

Two different approaches have been employed to produce biologically

active whole antibody in plants, a) transformation of the heavy and light-chain genes

separately into plants, followed by cross-pollination by conventional breeding of the

two transgenic parents to yield F1 individuals carrying both the heavy and light chain

genes (Austin 1994, De Neve 1993 and Ma 1994) and b) co- transformation of the

heavy and light chain genes on a single expression cassette (During 1990, Voss 1995

Page 37: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxxvii

and Yoder 1994). Although co-expressing the heavy and light chain genes on a single

expression cassette may be less involved in cross-pollination strategy, a wide range of

novel antibodies can be produced by combining plant lines carrying different sets of

modifications in either the heavy or light chains (Ma 1994). The co-transformation

system is widely used in Agrobacterium- mediated transformation (Komari, 1996).

This technique is rapid and the problem of segregation in the progenic plants can be

avoided, but the promoter and the terminator genes need to be chosen carefully to

coordinate the expression of the two transgene. An expression level of 1.1% of total

protein was achieved by this technique (Van Engelen, 1994). Super-binary vectors

carrying two separate T-DNAs are used for co-transformation were the plant cell is

transformed with two separate DNAs, one incorporating a gene of interest and the

other containing the selectable marker gene (During, 1990). The efficiency of co-

transformation and the frequency of unlinked integration of the two T-DNAs into

plants were higher through Agrobacterium-mediated transformation method than a

direct gene delivery method.

2.6.4. Protein targeting into plant subcellular compartments

In plant based antibody production, the recombinant protein can be targeted

into specific subcellular compartments, a plant specific strategy to increase the yield

and simplify the purification step by using specific promoters. Recombinant proteins

are targeted to plant cell compartments (endoplasmic reticulum (ER), chloroplast,

vacuole and oil body) for efficient protein expression (Hellwig 2004, Gomord 1999).

High-biomass yield and accumulation of large amount of therapeutic proteins were

obtained when proteins are targeted into various subcellular compartments of the plant

cell.

2.6.4.1. Endoplasmic reticulum

Page 38: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxxviii

The endoplasmic reticulum (ER) compartment allows the entry of proteins

into the secretory pathway and ensures correct folding and assembly of the newly

synthesized proteins in plants and animals (Van Droogenbroeck, 2008). Large amount

of recombinant protein accumulates in the periplasmic space, implying direct transport

of the proteins from the ER to the periplasmic space between the plasma membrane

and the cell wall. Overproduction of recombinant scFv-Fc disturbs normal ER

retention and protein-sorting mechanisms in the secretory pathway, which is proven by

aberrant localization of the ER chaperones calreticulin and binding protein (BiP) and

the endogenous seed storage protein cruciferin in the periplasmic space (Streatfield,

2006). The recombinant proteins produced so far in plants have been secreted into the

apoplast or intercellular space (Hellwig 2004, Borisjuk 1999). Some recombinant

proteins targeted to the secretory pathway was secreted by the roots into the culture

medium and these proteins accumulated in the medium in higher concentration than in

root tissues (Gaume 2003, Ma 2003) which makes the purification process simple

(Komarnytsky 2006). Recently, active form of antibody has been produced in the roots

of transgenic tobacco (Drake 2003, Sojikul 2003).

2.6.4.2. Protein storage vacuole

Proteins can also be targeted to protein storage vacuole (PSV), an

intracellular organelle of seeds, leaves and roots, where the proteins are stored (Park

2005, Vitale 2005). The PSV of seeds exhibits higher pH value and lower hydrolytic

activity which makes the compartment attractive for recombinant protein accumulation

(Humphrey, 2002). The human lysozyme and human serum albumin have been

expressed in the PSV of rice (Yang 2006, Yang 2003 and Arcalis 2004) and wheat

endosperm (Van Rooijen, 1995), where the protein is biologically active and showed

good stability.

Page 39: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xxxix

2.6.4.3. Oilbodies

Targeting of proteins to oilseeds enables the expression of high level of

proteins and cost effective recovery of therapeutic proteins. The proteins are targeted

to oilbodies as oleosin fusions (Seon, 2002), the major protein at the periphery of the

oilbody membrane, anchored by their hydrophobic domain exposing their N and C

terminal ends to the cytoplasm. The proteins can be purified by simple purification

system, where the proteins are recovered from the oilbodies and other seed

components by liquid- liquid phase separation. This strategy was used to produce

antibodies from different oilseed plants (Staub, 2000).

2.6.4.4. Chloroplast

Expression of proteins in the chloroplast offers several advantages

including very high yield and low proteolytic activity. Plants contain as many as

hundreds of chloroplast in each cell. The transgene coding for the protein can be

introduced into the chloroplast by particle bombardment or homologous recombination

(Daniell, 2002), this stable transformtion allows the amplification of transgene copies

and high accumulation of the recombinant proteins (De Cosa 2001, Faye 2006, and

Daniell 2001). Full length antibodies were produced in transgenic tobacco

chloroplasts, which were able to fold the complex proteins with disulfide bridges

(Mayfield, 2003). Similarly functional antibodies have been produced in the

chloroplast of algae (Cabanes, 1999). Though chloroplast has advantages,

Page 40: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xl

unfortunately chloroplast does not have the capacity to glycosylate proteins which is

essential for therapeutic proteins.

2.6.5. Signal peptide and ER retention signal (KDEL)

Targeting the protein to a specific compartment like ER or vacuoles

requires the addition of signal peptide and ER retention signal (KDEL). The function

of the signal peptide is to lead the protein molecules into the ER lumen for further

processing. In the absence of signal peptide, transgenic plants fail to express light and

heavy chains of the antibody (Austin, 1994). Expression level of recombinant protein

up to 1.3% of total leaf protein has been reported when expressed with signal peptide

at the N- terminus of the gene sequence (Austin, 1994). Recently, it has been reported

that the C-terminal peptide could also affect the expression level of plantibody

production. Expression level of the plantibody increases when the endoplasmic

reticulum retention signal peptide (KDEL) is added to the C-terminus of the scFv (Ma,

1995). Protein retention in the ER is not seen, when the gene sequence is expressed in

the absence of KDEL sequence at C-terminus and signal peptide at N-terminus. The

antibody expression level reached up to 0.01 % of total soluble protein, when the

signal peptide is linked to only N-terminus and 0.2% of total soluble protein, when

KDEL sequence was linked to only C-terminus. Expression level up to 1% of total

soluble protein was obtained when the signal peptide and KDEL sequence were linked

to both N-terminus and C-terminus of the gene sequence (Sriraman, 2004). It was

shown that KDEL -mediated protein retention in the ER could strongly increase the

stability and consequently the protein yield (Verch 1998, Zeitlin 1998, Ramirez 2002,

Page 41: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xli

Wandelt 1992, Tabe 1995, Pueyo 1995, Pagny 2003, Triguero 2005, Takaiwa 2007).

In transgenic alfalfa, accumulation of vicilin, the pea vacuolar storage protein

increases to 100-fold by the addition of ER retention signal (KDEL) to its C-terminus

(Tabe, 1995) and similar results have been obtained in other plant expression systems

(Hood 2007, Gomord 1997). Similar accumulation of storage protein sporamin has

been observed when HDEL, another ER- retention signal was fused to the C-terminus

(Park, 2004).

2.6.6. Protein N-glycosylation in plants

The antibodies produced in plants are proteolytically matured and

glycosylated with high-mannose and biantennary complex type N-glycans (Bakker

2001, Farran 2002).The high mannose-type N-glycans (Man 5-Man9 glycans) have

similar structure as mammalian glycoproteins and are glycosylated at the same Asn

residues, but the complex-type N-glycans remains structurally different from

mammalian glycoproteins. Despite these differences in the structure of N-glycans,

antibodies produced in plants have similar antigen binding specificity as antibodies

produced in mammalian cells (Figure 2.6.6.1).

Page 42: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xlii

Figure 2.6.6.1: N-glycans structure in plants and mammals.

Two classes of N-glycans are synthesized from the same oligosaccharide

precursor attached to the nascent glycoproteins. A. High mannose type N-glycans is

formed from the precursor by glycosidases. The N-glycans structure remains similar in

plants and mammals and contains two N-acetylglucosamine and 5–9 mannosyl

residues. B. The complex type N-glycans are formed from the precursor by the action

of glycosyltransferases. The biantennary complex type N-glycans are structurally

different in plants and mammals. The proximal N-acetylglucosamine core is

substituted by α1, 3 fucose (plants) and α1, 6 fucose (mammals) and the β mannose

core is substituted by a bisecting β1,2 xylose (plants) and a bisecting β1,4 N-

acetylglucosamine (mammals). In addition β1,3 galactose and fucose α1,4 linked to

the terminal N-acetylglucosamine of plant N-glycans and sialic acid in mammals

Page 43: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xliii

instead of β 1,3 galactose.

N-glycosylation of heavy chain determines the binding specificity of the

antibodies and half-life in the bloodstream, which is not affected by the presence of

plant N-glycans instead of mammalian N-glycans (Khoudi 1999, Bouquin 2002 and

Elbers 2001). A mouse immunoglobulin G antibody (MGR48) was expressed in

transgenic tobacco (Nicotiana tabacum cv Samsun NN) to study N-linked

glycosylation. The antibodies isolated from young leaves had relatively high amount

of high-mannose glycans compared with antibodies from older leaves, which contain

more terminal N-acetylglucosamine. The relative amount of N-glycans without

terminal N-acetylglucosamine increased with leaf age (Gomord 2005). Recently

engineering the glycosylated protein to humanize the production of recombinant

protein has been developed to make it clinically useful.

2.6.7. Glyco-engineering and humanization of plantibodies

The addition of α (1, 3)-fucose and β (1, 2)-xylose residues during N-

glycosylation in plants is not seen in mammals, and also they lack terminal galactose

and sialic acid residues, which are found on many native human glycoproteins. The

fucose and xylose residues have been reported to be immunogenic in several mammals

including humans, but curiously not in mice and only after multiple exposures in rats

(Faye 2005, Strasser 2005). Although animal studies indicate the immunogenic impact

of plant-specific glycans, glyco-engineering of the recombinant proteins have been

developed to produce non immunogenic N-glycans by removing the plant-specific

glycans or otherwise ‘humanize’ the glycan profiles of recombinant human

glycoproteins to increase in vivo activity and longevity of therapeutic antibodies

(Figure 2.6.7.1). Various strategies are used to glyco-engineer and humanize

recombinant therapeutic proteins.

Page 44: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xliv

Figure 2.6.7.1: Glycoengineering/ Humanization of plantibodies

Methods to modify plantibodies for clinical therapy

2.6.7.1. Mutating amino acid residues

N-glycosylation can be prevented in the recombinant protein, by mutating

Asn or Ser/Thr residues to inactivate N-glycosylation site. In several plant expression

systems, the plant specific Golgi glycosyltransferases are inhibited by modifying the

enzymatic machinery of the Golgi apparatus to prevent the addition of glyco-epitopes

to recombinant proteins. In Arabidopsis mutants, plant derived glyco-epitopes were

eliminated by Insertional mutation (Downing 2006, Koprivova 2004) and in

Page 45: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xlv

Physcomitrella patens elimination of glyco-epitopes were shown by inactivating the

target gene (Cox 2006). Insertional mutation or gene inactivation to eliminate glyco-

epitopes is done either by knocking out β (1, 2)-xylosylation and α (1, 3)-fucosylation

related genes (Von Schaewen 1993, Frey 2009) and/or by modifying the processing of

glycan structures by adding new glycosyltransferase (Strasser 2008, Misaki 2003). In

Arabidopsis plants, knocking out of the genes coding for β (1, 2)-xylosyltransferase

(XylT) and α (1, 3)-fucosyltranferase (FucT) resulted in the production of N-glycans

with two β-N-acetyglucosamine residues but lacked β (1, 2)-linked xylose and α (1, 3)

linked fucose (Von Schaewen 1993).

2.6.7.2. RNA interference

RNA interference has been used to knock out α 1, 3-fucosyltransferase and

β 1, 2-xylosyltransferase in two plant expression systems, Lemna minor and Medicago

sativa (Sriraman 2004). In Limna minor, plant glycans are humanized by co-

expression of RNAi transcript designed to silence endogenous α (1, 3)-

fucosyltransferase and β (1, 2)-xylosyltransferase activities to produce MDX-060

mAb. The produced mAb has a single major N-glycan species without detectable

plant-specific N-glycans (Sriraman 2004). RNAi technology has also been used for

expression of anti-HIV monoclonal antibody 2G12, to down-regulate the endogenous

XylT and FucT genes in N. benthamiana (Wright 1998.)

2.6.7.3. Protein retention in ER

Plant glycans can be humanized by retaining the recombinant proteins in

ER were the glycans structure remains similar in plants and animals. Further

modification of the glycoprotein in the Golgi apparatus can be avoided by retaining the

recombinant glycoprotein in ER, where plant-specific oligosaccharides are added

Page 46: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xlvi

(Fujiyama 2009). The ER retention signal SEKDEL (Ser-Glu-Lys-Asp-Glu-Leu) or

KDEL (Lys-Asp-Glu-Leu) or HDEL (His-Asp-Glu-Leu) peptides are often used to

retain the protein in endoplasmic reticulum (Tabe 1995, Park 2004, Haq 1995, Ko

2003, Strasser 2004). It has also been shown in tobacco that, by addition of KDEL ER

retention signal at the C-terminal ends of the heavy and light chains contains

exclusively non immunogenic high-mannose type N-glycans (Petruccelli 2006,

Palacpac 1999).

2.6.7.4. Addition of galactose

Plants do not add terminal galactose to the proteins which leads to

immunological reactions (Bakker 2006). It has been demonstrated that the human β1,

4 galactosyltransferase, expressed in plant cells, transfers galactose residues onto the

terminal N-acetylglucosamine residues of plant N-glycans (Misaki 2003, Fujiyama

2001, Jarvis 2003 and Farran 2002). In N. tabacum, human β (1, 4)-

galactosyltranferase (GalT) gene was transferred to add terminal galactose to plant

processed antibody proteins. In another approach, the genes encoding N-terminal

domain of Arabidopsis thaliana xylosyltransferase and the catalytic domain of human

β (1, 4)-galactosyltransferase I were fused and the chimeric gene expressed in tobacco

plant showed high galactosylation of N-glycan and decline in plant-specific Xyl and

Fuc residues (Kelm 1997).

2.6.7.5. Addition of sialic acid

In the absence of sialic acid residues at the termini of N- glycans, the

circulating proteins get rapidly eliminated from the blood by interacting with

Page 47: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xlvii

galactose-specific receptors on the surface of hepatic cells. Lack of terminal sialic acid

in plant glycoproteins (Fujiyama 2009) makes the proteins short-lived and biologically

inactive (Shah 2003). The production of sialylated N glycans is feasible in plants as

shown in insect cells (Lerouge 1998). Though endogenous sialylated glycol-conjugates

have been found in the plants (Paccalet 2007), it has been found that Neu5Ac, the

major sialic acid present in humans is not synthesized in plants in detectable amounts

(Seveno 2004, Zeleny 2006, Wee 1998). Addition of sialic acid has been successfully

engineered in plants to produce biologically active recombinant proteins. In

Arabidopsis, addition of mammalian α-2, 6-sialtransferase led to the expression of

sialylated N glycans (Misaki 2006). Similarly, addition of human CMP-N-

acetylneuraminic acid synthetase and CMP sialic acid transporter was demonstrated in

tobacco suspension-cultured cells (Saint 2007) and addition of genes encoding

Neu5Ac lyase and Neu5Ac synthase (neuB2) was shown in BY2 cells as well as in

alfalfa (Seveno 2004). Engineering the gene coding for epimerase along with other

components in the same expression system would complete the sialation pathway in

plants (Stein 2001). From this it is clear that plant produced antibodies or proteins can

be glycoengineered or humanized for the production of glycosylated therapeutic

proteins without immunogenic glyco-epitopes.

2.6.8. Successfully produced plantibodies

Several human therapeutic antibodies have been produced in plants.

Currently most of the antibodies and antibody fragments are expressed in tobacco or

corn (Larrick 2001). There are more than 20 companies worldwide that have

developed antibodies in various plant expression systems. The most advanced product

is CaroRX™, produced in tobacco by Planet Biotech (MountainView, CA); a chimeric

secretory antibody drug that has been shown to reduce tooth decay (Yuan 2000).

Epicyte Pharmaceutical (San Diego, CA) is developing several plantibodies to treat

Page 48: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xlviii

inflammatory and infectious diseases. Antibody against herpes simplex virus was

initially produced in soya bean for pre-clinical trials and now in rice expression

systems (Stoger 2002). The other antibodies in Epicyte’s pipeline are being produced

in corn through collaborations with Dow and Dow AgroSciences (Indianapolis, IN).

Meristem Therapeutics (Clermont-Ferrand, France) and Goodwin Biotech (Plantation,

FL) are using tobacco and corn systems to produce therapeutic antibodies. Monsanto

Protein Technologies (St Louis, MO) have also genetically modified corn to produce

monoclonal antibodies.

Chimeric secretory IgG-IgA antibody against surface antigen of

streptococcus mutants, causative agent of tooth decay has been produced in plants and

tested in humans (Torres 1999). Antibody 2G12, one of human immunoglobulin G

(IgG) monoclonal antibody which has the ability to prevent HIV-1 infection in animal

models has been expressed in maize. A humanized antibody, anti-herpes-simplex

virus (HSV) antibody for HSV-2 protein has been produced in soybean, effective in

the prevention of vaginal HSV-2 transmission in a mouse model (Stoger 2000).

Several therapeutic monoclonal antibodies specific for tumor antigens have also been

produced in plants. Antibody against carcinoembryonic antigen (CEA), a tumor-

associated antigen, which is a surface glycoprotein, has been expressed in rice and

wheat (Stoger 2002, McCormick 1999). Antibodies for lymphoma treatment have been

produced in plants (Pujol 2005). Functionally active antibody against anti-hepatitis B

surface antigen has been produced in the seeds of transgenic tobacco plants (Stoger

2000). Other antibodies produced in plants were Co-17 A IgG, scFv for mycotoxin

(Zearalenone), MAK33 IgG1, Fab fragment, MAK33 ScFv, MAK33 Fab and 38C 13

ScFv (Gomord 2004). Several plantibodies have been produced in plant expression

system (Table 2.6.8.1).

Page 49: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xlix

Page 50: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

l

Page 51: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

li

Page 52: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lii

Secretory immunoglobulin A (sIgA), produced in transgenic tobacco in

1998 (Ma 1995, Ma 1998) is the most extensively studied plantibody and the first

successfully assembled and expressed antibody. At present, six different plant-derived

monoclonal antibodies are being tested and are under clinical trials (Bardor 2003).

CaroRX™ for the prevention of dental carries is in Phase II.

Single-chain Fv antibody fragments produced by viral vectors in tobacco

against non-Hodgkin’s Disease is in Phase I.

IgG (ICAM1) for the prevention of common cold (Phase I)

Antibody against cancer (Phase II)

Rhino RX for the treatment of respiratory syncytial disease (Phase I)

Anti-hepatitis B surface-antigen antibody received regulatory approval in

Cuba for large-scale plant-made antibody production

2.6.9. Current limitations of plantibodies

Though plant expression systems are excellent bioreactors for antibody

production, there are a few limitations like low yield of some therapeutic proteins,

non-mammalian glycosylation, immunogenicity and allergenicity of glycosylated plant

produced antibodies. One of the major limitation to use plant made pharmaceuticals

Page 53: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

liii

(PMP) is plant N-glycans harboring specific α (1, 3)-fucose and β (1, 2)-xylose

residues that differs from animal glycoproteins. These non-mammalian glyco-epitopes

have been attributed to elicit immune responses in humans (Jin 2006, Koprivova

2004). Hence controlling the N-glycosylation of plant-made pharmaceuticals remains a

prerequisite for their use in human therapy. To overcome the limitation of plant N-

glycosylation, strategies have been developed to humanize plant derived antibodies by

inhibiting the plant endogenous Golgi glycosyltransferase and/or adding new

glycosyltransferases from mammals (Farran 2002, Bruyns 1996, Streatfield 2006, Haq

1995, Strasser 2004, Von Schaewen 1993, Frey 2009, Strasser 2008) and by retaining

the recombinant glycoproteins in the endoplasmic reticulum (ER), the site where the

plants and animals share the same N-glycan modifications. Another major hurdle is

antibody purification from plants, which can be sorted out easily. A major concern

prevails for the presence of phenols in the plant, which alters the property of the

expressed protein dramatically and irreversibly, which can be removed by

ultrafiltration or diafiltration. Other concerns include secondary metabolites,

endotoxins and mycotoxins, which can be minimized by rapid processing and early

filtration. The removal of these constituents is an important aspect for progressing

towards the clinical trials of plantibodies, which is really not a major obstacle as

several antibodies have been purified to homogeneity for therapeutic use.

Contamination risk by viral pathogens and prions is no longer an issue, but the

regulatory guidelines demand for the screening of herbicide and pesticide residues.

The regulatory authorities are also concerned about the labeling and containment of

the transgenic crops expressing recombinant proteins to prevent the entry of transgene

into the food chain. Strategies for containment include marker selection, self

pollinating crops and male sterility are used.

2.7.0. Future prospects

Page 54: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

liv

Plants offer many theoretical advantages over microbial and mammalian

expression systems for the production of antibodies, in terms of cost, safety,

practicality and scalability. Current strategies to improve plant expression systems will

potentially result in increased yield and simplified down-stream processing of

therapeutic proteins. It is interesting to note that several strategies have been

developed to overcome these limitations of plant based antibody production.

Progresses in glyco-engineering, humanization of glycans and reduced heterogeneity

of plant made antibodies are currently making plants as one of the major expression

systems, particularly when large quantities of multimeric recombinant proteins are

required. Although some plant-derived antibody products have successfully completed

early phase clinical trials, several issues including regulatory issues must still be

resolved. A small number of plant derived antibodies have met the technological

challenges, cleared the regulatory hurdles and have approached commercialization.

Owing to the increasing demand of pharmaceutically important therapeutic and

diagnostic proteins, plant expression system will act as an excellent bioreactor for

unlimited antibody production.

Page 55: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lv

CHAPTER – 3

Page 56: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lvi

AIM AND OBJECTIVE

OF THE WORK

CHAPTER 3

AIM AND OBJECTIVE OF THE WORK

AIM

The aim of this research work is to clone, transform and express cDNA of

humanized monoclonal antibody (scFv) against HCC in IR-64 rice variety.

Page 57: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lvii

OBJECTIVES

To clone cDNA of scFv fragment gene against hepatocellular

carcinoma (HCC-scFv) using Vector NTI advance 11.5 software.

To synthesis gene (HCC-scFv) – GENEART.

To transform plasmid (pCAMBIA 1301 and pMA-T) into E. coli

DH5α, by calcium chloride transformation method and to screen the

transformed E. coli DH5α strains.

To isolate the plasmid pCAMBIA 1301 and pMA-T from

transformed E. coli DH5α and confirm by gel electrophoresis.

To clone HCC-scFv from pMA-T plasmid into pCAMBIA 1301

To transform pCAMBIA 1301-HCC-scFv from E.coli DH5α into

Agrobacterium tumefaciens EHA105.

To induce callus in IR64 rice variety for transformation.

To transform pCAMBIA 1301-HCC-scFv from Agrobacterium

tumefaciens EHA 105 into callus by Agrobacterium- mediated

transformation.

To extract total genomic DNA from plant tissues for Molecular

Analysis.

To confirm Putative transformed plants by molecular analysis using

Polymerase Chain Reaction (PCR).

Page 58: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lviii

CHAPTER – 4

Page 59: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lix

MATERIALS

AND

METHODS

CHAPTER 4

MATERIALS AND METHODS

Page 60: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lx

4.1. GENE CLONING USING VECTOR NTI SOFTWARE

4.1.1. OBJECTIVE

To clone cDNA of scFv fragment gene against hepatocellular carcinoma

(HCC-scFv) using Vector NTI advance 11.5 software.

4.1.2. PRINCIPLE

The Vector NTI software contains cloning Tools known as Clone2Seq*

which was designed to recombine two restriction fragments inserted into an

appropriately digested vector. The interface makes it easy to select molecules and

fragments for cloning, to modify 3’ and 5’ ends of the fragment for compatibility and

to create the required recombinant DNA, whether circular or linear. The DNA

fragment can be restricted with various restriction enzymes and the fragments can be

cloned using the software. The cloned fragments are confirmed by electrophoresis

using the software. All cloning functions are present in vector NTI Advance software

including graphical map creation and parent descendant lineage tracking. Any

individual search result can be opened in the Molecule Viewer and sent to Clone2Seq*

for rapid cloning experiment.

4.1.3. MATERIALS

Vector NTI software advance 11.5 version (Invitrogen).

Page 61: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxi

Gene sequence of anti-HCC scFv antibody (Gene bank) (ref: no: AY686498).

Gene sequence of the plasmid 1301(Gene bank) (ref: no: AF234297).

4.1.4 METHODS

4.1.4.1. Gene sequence of anti-HCC scFv antibody gene: AY686498

Cloning of the anti-HCC scFv antibody gene into plasmid vector

(pCAMBIA 1301) was initially carried out using vector NTI software advance 11.5

version. The gene sequence was downloaded from the Gen bank accession no:

AY686498 (Synthetic construct clone SLH-04-scfv anti-HCC scFv antibody gene).

The sequence of the gene is shown below.

Gene sequence of anti-HCC scFv antibody gene: AY686498

LOCUS AY686498 894 bp DNA linear SYN 26-JUL-2004

DEFINITION Synthetic construct clone SLH-04-scfv anti-HCC scFv antibody gene,

complete cds.

ACCESSION AY686498

VERSION AY686498.1 GI:50428757

KEYWORDS .

SOURCE synthetic construct

ORGANISM synthetic construct

other sequences; artificial sequences.

REFERENCE 1 (bases 1 to 894)

AUTHORS Yu,B., Ni,M. and Shen,G.

TITLE Sceening of special scFv which can ligate to HCC from a large naive

antibody library

JOURNAL Unpublished

REFERENCE 2 (bases 1 to 894)

AUTHORS Yu,B., Ni,M. and Shen,G.

TITLE Direct Submission

JOURNAL Submitted (15-JUL-2004) Tongji Medical College, Department of

Page 62: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxii

Immunology, Hangkong Road, Wuhan, Hubei 430030, China

FEATURES Location/Qualifiers

source 1..894

/organism="synthetic construct"

/mol_type="genomic DNA"

/db_xref="taxon:32630"

/clone="SLH-04"

CDS 1..894

/codon_start=1

/transl_table=11

/product="anti-HCC scFv antibody"

/protein_id="AAT77091.1"

/db_xref="GI:50428758"

/translation="MKYLLPTAAAGLLLLAAQPAMAQANLRESGPALVKPTQTLTLTC

TFSGFSLSTSGMCVSWIRQPPGKALEWLALIDWDDDKYYSTSLKTRLTISKDTSKNQV

VLTMTNMDPVDTAVYYCARHWPTSFDYWGQGTLVTVSSGGGGSGGGGSGGSALEIVMT

QTPLSSPVTLGQPASISFRSSQSLVHSDGNTYLSWLQQRPGQPPRLLIYKVSNRFSGG

PRQIQWQWGQGQIFTLKNQQGWKLRMSGFITARKLHNFRVGTFGPRDQAGNRRAAAHH

HHHHGAAEQKLISEEDLNGAA"

sig_peptide 1..60

misc_feature 415..462

/note="Region: linker peptide"

misc_feature 823..840

/note="Region: His-tag"

misc_feature 850..891

/note="Region: myc-tag"

ORIGIN

1 atgaaatacc tattgcctac ggcagccgct ggattgttat tactcgcggc ccagccggcc

61 atggcccagg ccaacttaag ggagtctggt cctgcgctgg tgaaacccac acagaccctc

121 acactgacct gcaccttctc tgggttttca ctcagcacta gtggaatgtg tgtgagctgg

181 atccgtcagc ccccagggaa ggccctggag tggcttgcac tcattgattg ggatgatgat

241 aaatactaca gcacatctct gaagaccagg ctcaccatct ccaaggacac ctccaaaaac

301 caggtggtcc ttacaatgac caacatggac cctgtggaca cggccgtgta ttactgtgca

361 agacattggc cgacgagttt tgactattgg ggccaaggta ccctggtcac cgtctcgagt

421 ggtggaggcg gttcaggcgg aggtggctct ggcggtagtg cacttgagat tgtgatgacc

481 cagactccac tctcctcgcc tgtcaccctt ggacagccgg cctccatctc cttcaggtct

541 agtcaaagcc tcgtacacag tgatggaaac acctacttga gttggcttca gcagaggcca

601 ggccagcctc caagactcct aatttataag gtttctaacc ggttctctgg gggtcccaga

661 cagattcagt ggcagtgggg gcagggacag attttcacac tgaaaaatca gcaggggtgg

721 aagctgagga tgtcggggtt tattactgca cgcaagctac acaattttcg cgttggtacg

781 ttcgggccaa gggaccaagc tggaaatcga cgtgcggccg cacatcatca tcaccatcac

841 ggggccgcag aacaaaaact catctcagaa gaggatctga atggggccgc ctag

Page 63: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxiii

4.1.4.2. Modification of Gene sequence of anti-HCC scFv antibody gene:

AY686498

The downloaded gene sequence was further modified to clone the gene into

pCAMBIA 1301 for plant transformation. The modified gene sequence is shown

below.

Gene sequence modification of anti-HCC scFv antibody gene: AY686498-

1 to 951 bp

1 ccatggatgc aggtgctgaa cacgatggtg aacaaacact tcttgtccct ttcggtcctc

61 atcgtcctca tcgtcctctc ctccaacttg acagccggca tggcccaggc caacttaagg

121 gagtctggtc ctgcgctggt gaaacccaca cagaccctca cactgacctg caccttctct

181 gggttttcac tcagcactag tggaatgtgt gtgagctgga tccgtcagcc cccagggaag

241 gccctggagt ggcttgcact cattgattgg gatgatgata aatactacag cacatctctg

301 aagaccaggc tcaccatctc caaggacacc tccaaaaacc aggtggtcct tacaatgacc

361 aacatggacc ctgtggacac ggccgtgtat tactgtgcaa gacattggcc gacgagtttt

421 gactattggg gccaaggtac cctggtcacc gtctcgagtg gtggaggcgg ttcaggcgga

481 ggtggctctg gcggtagtgc acttgagatt gtgatgaccc agactccact ctcctcgcct

541 gtcacccttg gacagccggc ctccatctcc ttcaggtcta gtcaaagcct cgtacacagt

601 gatggaaaca cctacttgag ttggcttcag cagaggccag gccagcctcc aagactccta

661 atttataagg tttctaaccg gttctctggg ggtcccagac agattcagtg gcagtggggg

721 cagggacaga ttttcacact gaaaaatcag caggggtgga agctgaggat gtcggggttt

781 attactgcac gcaagctaca caattttcgc gttggtacgt tcgggccaag ggaccaagct

841 ggaaatcgac gtgcggccgc acatcatcat caccatcacg gggccgcaga acaaaaactc

901 atctcagaag aggatctgaa tggggccgcc aaggatgagc tctagcacgt g

Modifications made to the sequence:

1. Adaptor for NcoI was attached at the 5’end (1st bp- 6

th bp) -ccatgg.

2. Rice α amylase signal peptide was attached at the 5’end (7th

bp-99th

bp).

3. Variable heavy and light chain with linker peptide from 100th

bp-930th

bp.

4. Kdel retention signal peptide was attached from 931th

bp-942th

bp.

5. Adaptor for PmlI was attached at the 3’end (946th

bp -951st bp)- cacgtg.

6. The 24th

code c was replaced by g –cga.

Page 64: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxiv

The gene sequence (single strand shown in the illustration) from 1to 6 and

23 to 28 base pair (bp) codes for NcoI restriction site and when restricted with NcoI

enzymes, it forms 2 fragments. Therfore the 24th

code c i.e. cca coding for theronine

was replaced by cga which also codes for the same amino acid theronine. This

sequence modification will help to clone the gene into pCAMBIA 1301 at NcoI and

PmlI, were the entire cDNA strand will remain unrestricted between 7th

bp to 945th

bp.

The rice α amylase signal peptide was added at 5’end (ATG CAG GTG

CTG AAC ACC ATG GTG AAC AAA CAC TTC TTG TCC CTT TCG GTC

CTC ATC GTC CTC ATC GTC CTC TCC TCC AAC TTG ACA GCC GGC

ATG) to target the protein into the Endoplasmic reticulam (ER) and KDEL retention

signal peptide at 3’ end (AAGGATGAGCTC) to retaining the protein in

Endoplasmic reticulum(ER).

The modified gene sequence was cloned into pCAMBIA 1301 T-DNA

plasmid vector sequence restricted with NcoI and PmlI restriction enzymes and

ligated. The cloned construct was further restricted with the same NcoI and PmlI

restriction enzymes and confirmed by running electrophoresis using the software. All

the steps were carried out in the software, prior to gene synthesis.

Page 65: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxv

4.2 SYNTHESIS OF HCC-scFv GENE-GENEART

4.2.1. OBJECTIVE

To synthesis modified AY686498: gene (HCC-scFv) sequence –

GENEART.

4.2.2. METHOD

The modified gene squence confirmed by the analysis of Vector NTI

software for cloning was sent for gene synthesis to GENEART, Germany. The

stability of the gene strand was analyzed with Gene Optimizer for G+C content. When

the G+C content was confirmed to be above 50%, the gene synthesis was initiated. The

synthesized gene was confirmed by gene sequencing and cloned into a standard

plasmid vector pMAT-T (3329 bp) containing ampicillin resistance marker with colE1

replicon, using Sfil and Sfil cloning sites. Plasmid containing the synthesized gene of

interest of 5 µg was delivered in lyophilized form.

Page 66: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxvi

4.3. TRANSFORMATION OF PLASMID pCAMBIA 1301 AND pMA-T INTO

E. coli DH5α AND SCREENING OF TRANSFORMED E. coli DH5α STRAIN

4.3.1. OBJECTIVE

To transform plasmid (pCAMBIA 1301 and pMA-T) into E. coli DH5α, by

calcium chloride transformation method and to screen the transformed E. coli DH5α

strains.

4.3.2. PRINCIPLE

Certain species of bacteria naturally take up the DNA at certain stage of

growth. Transformation is the uptake of naked DNA by an organism and competence

is the state of being able to take up the DNA. Certain bacterial species have natural

transformation system where bacterial cells acquire competence for DNA uptake.

Some species are not naturally competent at any stage of growth. In such cells,

competence can be artificially induced by treating them with calcium chloride (Dagert

1979, Hiroaki 1990).

The exact mechanism of plasmid DNA uptake by competent E.coli cells is

unknown. Unlike salts and small organic molecules such as glucose, DNA molecules

Page 67: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxvii

are too large to diffuse or transport through the cell membrane. Some bacteria possess

membrane proteins that recognize the DNA and facilitate the absorption of short DNA

sequences derived from the relative species. One hypothesis states that DNA molecule

pass through any of several hundred channels formed at adhesion zones, where the

outer and inner cell membranes are fused to pores in the bacterial cell wall.

The fact that the adhesion zones are only present in growing cells, in

consistent with the observation that the cells in logarithmic phase can render most

competence for plasmid uptake. However, acidic phosphates of the DNA helix are

negatively charged, as well as the phospholipid composition of the cell membrane and

the membrane pores are negatively charged. Thus electrostatic repletion between

anions may effectively block the movement of the DNA through the adhesion zones.

Analysis of this ionic interaction produces a possible hypothesis for DNA

uptake in bacteria. Treatment of the cells at 0°C crystallizes the fluid cell membrane,

stabilizing the distribution of charged phosphates. The cations in the transformation

solution (Ca++

,Mn++

,K+,Co

+++) forms complex with exposed phosphate groups,

shielding the negative charges. With this ionic shield in place, a plasmid molecule can

then move through the adhesion zone. Heat shock complements this chemical process

perhaps by creating a thermal imbalance on either side of the E.coli membrane that

physically helps to pump the DNA molecule through the adhesion zone.

The transformed plasmids are selected based on the presence of antibiotic

marker genes present on the plasmid. The transformed pCAMBIA 1301 was screened

based on kanamycin antibiotic and the pMA-T was screened based on ampicillin

antibiotic. The plasmid pCAMBIA 1301 was also screened for the presence of lacZ

gene.

4.3.3. MATERIALS

Page 68: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxviii

pCAMBIA 1301

pCAMBIA Legacy Vector Kit obtained from CAMBIA, Australia. For cloning,

pCAMBIA 1301was used as plasmid vector- Appendix 2.

HCC-scFv-pMA-T

The synthesized gene was inserted into pMA-T vector and supplied by GENEART in

lyophilized form (5µg of plasmid).

Recipient strain

E. coli DH5α obtained from Madurai Kamaraj University (MKU).

2X YT medium (200 ml) pH 7.0

Tryptone 3.2g

Yeast Extract 2.0g

Nacl 1.0g

Mgcl2.6H2o 0.8132g

LB medium (1000 ml) pH 7.2

Tryptone 10.0g

Yeast Extract 5.0g

Nacl 10.0g

Agar (1.5%) 15.0g

100mM Cacl2 (400ml)

Page 69: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxix

Weigh 5.88 g of Cacl2 (1M=147.01g/mol-Merck: 10238005001730) and dissolve in

350 ml of autoclaved millipore water. Make up the volume to 400ml. Autoclave and

store at 4°C.

X-gal (5-Bromo-4-chloro-3-indolyl-β-D-galactoside)- Sigma B4252

Stock: (20mg/ml): 20mg of X-gal was dissolved in DMF (dimethyl formamide). The

tube was wrapped in aluminum foil and stored at -20°C.

Working stock: X-gal was used in plates at a concentration of 40µg/ml: 2µl/ml of LB

media.

IPTG (Iso propyl thiogalactoside or isopropyl beta-D-thiogalactopyranoside)-

Sigma-11502

Stock: 0.1 M IPTG :( 1M=238.3g/mol): 0.2383 g of IPTG was dissolved in 10 ml of

autoclaved Millipore water and stored at -20◦C.

Working stock: IPTG was used in the concentration of (0.1mM): 1µl/ml of LB media.

Kanamycin

Stock: (100mg/ml)-Kanamycin of 100mg was dissolved in 1 ml of sterile autoclaved

Millipore water.

Working stock: (50µg/ml)

Ampicillin

Stock: (100mg/ml)-Ampicillin of 100mg was dissolved in 1ml of sterile autoclaved

Millipore water.

Working stock: (100 µg/ml)

4.3.4. METHOD

Page 70: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxx

4.3.4.1. Plasmid preparation of pCAMBIA 1301vector

The paper containing the plasmid was cut and placed in 0.5 ml centrifuge

tube pierced with a needle at the base, and placed over a 1.5 ml eppendorf tube. TE

buffer of 50µl was added and incubated at room temperature for 10 minutes. The

content was spinned down into 1.5 ml eppendorf tube and stored at -20◦C until

transformation. The concentration of plasmid was checked using Biophotometer

(Germany).

4.3.4.2. HCC-scFv-pMA-T vector

Lyophilized plasmid of 5µg was dissolved in 50 µl of sterile syringe water

and stored at -20◦C until transformation.

4.3.4.3. Competent cell preparation

A single colony of E. coli (DH5α) was inoculated into 2 ml of LB culture

broth and incubated (Scigenics, India) at 37°C for 10 hours to 12 hours overnight.

From the overnight culture, 500µl was inoculated into 50 ml of 2X YT medium and

incubated at 37°C in a rotary shaker until it reached an O.D of 0.5 to 0.6 at 600nm

(Biophotometer, Germany). To a sterile 50 ml polypropylene tube, 30 ml of the culture

was transferred and incubated in ice for 30 minutes. The culture was centrifuged at

5000 rpm at 4°C for 10 minutes. The supernatant was discarded and the pellet was

resuspended in a small volume of medium left behind. Finally the pellet was gently

resuspended in 30 ml of ice cold 100mM CaCl2 and incubated in ice for 30 minutes.

The resuspended cells were centrifuged at 5000 rpm for 10 minutes at 4°C. The

Page 71: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxi

supernatant was discarded and the pellet was resuspended gently in 3ml (1/10th

volume

of CaCl2) of ice cold 100mM CaCl2.The competent cells were stored in ice for at least

30 minutes before use.

4.3.4.4. Transformation of competent cells (E. coli)

Competent cells of 200 µl was aliquated into 2ml sterile eppendorf tubes

and incubated in ice (cut tips were used to aliquot the cells). Plasmid pCAMBIA 1301

1301 and pMA-T-HCC-scFv of 200ng was dispensed into the above eppendorf tubes

containing competent cells separately and incubated in the ice for 30 minutes. Heat

shock was given for 2 minutes at 42°C in water bath and then transferred and

incubated in ice for 10 minutes. The cells were transferred to 0.8 ml of LB media and

incubated at 37°C in a rotary shaker for 1 hour at 220rpm.The LB mixture was then

centrifuged at 10,000 rpm for 5 minutes and the supernatant was discarded.

From the pellet, 100µl of the cells were plated onto LB agar plate

containing antibiotics kanamycin (50µg/ml) for pCAMBIA 1301 and 200 µl of the

cells were plated onto LB agar plate containing Ampcillin (100 µg/ml) for pMA-T-

HCC-scFv. The plates were incubated overnight at 37°C. In case of pCAMBIA 1301,

the transformed colonies were further screened for the presence of blue colonies by

streaking the transformed cells onto LB medium containing X-gal (40µg/ml) and IPTG

(0.1mM).

Page 72: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxii

4.4. ISOLATION OF PLASMID pCAMBIA 1301 AND pMA-T FROM

TRANSFORMED E. coli DH5α AND CONFIRMATION BY GEL

ELECTROPHORESIS

4.4.1. OBJECTIVE

To isolate plasmid pCAMBIA 1301 and pMA-T from transformed E.

coli DH5α and to confirm by gel electrophoresis.

4.4.2. PRINCIPLE

A rapid method for making a small preparation of purified plasmid DNA

from bacterial culture as low as 1ml is called “miniprep”. The alkaline lysis method is

a modified method of Birnbroim and doly (1979) and Ish.horowwicz and burkes

Page 73: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxiii

(1981). In this method the cells are lysed and the DNA is denatured by high pH of

solution II. The Solution III returns the lysis mixture to the neutral pH. The plasmid

DNA which remains partially intact, renatures rapidly than the chromosomal DNA

fragments, which gets pelleted along with the denatured proteins. The RNA which

remains along with the plasmid in the supernatant is removed by RNase treatment. The

Plasmid can then be concentrated by the addition of sodium acetate salt and 95 %

ethanol. Based on the salting out principle the sodium acetate salt removes the water

molecule, which causes the double stranded plasmid DNA to aggregate and

precipitate.

4.4.2.1. Principle behind the reagents used

Solution I: It acts a buffer in which glucose increases the osmotic pressure of the cell,

EDTA removes the magnesium ions that are essential for preserving the overall

structure of the cell membrane and also inhibits the cellular enzymes that could

degrade DNA and Tris helps in maintaining the cytoplasmic pH of the cell.

Solution II: The alkanine mixture (SDS/NaOH) lysis the bacterial cells. The detergent

SDS dissolves the lipd components of the cell membrane, as well as the cellular

proteins. The NaOH denatures the chromosomal and plasmid DNA into single strands.

The intact circles of plasmid DNA remain intertwined.

Solution III: The acetic acid returns the pH to netural, allowing DNA strands to

renature. The large disrupted chromosomal strands cannot rehybridize perfectly, but

instead collapse into a partially hybridized tangle. At the same time, potassium acetate

precipitates the SDS from the cell suspension along with the protein and the lipids.

The renaturing chromosomal DNA is trapped in the SDS/ Lipid / Protein precipitate.

Only smaller plasmid DNA and RNA molecules escape the precipitate and remain in

the solution.

Page 74: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxiv

Isopropanol: The isopropyl alcohol rapidly precipitates the nucleic acid but slowly

precipitates the protein. The organic solvent content increases as the availability of the

water decreases. Thus the solubility of the DNA comes down and plasmid DNA gets

pelleted.

Tris / EDTA: Tris buffers the DNA solution, EDTA protects the DNA from

degradation by DNase by binding divalent cations that are necessary co factors for

DNase activity, buffering DNA is important as low pH(<6) leads to the loss of purines

(adenine and guanine) called depurination.

4.4.3. MATERIALS

Solution I: 100ml (pH8.0)

M.wt for 100ml

Tris (25mM) - 121.1 0.303g

EDTA (10mM) - 372.0 0.372g

Glucose (50mM) -180.16 0.901g

Page 75: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxv

The salts were weighed and dissolved in 80 ml of millipore water and the pH was

adjusted to 8.0 using 1 N HCl. The volume was made up to 100 ml. Autoclaved and

stored at room temperature.

Solution II: (Prepared fresh every time)

NaOH (0.2 M)

SDS (1.0 %)

NaOH (0.4 N) was prepared and stored in a plastic reagent bottle. SDS (2%) was

prepared and autoclaved. The solutions were mixed in the ratio of 1:1 before use.

(NaOH- was not autoclaved)

Solution III: 100ml (pH 5.5)

3 M potassium acetate

Glacial acetic acid

For 100 ml, 29.4 gm of potassium acetate was weighed and dissolved in 25 ml to 30

ml of autoclaved millipore water. The pH was adjusted with glacial acetic acid and the

volume was made up to 100ml. Autoclaved and stored at 4°C.

RNase-(Cat- RD0187)

Stock: 10mg/ml

Working stock concentration: 1 µl

RNase (Fermentas) was treated initially by heating to 100 °C for 15 minutes in boiling

water bath (to denature DNase) and allowed to cool to room temperature and stored at

-20°C.

TE(0.1X) pH 8.0

Tris HCl (1mM)

EDTA (0.1mM)

Page 76: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxvi

For 100ml, 100µl from 1 M stock (pH 8.0) and 20 µl from 0.5 M stock (pH 8.0) was

added and made up to 98.8 ml with sterile autoclaved milli Q water.

Chloroform: Isoamyl alcohol

Chloroform: Isoamyl alcohol was prepared in the ratio of 24: 1.

3M sodium acetate:100ml (pH 5.2)

24.61 g of sodium acetate was weighed and dissolved in 80 ml of autoclaved millipore

water. The pH was adjusted with glacial acetic acid. The volume was made up to 100

ml. Autoclaved and stored at 4°C.

70% Ethanol

To sterile 30ml of autoclaved Millipore water 70 ml of absolute ethanol was added and

mixed.

10X TBE: 1000ml (pH8.2)

Tris : 107.78g

EDTA: 8.41g

Boric acid :55.00g

The above chemicals were dissolved in 600ml of autoclaved water and made upto one

liter and autoclaved.

Ethidium Bromide

Stock: 10mg/ml

Working stock: 1µg/ml.

Agarose gel preparation (0.7%)

Page 77: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxvii

In a 50 ml 1X TBE buffer 0.35g of agarose was added, melted in microoven for 3

minutes, cooled to about 44°C to 50°C and poured into a gel platform with the comb in

position.

Running gel

After the solidification of the gel (approx.45 min), the gel was placed in a gel tank

with 1X TBE buffer. The buffer was filled to the surface of the gel. The samples were

loaded in each well and the electrophoresis was carried out at 60V till the blue dye

runs to the end.

Staining the gel

Staining solution was prepared by adding 10 µl of 10mg/ml stock of Ethidium bromide

in 100ml of sterile autoclaved millipore water.

Gel loading dye: 10X stock (10ml)

Bromophenol blue – 0.25%

Ficoll - 25%

Bromophenol blue of 25mg was weighed and dissolved in 7 ml of sterile autoclaved

Millipore water, in screw cap centrifuge tube. Ficoll of 2.5 g was added and dissolved

completely (kept in shaker, overnight). The volume was measured and made up to

10ml using autoclaved Millipore water and stored at 4°C.

4.4.4. METHOD

Page 78: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxviii

4.4.4.1. Plasmid isolation by alkaline lysis method

The isolation of plasmid was carried out following the steps given below.

1. Transformed E. coli DH5α (pCAMBIA 1301) and E. coli DH5α (pMA-T) was

inoculated into 10 ml of LB broth containing Kanamycin and Ampicillin antibiotic

for 4-5 hours at 37 °C in a rotary shaker till late log phase.

2. In 1.5 ml eppendorf tubes, 1.5 ml of the culture was taken and centrifuged at 12,000

rpm for 5 minutes and the supernatant was discarded. Another 1.5 ml culture was

centrifuged in the same eppendorf tube and the supernatant was discarded.

3. The pellets were resuspended in 100 µl of Solution I (the cells were suspended well

using vortex mixer).

4. Immediately 200 µl of freshly prepared Solution II was added. The contents were

mixed by inverting the tubes 4 to 6 times and kept in ice for 15 minutes.

5. To each tube 150 µl of ice cold Solution III was added and gently mixed by

inverting the tubes upside down and kept in ice for 15 minutes.

6. The tubes were centrifuged in a microfuge for 15 minutes at 12,000 rpm.The

supernatant was collected and 300 µl of neutral phenol was added. The content was

mixed and microfuged for 5 minutes.

7. The aqueous phase was carefully transferred into fresh eppendorf tube avoiding

interphase.

Page 79: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxix

8. Ice cold isopropanol of 300 µl was added, mixed and left in the room temperature

for 2 minutes.

9. The content was centrifuged at 12,000 rpm for 10 minutes. The supernatant was

discarded and drained over tissue paper for 5 minutes. To the pellet 100 µl TES was

added and the pellets were dissolved.

10. Neutral phenol: chloroform mix of 150 µl was added and spinned for 5minutes.

11. Aqueous phase was collected and ether saturated with water was added (200 µl to

250 µl) mixed and spinned for 5 minutes.

12. The top ether phase was removed and ether extraction was repeated.

13. The tubes were kept open in the water bath for 10 minutes at 50°C to remove

ether.

14. The ether extracted aqueous phase was measured and 1/10th

volume of 3 M sodium

acetate (pH 5.2) was added.

15. To the above aqueous phase 2.5 volumes of 95% ethanol (ice cold) was added and

mixed by inverting the tubes.

16. The tubes were kept in -20°C overnight.

17. The tubes were microfuged at 12,000 rpm for 10min at 4°C.

Page 80: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxx

18. Ethanol was decanted and 500 µl of 70% ethanol was added to the pellet. Gently

the tubes were mixed by inverting and spinned at 12,000 rpm for 5 minutes at 4°C.

19. The pellets were dried for 10 minutes.

20. The dried pellets were dissolved in 20 µl of 0.1 X TE (pH 8.0).

21. The presence of the plasmid DNA was checked by loading 5 µl on agarose

minigel.

Note: some samples were treated with Heat treated RNase of 1 µl (10mg/ml) was

added and incubated in a water bath at 37°C for 5 minutes after step9.

4.4.4.2. Gel electrophoresis

Gel electrophoresis was carried out following the steps given below.

1. Gel was casted using agarose (8.0%). Agarose was weighed and dissolved in 1X

TBE buffer by heating in a microwave oven until the agarose melted. After the

solution was cooled to about 60°C, it was poured into a casting tray containing a comb

and allowed to solidify at room temperature for 45 minutes.

2. The comb was carefully removed after the gel was solidified without ripping the

bottom of the gel. The casting tray along with the gel was placed horizontally in the

electrophoresis tank containing 1X TBE buffer.

3. The plasmid samples (5 µl) and gel loading dye (3 µl) was added to the wells and

electrophoresis was carried out at 60V till the blue dye runs 75% of the gel.

Page 81: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxxi

3. The gel was placed in a staining solution containing ethidium bromide for 30

minutes and viewed in UV transilluminator.

4. The image of gel was captured using gel documentation and the gel was analysed

in Biorad and Biocaputre software.

Page 82: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxxii

4.5. CLONING OF HCC-scFv FROM pMA-T PLASMID INTO pCAMBIA 1301

4.5. PRINCIPLE

Cloning the gene of interest into plasmid vector requires restriction

enzymes and T4 DNA ligase. Cloning of HCC-scFv from pMA-T into pCAMBIA

1301 plasmid is shown in the Figure 4.5.1.

Page 83: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxxiii

Figure 4.5.1: Illustration of cloning HCC-scFv gene into pCAMBIA 1301.

4.5.1. RESTRICTION DIGESTION OF pCAMBIA 1301 AND pMA-T FOR

CLONING

4.5.1.1. OBJECTIVE

To restrict digest pMA-T (HCC-scFv) and pCAMBIA 1301 (Ti-binary-T-

DNA vector) using restriction enzymes.

4.5.1.2. PRINCIPLE

RESTRICTION ENZYMES

Restriction enzymes are endonucleases that cleave the deoxy ribose sugar-

and phosphate backbone of the DNA. This leaves a phosphate group on the 5' ends and

a hydroxyl on the 3' ends of both strands. Restriction enzymes recognise 6 to 8 bp on

double stranded DNA and usually recognise palindrome sequence. Type II restriction

enzymes recognise specific DNA sequence and cleave the DNA at the specific

recognition site or near to the recognition site. Fast digest enzymes are an advance line

of restriction enzymes for rapid DNA digestion. The fast digest enzymes are able to

digest the DNA in 5-15 minutes. This enables any combination of restriction enzymes

to work simultaneously in one reaction tube and eliminates the need for sequential

digestions. These enzymes can be used for digestion of plasmid, genomic, PCR and

viral DNA and does not show star activity even in prolonged incubations. The

enzymes used in this experiment are PmlI and NcoI.

Page 84: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxxiv

PmlI recognises the sequence given below

NcoI recognises the sequences given below

PmlI causes blunt end restriction and NcoI causes sticky end restriction. One unit of

enzyme is usually defined as the ability to digest 1 µg of lambda DNA with 1µl of the

enzyme in 5 minutes at 37°C in 20 µl of reaction mixture.

4.5.1.1. MATERIALS

Restriction enzyme

1. Fast Digest® NcoI (FD0574)-Fermentas

2. Fast Digest® PmlI (Eco721)(FD0364)- Fermentas

Eppendorf tube

Biophotometer

Water bath

TAE buffer (50X)-pH: 8.0

Stock: -100ml (autoclaved Millipore water)

Tris Base (MW: 121.1) - 24.20g

EDTA (MW: 372.2) -1.86g

Page 85: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxxv

Glacial acetic acid -5.71ml

Working stock (500ml)

To 490ml of autoclaved Millipore water, 10 ml of TAE buffer (50X) was added and

the buffer was used for 0.8% agarose gel preparation and as buffer for electrophoresis.

4.5.1.2. METHOD

1. The following reaction components were combined at room temperature in the

order indicated in Table 4.5.1.1 and Table 4.5.1.2. The reaction volume was

increased based on the amount of plasmid DNA used for restriction digestion.

Water,nuclease-free(R0581) 14µl 24µl

10 X Fast Digest® 2 µl 4 µl

Plasmid pCAMBIA1301 2 µl(up to1µg) 4 µl (4 µg/ µl)

Restriction enzyme Fast Digest® NcoI 1 µl 4 µl

Fast Digest® PmlI 1 µl 4 µl

Total volume 20 µl 40 µl

Table 4.5.1.1: Restriction Digestion set up: pCAMBIA 1301: Vector DNA

Water,nuclease-free(R0581) 14µl 24µl

10 X Fast Digest® 2 µl 4 µl

Plasmid pMA-T (HCC-

scFv)

2 µl(up to1µg) 4 µl (4 µg/ µl)

Page 86: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxxvi

Restriction enzyme Fast Digest® NcoI 1 µl 4 µl

Fast Digest® PmlI 1 µl 4 µl

Total volume 20 µl 40 µl

Table 4.5.1.2: Restriction Digestion set up: pMA-T (HCC-scFv): Insert DNA

2. The mixture was gently mixed and spinned down in microfuge.

3. The tubes were incubated at 37°C (both enzymes restrict at the same time) in a

thermostat water bath for 10 minutes or15 minutes (when the volume exceeds 20 µl).

4. Digested sample of 5 µl was loaded onto 0.8% agarose gel prepared in TAE

with 3 µl of gel loading dye and electrophoresis was carried out at 60V till the

blue dye runs 75% of the gel.

5. The gel was placed in a staining solution containing Ethidium bromide for 30

minutes and viewed in UV transilluminator.

6. The image of gel was captured using gel documentation and the gel was

analyzed in Biorad and Biocaputre software.

Page 87: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxxvii

4.5.2. LIGATION OF HCC-scFv AND pCAMBIA 1301

4.5.2.1. OBJECTIVE

To ligate restrict digested fragments using T4 DNA ligase enzyme.

4.5.2.2. PRINCIPLE

T4 DNA ligase enzyme catalyzes the formation of a phosphodiester bond

between 5' phosphate and 3' hydroxyl termini in duplex DNA or RNA (Weiss and

Richardson (1967), Lehnman 1974). This enzyme will join blunt end and cohesive end

termini and also repairs single stranded nicks in duplex DNA, RNA or DNA/RNA

hybrids. T4 DNA ligase requires ATP as the cofactor for ligation reaction, whereas E.

coli DNA ligase requires NAD as the cofactor. Ligation reaction was carried out at the

room temperature (20°C to 25°C). The Rapid DNA ligation kit enables sticky-end or

blunt-end DNA ligation in only 5 minutes at room temperature. The kit includes T4

Page 88: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxxviii

DNA Ligase and a specially formulated 5X Rapid Ligation Buffer optimized for fast

and efficient DNA ligation. The ligation reaction mixture can be used directly for

bacterial transformation using calcium chloride transformation method or bacterial

transformation kit. The transformed ligated clone was confirmed on 0.8% agarose gel

electrophoresis (Adkins 1996).

4.5.2.3. MATERIALS

4.5.2.3.1. GEL ELUTION OF THE PARTICULAR BANDS RESTRICTED

WITH RESTRICTION ENZYMES

Gel elution Kit (Real Genomics, Hi Yield TM

Gel/PCR DNA MINI KIT-

YDF100/YDF300)

100 mini preps/kit

DF Buffer: 80ml

W1 Buffer: 45ml

Wash Buffer (concentrated):25ml*

Elution Buffer: 6ml

DF Column: 100pcs

2ml Collection Tube: 100pcs

*Initially 100ml of ethanol (96-100%) was added to 25 ml of wash buffer prior to use

Sterile Lancet Blade

Plastic cover

Scale

2ml eppendorf tubes

DNase-free pipette tips

4.5.2.3.2. LIGATION OF THE VECTOR (pCAMBIA 1301) AND INSERT

(HCC-scFv)

Page 89: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

lxxxix

Rapid DNA Ligation Kit (K1422- Fermantas)

Linearized vector DNA- (pCAMBIA 1301-9.802 kb)

Insert DNA - (at 3:1 molar excess over vector)-(HCC-scFv-0.946 kb)

5X Rapid Ligation Buffer

T4 DNA Ligase (5U/ µl)

Water, nuclease-free

Autoclaved microfuge tubes, microtips etc.

4.5.2.3.3. TRANSFORMATION OF THE LIGATED SAMPLES

2X YT medium (200 ml) pH 7.0

Tryptone 3.2g

Yeast Extract 2.0g

Nacl 1.0g

Mgcl2.6H2o 0.8132g

LB medium (1000 ml) pH 7.2

Tryptone 10.0g

Yeast Extract 5.0g

Nacl 10.0g

Agar (1.5%) 15.0g

100mM Cacl2 (400ml)

Page 90: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xc

Weigh 5.88 g of Cacl2 (1M=147.01g/mol-Merck: 10238005001730) and dissolve in

350 ml of autoclaved millipore water. Make up the volume to 400ml. Autoclave and

store at 4°C.

X-gal (5-Bromo-4-chloro-3-Indolyl-β-D-galactoside)-Sigma B4252

Stock: (20mg/ml): 20mg of X-gal was dissolved in DMF (dimethyl formamide).The

tube was wrapped in aluminum foil and stored at -20°C.

Working stock: X-gal was used in plates at a concentration of 40µg/ml: 2µl/ml of LB

media.

IPTG (Iso propyl thiogalactoside or isopropyl beta-D-thiogalactopyranoside-

Sigma-11502

Stock: 0.1 M IPTG :( 1M=238.3g/mol): 0.2383 g of IPTG was dissolved in 10 ml of

autoclaved Millipore water and stored at -20◦C.

Working stock: IPTG was used in the concentration of (0.1mM): 1µl/ml of LB media.

Kanamycin

Stock: (100mg/ml)

Kanamycin of 100mg was dissolved in 1 ml of sterile autoclaved Millipore water.

Working stock: (50µg/ml)

4.5.2.4. METHOD

4.5.2.4.1. GEL ELUTION OF THE PARTICULAR BANDS RESTRICTED

WITH RESTRICTION ENZYMES

Gel Dissociation

Page 91: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xci

1. The agarose gel containing the samples restricted with enzymes was placed on a

plastic paper over the UV illuminator. The bands were visualized and particular

band of interest was marked.

2. The band of particular size of approximately 10Kb (pCAMBIA1301) from lane

2-5 and approximately 1Kb (HCC-scFv-pMA-T) from lane6-9 was excised

from the gel using sterile lancet blade.

3. Extra agarose gel was removed to minimize the size of the gel. The gel was

made using TAE buffer as TBE buffer might affect the downstream experiment.

4. The gel slice of 300mg was transferred into 1.5 ml microcentrifuge tube.

5. DF Buffer of 500µl was added to the samples and mixed by vortexing.

6. The tubes were incubated at 55 to 60°C for 10-15 minutes until the gel slice

have been completely dissolved. The tubes were inverted every 2-3 minutes.

7. The dissolved samples were cooled to room temperature.

DNA Binding

8. DF column was placed in a 2 ml collection tube.

9. The sample mixture of 800µl from step 7 was added into DF Column.

10. The column was centrifuged at full speed (approximately 13, 000 rpm) for 30

seconds.

11. The flow through was discarded and the DF Column was placed back into the 2

ml collection tube. When the samples were more than 800µl, this step was

repeated.

Wash

12. W1 Buffer of 400 µl was added into the DF Column.

13. The columns were centrifuged at full speed (approximately13, 000 rpm) for 30

seconds.

Page 92: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xcii

14. The flow through was discarded and the DF column was placed back into the 2

ml collection tube.

15. Wash buffer of 600 µl was added into the DF column and allowed to stand for 1

minute.

16. The column was centrifuged at full speed (approx.13, 000 rpm) for 30 seconds.

17. The flow through was discarded and the DF column was placed back in 2ml

collection tube.

18. The columns were centrifuged again for 3 minutes at full speed (approximately

13000rpm) to dry the column matrix.

DNA Elution

19. The dried DF column was transferred into a new 1.5ml microcentrifuge tube.

20. Elution buffer or TE of 20 µl to 50 µl was added into the center of the column

matrix.

21. The elution buffer was allowed to stand for 2 minutes until the buffer is

absorbed by the matrix.

22. The column was centrifuged at full speed for 2 minutes to elute the purified

DNA

23. The eluted DNA of 2 µl was diluted with 48 µl of sterile water and the quantity

was measured using biophotometer.

4.5.2.4.2. LIGATION OF pCAMBIA 1301 FRAGMENT AND Hcc-scFv

FRAGMENT OF pMA-T

1. The 5 X Rapid ligation buffer was thoroughly mixed prior to use.

2. The ligation reaction was setup as below (Table 4.5.2.1) in a microfuge tube.

Page 93: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xciii

S. No Reaction mixture volume

1 Linearized vector DNA

(pCAMBIA 1301-9.802 kb)

2 µl (100ng)

2 Insert DNA(at 3:1 molar excess over vector)

(HCC-scFv-0.946 kb)

1 µl (28.95ng )

(approx. 29ng)

3 5X Rapid Ligation Buffer 4 µl

4 T4 DNA Ligase(5U/ µl) 1 µl

5 Water, nuclease-free 12 µl

Total volume 20 µl

Table 4.5.2.1: Setup of Ligation reaction

3. The vector and insert DNA ratio was calculated using the Equation (4.5.2.1).

Vector: Plasmid (pCAMBIA 1301) - concentration of plasmid used was 100ng and it

was about 9.802 Kb size after restriction with NcoI and Pml1

Insert: HCC-scFv- the size of the insert DNA was 0.946 Kb which was restricted with

NcoI and Pml1 restriction enzyme. The concentration of the insert to be taken was

calculated using the Equation (4.5.2.1) and it was found to be 28.95ng. The insert

DNA was taken in 3 moles excess of vector DNA.

4. The controls used for ligation setup were carried out as in Table 4.5.2.2.

Page 94: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xciv

Content DNA Water,

nuclease-

free

5X Rapid

Ligation

Buffer

T4 DNA

ligase(5 unit/

µl)

Uncut pCAMBIA

1301

2 µl(100ng) 18 µl

Cut

pCAMBIA1301

2 µl(100ng) 18 µl

Cut (pCAMBIA

1301)+ligated

2 µl(100ng) 13 µl 4 µl 1 µl

Experiment-1

Vector(cut-9.802 kb)

+

Insert(0.946 kb)

2 µl(100ng)

1 µl(29ng)

12 µl

4 µl

1µl

Experiment-2

Vector(cut-9.802 kb)

+

Insert(0.946 kb)

2 µl(100ng)

2 µl(58ng)

11 µl

4 µl

1 µl

Table 4.5.2.2: Controls for Ligation setup.

5. The contents were vortexed and spinned briefly to collect the drops.

6. The mixture was incubated at 22°C for 5 min.

7. Ligation mixture of 2-5 µl was used for transformation.

The reaction mixture can be stored at 0°C to 4°C until used for transformation.

4.5.2.4.3. TRANSFORMATION OF THE LIGATED SAMPLES

4.5.2.4.3.1. COMPETENT CELL PREPARATION

A single colony of E. coli (DH5α) was inoculated into 2 ml of LB culture

broth and incubated (Scigenics, India) at 37°C for 10 hours to 12 hours overnight.

From the overnight culture, 500µl was inoculated into 50 ml of 2X YT medium and

incubated at 37°C in a rotary shaker until it reached an O.D of 0.5 to 0.6 at 600nm

(Biophotometer, Germany).To a sterile 50 ml polypropylene tube, 30 ml of the culture

Page 95: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xcv

was transferred and incubated in ice for 30 minutes. The culture was centrifuged at

5,000 rpm at 4°C for 10 minutes. The supernatant was discarded and the pellet was

resuspended in a small volume of medium left behind and finally the pellet was gently

resuspended in 30 ml of ice cold 100mM CaCl2 and incubated in ice for 30 minutes.

The resuspended cells were centrifuged at 5,000 rpm for 10 minutes at 4°C.The

supernatant was discarded and the pellet was resuspended gently in 3ml (1/10th

volume

of CaCl2) of ice cold 100mM CaCl2.The competent cells were stored in ice for atleast

30 minutes before use.

4.5.2.4.3.2. TRANSFORMATION OF COMPETENT CELLS (E. coli)

Competent cells of 200 µl was aliquated into 2ml sterile eppendorf tubes and

incubated in ice (cut tips were used to aliquot the cells). The liagted samples were

dispensed into the above eppendorf tubes containing competent cells separately and

incubated in the ice for 30 minutes. Heat shock was given for 2 minutes at 42°C in

water bath and then transferred and incubated in ice for 10 minutes.The cells were

transferred to 0.8 ml of prewarmed LB mix and incubated at 37°C in a rotary shaker

for 1 hour at 220rpm.The LB mixture was then centrifuged at 10,000 rpm for 5

minutes and the supernatant was discarded. From the pellet, 50 µl and 200µl of cells

were plated onto LB agar plate containing antibiotics kanamycin (50µg/ml). The plates

were incubated overnight at 37°C. The controls kept for the cloning experiment are

shown below in the Table 4.5.2.3.

S.No Innoculum Medium Volume to be

plated(µl)

Page 96: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xcvi

1.

2.

3.

4.

5.

Competent cells

Uncut plasmid vector

Vector cut

Vector cut+ ligated

Digested vector + insert +

ligase(Experiment:1&2)

(a) LB [plain]

(b) LB+antibiotic

LB+antibiotic

LB+antibiotic

LB+antibiotic

LB+antibiotic

100

100

100

100

100

50 µl

200 µl

Table 4.5.2.3: Controls kept for the cloning experiment.

4.5.2.4.3.3. CONFIRMATION OF THE LIGATED CLONE

From the patched colony of clone, plasmid DNA was extracted miniprep

(chapter 4.4) and digested with restriction enzyme (chapter 4.5.1). The digested

samples were seperated in 0.8% agarose gel and the clones were confirmed using 1 kb

ladder.

4.6. TRANSFORMATION OF pCAMBIA 1301-HCC-scfv FROM E. coli DH5α

INTO Agrobacterium tumefaciens EHA105

Page 97: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xcvii

4.6.1. OBJECTIVE

To transform pCAMBIA 1301-HCC-scFV from E. coli DH5α into

Agrobacterium tumefaciens EHA105 by triparental mating.

4.6.2. PRINCIPLE

The gene of interest can be transformed into Agrobacterium tumefaciens by

electroporation, free/thaw transformation and triparental mating (Figure 4.6.1).

Triparental mating is an effective method to transfer a non-conjugative plasmid but

mobilizable plasmid into Agrobacterium. In this method two E. coli strains are used to

transfer the plasmid into Agrobacterium. E. coli strain (helper strain) carrying a

conjugative plasmid (helper plasmid), which encodes the gene of all the proteins for

the formation of mating bridge, to transfer itself or mobilize another plasmid into a

recipient cell. The helper plasmid contains mob (mobilization) and tra (transfer

functions) genes. The donar plasmid (cloning vector) contains oriT (specific origin of

transfer) and bom (activation site at which mob and tra gene product can act). The

plasmid from the E. coli (donar strain) containing the gene of interest to be

transformed into Agrobacterium is transferred by the trans-acting elements in the

helper plasmid. The trans-acting functions include single stranded break within the ori

T region and target the nicked DNA (relaxosomes) into the mating bridge that links

donor and the recipient cells(Waters 1999).

The interaction between oriT and transfer functions are specific (Waters

1999, Hamilton 2000).Widely used helper plasmid is pRK2013 which carries the

transfer and mobilization functions of RK2 on a colE1 replicon (Ditta 1985). Thus

Page 98: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xcviii

pRK2013 can transfer plasmid carrying RK2 derived oriT from E. coli into

Agrobacterium tumefaciens. Transfer of T-DNA from E. coli into Agrobacterium by

triparental mating has been described by Roger et al (2000). After mating

Agrobacterium strains harboring the transformed vector (Ti plasmid containing gene

of interest) are selected on antibiotic plates depending on the antibiotic resistant

marker present on the plasmid.

Figure 4.6.1: Illustration of transformation of pCAMBIA 1301-HCC-scFv into

A.tumefaciens EHA 105 by triparental mating

Page 99: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

xcix

4.6.3. MATERIALS

Recipient strain – Agrobacterium tumefaciens EHA 105 (super-virulent strain)-

obtained from Dr. Tomasz Pniewski, (Institute of Plant Genetics, Polish Academy of

Sciences, Poland) – Appendix 2.

Strain grows at 30°C on AB minimal medium with relevant antibiotics.

Donor strain - E. coli DH5α (containing transformed pCAMBIA 1301-ScFv)-the

plasmid to be mobilized. The strain grows at 37°C on LB medium with relevant

antibiotics (Kanamycin).

Helper strain - E. coli HB101 (containing pRK2013)-MTCC -398 (IMTEC, India)-

This plasmid helps the mobilization of the plasmid from donor to recipient strain. The

strain grows at 37°C on LB medium containing the relevant antibiotic (Kanamycin or

ampicillin)

LB medium (1000 ml) pH 7.2- (Kanamycin-50 µg/ml- E. coli DH5α, E. coli HB101)

Tryptone 10.0g

Yeast Extract 5.0g

Nacl 10.0g

Agar (1.5%) 15.0g

YEP medium (50ml) pH 7.1

Yeast Extract 1% 0.5g

Peptone 1% 0.5g

NaCl 0.5% 0.26g

Agar 1.6% 0.8g

Page 100: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

c

AB Buffer (20X): pH 7.0 100ml

K2HPO4 (anhydrous) -6.0g

NaH2PO4 (anhydrous) -2.0g

Or

NaH2PO4 .2H2O (hydrated)-2.6g

Each salt was dissolved separately in about 50 ml of autoclaved Millipore water and

made up to 100ml. The pH was adjusted to 7.0. The buffer was autoclaved and stored

at room temperature.

AB salts (20X) -100ml

NH4cl - 2.0g

MgSo4.7H2O - 0.6g

Kcl - 0.3g

Cacl2.2H2O - 0.31g

FeSO4.7H2O - 0.005g

All the salts were dissolved in 80ml of Millipore water and made up to 100ml. The

contents were autoclaved and stored at room temperature.

AB minimal medium (Agrobacterium tumefaciens EHA 105)

For pour plate inoculation of the serially diluted samples, 400 ml of AB minimal

medium was prepared by adding 2 g glucose and 6 g agar to 360 ml of millipore water

and autoclaved. To this 20 ml of AB salt and 20 ml AB buffer was added aseptically

and respective amount of antibiotics, rifampicin and kanamycin were added.

For streaking of the colonies obtained from serial dilution, 300ml of AB minimal

medium was prepared by adding 1.5 g glucose and 4.5 g Agar to 270 ml of millipore

water and autoclaved. To this 15 ml of AB salt and 15 ml of AB buffer was added

Page 101: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ci

aseptically and respective amount of antibiotics from rifampicin and kanamycin stock

were added.

Antibiotic stocks

Stock: Rifampicin (10mg/ml)

Working concentration: Rifampicin (10µg/ml)

Stock: Kanamycin (100mg/ml)

Working concentration: Kanamycin (100µg/ml, 50µg/ml)

4.6.4. METHOD

The day on which triparental mating is performed was considered as day

one.

Day: -4

Agrobacterium tumefaciens (EHA 105) was streaked to get single colonies on AB

minimal medium agar or YEP agar plates containing antibiotic (rifampicin-10µg/ml)

and incubated at 30°C.

Day: -1

E. coli HB101 (containing pRK2013) was streaked to get single colonies on LB agar

with 50µg/ml of kanamycin and incubated at 37°C.

E. coli DH5α (containing transformed pCAMBIA 1301-ScFv) was also streaked on

LB agar with 50µg/ml of kanamycin and incubated at 37°C.

Day: +1

A plate with YEP agar plate was prepared without antibiotics. One colony of E. coli

DH5α(pCAMBIA 1301-HCC-scFv) and E. coli HB101(pRK2013) were spotted over

YEP agar medium separately and 3 closely lying colonies of A. tumefaciens (EHA

Page 102: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cii

105) were picked and spotted near to the other 2 strains of E. coli using a sterile loop,

all three bacterial strains were mixed well. Then the plates were incubated at 30°C for

12 to 18 hours.

Day: +2

The colony obtained as a result of mating was scrapped and suspended in 1.0 ml of

0.9% sterile sodium chloride solution. Serial dilution was performed by transferring

0.1 ml of bacterial suspension into 0.9% NaCl. Similarly six dilutions were made up to

10-6

. 100 µl of each dilution was plated on AB minimal medium containing

Rifampicin (10µg/ml) and Kanamycin (50µg/ml). The cultures were uniformly spread

onto the surface of the media using L rod by spread plate technique. The plates were

incubated at 30°C for 3 to 5 days.

*Note: Before each transfer the contents were vortexed thoroughly.

Day: +6

At one or two dilutions, single colonies appear on AB minimal medium. These

colonies are Agrobacterium tumefaciens into which the donor plasmid has been

transformed.

Day: +7

Six to eight single colonies of Agrobacterium tumefaciens were then streaked on AB

minimal medium containing antibiotics and incubated at 30°C for 2 to 3 days to get

single colonies.

Day: +11

Then the single colonies were patched and maintained as master plate for further use in

duplicates.

Page 103: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ciii

A glycerol stock of the transconjugants was made immediately and used for further

studies.

4.7. CALLUS INDUCTION OF IR64 RICE VARIETY FOR

TRANSFORMATION

4.7.1. OBJECTIVE

To induce callus by indirect organogenesis in IR64 rice variety for

Agrobacterium mediated transformation

4.7.2. PRINCIPLE

Plant tissue culture is a technique whereby the explants are isolated from

the plant and grown aseptically on a nutrient medium under controlled environmental

conditions (16/8 hours of light/dark and 23ºC to 25ºC)

Organogenesis is the formation of either shoots or roots and depends on the

balance between the auxin and cytokinin phytohormones during invitro culture.

Organogenesis is of two types indirect and direct. During indirect organogenesis, the

organs are formed indirectly via a callus phase and in direct organogenesis, the organs

are formed without undergoing callus phase. Induction of plants via callus phase has

been widely used for the production of transgenic plants.

Page 104: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

civ

Totipotency is the ability of a single plant cell to regenerate into an entire

plant. Callus is undifferentiated mass of parenchymatous cells. Callus induction from

the embryo is very useful to obtain an increased number of hybrid plants. Formation of

shoots or roots from a callus is known as organogenesis. First report on organogenesis

was made (White 1939). Induction of callus shoots and roots can be done from any

part of the plant using different medium. The two major classes of growth regulators

essential for both callus induction and organogenesis are auxins and cytokinins.

Auxin induces cell division, cell elongation, swelling of tissues and the

formation of adventitious roots. At low concentration of auxin, adventitious root

formation takes place, whereas at high concentration of auxin root formation fails to

occur and callus formation takes place. Cytokinins are derivatives of adenine and have

important role in shoot induction. They usually promote cell division if added together

with an auxin. At higher concentration (1 to 10 mg/L), adventitious shoot formation is

induced but root formation is generally inhibited.

In this experiment 2,4D was used for callus induction and later for

regeneration NAA (Naphthalene acetic acid) and kinetin was used as auxin and

cytokinin respectively.

4.7.3. MATERIALS

1. Explant: Healthy matured Rice (Oryza sativa L) seeds of IR64 (Indica rice variety

64) was obtained from ICAR, Cuttack, India.

2. Sterilizing Solutions: 70% ethanol, 3-4% sodium hypochloride (Merck) with 1-2

drops of Tween-20(Sigma), 0.2 % (w/v) HgCl2

(mercuric chloride) (Rankem), sterile

millipore water.

Page 105: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cv

3. Stock solution: 2, 4-Dichlorophenoxy acetic acid (2, 4-D; Sigma)

20mg/100ml.Dissolve the flake in a few drops of 0.1 N KOH and make the volume to

100ml with autoclaved millipore water and store at 4°C.

4. MS 2, 4-D medium: MS salts(Merck) and vitamins (Sigma), 300mg/L casamino

acid, 2mg/L 2, 4-D and 8 g/L agar, pH 5.8 (Autoclaved) (Table 4.7.1).

Name of

stock

solution

Chemicals

1000ml

500ml

250ml 1Quantit

y

g/L

*V of

stock

1Quantit

y

g/L

*V of

stock

1Quantit

y

g/L

*V of

stock

MS 1 NH4NO3 82.5 20 41.25 10 20.625 5

KNO3 95.0 47.5 23.75

MS2 MgSO4.7H2O 37.0 10 18.5 5 9.25 2.5

MnSO4.4H2O 2.23 1.115 0.5575

ZnSO4 1.058 0.529 0.2645

CuSO4.5H2O 0.0025 0.00125 0.000625

MS3 Cacl2.2H20 44.0 10 22.0 5 11.0 2.5

KI 0.083 0.0415 0.02075

CoCl2.6H2O 0.0025 0.00125 0.000625

MS4 KH2PO4 17.0 10 8.5 5 4.25 2.5

H3BO3 0.62 0.31 0.155

Na2MoO4.2H2

O

0.025 0.0125 0.00625

MS5 FeSO4.7H2O 2.785 10 1.3925 5 0.69625 2.5

Na2EDTA.2H2

O

3.725 1.8625 0.93125

Vitamins Nicotinic acid 10mg/10

0ml

5 10mg/10

0ml

2.5 10mg/10

0ml

1.25

Pyridoxine

HCl

10mg/10

0ml

10mg/10

0ml

10mg/10

0ml

Thiamine HCl 20mg/10

0ml

20mg/10

0ml

20mg/10

0ml

Glycine 40mg/10

0ml

40mg/10

0ml

40mg/10

0ml

Hormone 2,4D 10mg/10 20 10mg/10 10 10mg/10 5

Page 106: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cvi

0ml 0ml 0ml

Myoinositol 0.1g 0.05g 0.025g

Casamino

acid

0.3g 0.15g 0.075g

Sucrose/

Maltose

30g 15g 7.5g

Agar 8.0 4g 2g

Table 4.7.1: Chemical composition of MS 2, 4-D medium

*V- Volume of stock to prepare the media 1Quantity- quantity of chemicals required to prepare the stock

4.7.4. METHODS

1. The seeds of IR 64 were manually dehusked.

2. The dehusked seeds were surface sterilized with 70% (v/v) ethanol for 1 min and

then with 0.2 % (w/v) HgCl2 (mercuric chloride) for 10 minutes with gentle agitation

and rinsed several time with sterile Millipore water to remove HgCl2. It was further

sterilized with 3 to 4% sodium hypo chloride (containing 2 to 3 drops of tween 20) for

30 minutes with gentle agitation and rinsed with autoclaved sterile Millipore water.

3. Individually twenty five embryos from the sterilized seeds were placed in each Petri

dish containing 20 ml of MS 2, 4-D medium.

4. The plates were incubated in the dark at 25 ± 1ºC.

5. After 3 to 4 days, the emerging shoots and roots were cut. The remaining scutellar

tissues were subcultured onto a fresh medium with the same orientation.

Page 107: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cvii

6. Yellowish white soft embryogenic calli developed on the surface of the scutellar

tissue after 2 to 3 weeks.

7. Callus (total/embryogenic) induction frequency (%) can be calculated as in Equation

(4.7.1).

4.8. TRANSFORMATION OF pCAMBIA 1301-HCC-scFv FROM

Agrobacterium tumefaciens EHA 105 INTO CALLUS BY Agrobacterium

MEDIATED TRANSFORMATION

4.8.1. OBJECTIVE

To transform pCAMBIA 1301-HCC-scFv gene from Agrobacterium

tumefaciens EHA 105 into callus by Agrobacterium- mediated transformation.

4.8.2. PRINCIPLE

The Plant pathogenic Agrobacterium tumefaciens have the capacity to

transfer part of its T-DNA into the nuclear genome of the plant cells. Two types of

Agrobacterium strains that are used for plant genetic transformation are A. tumefaciens

Page 108: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cviii

and A. rhizogenes. In case of A. tumefaciens strains, the oncogenes are encoded in the

T-DNA (Ti) which will induce tumor formation on the infected plant tissue and in case

of A. rhizogenes strains, the T-DNA (Ri) genes encode oncogenes that will induce the

production of adventitious roots called the hairy root tissue. The transfer of T-DNA

depends on the expression of the vir genes which transfers the DNA into plant nucleus

by recognizing specific sequence called right and left border (RB and LB) sequence.

Several strategies to introduce foreign genes directly into the T-region of Ti

plasmids have been developed by Hoekema et al (1983) and de Frammond et al

(1983). A binary vector system was developed were the replicon harboring the T-DNA

region constituted the binary vector, whereas the replicon containing the vir genes was

known as vir helper. The vir helper plasmid does not induce tumor but contain

complete or partial T-region. A number of Agrobacterium strains containing non-

oncogenic vir helper plasmids have been developed, including LBA4404 (Ooms

1981), GV3101 MP90 (Koncz 1986), AGL0 (Lazo 1991), EHA101 and its derivative

strain EHA105 (Hood 1986, 1993) and NT1 (pKPSF2) (Palanichelvam 2000).

Commonly used non- oncogenic A. tumefaciens vectors have the oncogenic

portion of the T-DNA replaced by a dominant selectable marker gene. In this

experiment A. tumefaciens EHA 105 transformed (Karabi Datta and Swapan Datta,

2006) with pCAMBIA 1301-HCC-scFv was allowed to infect the callus of IR 64 (

Figure 4.8.1) .The selectable marker gene present in pCAMBIA 1301 is hpt gene

which codes for hygromycin phosphotransferase which detoxifies the aminocyclitol

antibiotic hygromycin B. Most of the cereals exhibit higher sensitivity to hygromycin

B than to kanamycin. So this gene can be used to screen the transformed callus. The

reporter gene gus A was not used as it was excised and inserted with the gene of

interest.

Page 109: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cix

Figure 4.8.1: Illustration of transformation of pCAMBIA 1301-HCC-scFv into IR

64.

4.8.3. MATERIALS

AB Buffer (20X): pH 7.0 100ml

K2HPO4 (anhydrous) -6.0g

NaH2PO4 (anhydrous) -2.0g

Or

NaH2PO4 2H2O (hydrated)-2.6g

Each salt was dissolved separately in about 50 ml of autoclaved Millipore water and

made up to 100ml. The pH was adjusted to 7.0. The buffer was autoclaved and stored

at room temperature.

AB salts (20X) -100ml

Page 110: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cx

NH4cl - 2.0g

MgSo4.7H2O - 0.6g

Kcl - 0.3g

Cacl2.2H2O - 0.31g

FeSO4.7H2O - 0.005g

All the salts were dissolved in 80ml of Millipore water and made up to 100ml. The

contents were autoclaved and stored at room temperature.

AB minimal medium (Agrobacterium tumefaciens EHA 105)

For pour plate inoculation of the serially diluted samples, 400 ml of AB minimal

medium was prepared by adding 2 g glucose and 6 g agar to 360 ml of millipore water

and autoclaved. To this 20 ml of AB salt and 20 ml AB buffer was added aseptically

and respective amount of antibiotics rifampicin and kanamycin were added.

For streaking of the colonies obtained from serial dilution, 300ml of AB minimal

medium was prepared by adding 1.5 g glucose and 4.5 g Agar to 270 ml of millipore

water and autoclaved. To this 15 ml of AB salt and 15 ml of AB buffer was added

aseptically and respective amount of antibiotics from rifampicin and kanamycin stock

were added.

Antibiotics

Stock: Rifampicin (10mg/ml)

Working concentration: Rifampicin (20µg/ml)

Stock: Kanamycin (100mg/ml)

Working concentration: Kanamycin (50µg/ml)

Stock: Cefotaxime (Invitrogen):100mg/ml. Filter sterilize. Store at -20°C.

Page 111: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxi

Working concentration (250mg/L)

Selective marker agent

Hygromycin B: (H3274-50MG- Sigma)

10mM MgSO4

10mM MgSO4 was mixed in water and sterilized by autoclaving and stored at -20°C.

Acetosyringone (3, 5, dimethoxy 4 hydroxy-acetophenone) (Sigma-Aldrich)

40mg/ml 200mM): The powder was dissolved in DMSO (dimethyl sulfoxide, Sigma)

and the solutions were made up using autoclaved Millipore water. The solution was

filter sterilized and stored at 4°C.

Kinetin (6 furfurylaminopurine) (Sigma):20mg/ml. The powder was dissolved in a

few drops of 1N HCl, and then the volume was made up with autoclaved Millipore

water and stored at 4°C.

Components Quantity(mg/L)

1. Macronutrients

Cacl2.2H2O

MgSO4.7H2O

NaH2PO4.H2O

KCl

150.0

250.0

150.0

2950.0

2 Micronutrients

KI

H3BO3

MnSO4.H2O

ZnSO4.7H2O

Na2MoO4.2H2O

CuSO4.5H2O

0.75

3.0

10.0

2.0

0.25

0.025

Page 112: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxii

CoCl2.6H2O 0.025

3 Iron composition

Na2EDTA

FeSo4.7H2O

Vitamins

Nicotinic acid

Pyridoxine HCl

Thiamine HCl

Glycine

Inositol

37.3

27.8

0.5

0.5

1.0

2.0

100.0

4 L-glutamine

Aspartic acid

Arginine

Casamino acid

Sucrose

Glucose

Acetosyringone(3,5,dimethoxy 4 hydroxy-

acetophenone)

876.0

266.0

174.0

500.0

68.5g/L

36g/L

200µM

Table 4.8.1: Chemical composition of AAM Medium

The components were mixed and then pH was adjusted to 5.2. The medium was

sterilized by filter sterilization (Hiei 1994).

Name of stock

solution

Components Quantity

g/L

Volume of

stock for

1L(ml)

Final

concentration

(mg/L)

N61

KNO3 141.50 20 2830.0

Page 113: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxiii

N62 MgSO4.7H2O

MnSO4.4H2O

ZnSO4

(NH4)2SO4

18.5

0.44

0.15

46.3

10 185.0

4.4

1.5

463.0

N63 Cacl2.2H20 16.6 10 166.0

N64 KI

KH2PO4

H3BO3

0.08

40

0.16

10 0.8

400

1.6

N65 FeSO4.7H2O

Na2EDTA.2H2O

2.785

3.725

10 27.85

37.25

Vitamins Nicotinic acid

Pyridoxine HCl

Thiamine HCl

Glycine

10mg/100ml

10mg/100ml

20mg/100ml

40mg/100ml

5 0.5

0.5

1.0

2.0

Hormones

Myoinositol

Sucrose/maltose

Agar

2,4 D 10mg/100ml 20 10.0

100.0

30,000.0

8000.0

Table 4.8.2: Chemical composition of N6 Medium

The pH of the N6 Medium was adjusted to 5.6-5.8. All the chemicals were obtained

from Merck. Vitamins and hormones were obtained from Sigma (Karabi Datta, 2006)

N6-AS medium for infiltration (liquid) and co-cultivation (solid)

N6 salts from the N6 salts Table 4.8.2, MS vitamins from the MS Table 4.7.1,

300mg/L casamino acid, 2mg/L2, 4-D, 30 g/L sucrose and 10 g/L glucose. In case of

cocultivation medium 9 g/L agar was added. The pH was adjusted to 5.2. The medium

was autoclaved and cooled to 55°C before adding acetosyringone (200µM).

Page 114: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxiv

NAA (1-Naphthylacetic acid-Sigma):20mg/ml. the powder was dissolved in a few

drops of 0.1N NaOH and finally made up the volume with autoclaved Millipore water

and stored at 4°C.

Selection Medium

MS salts, MS vitamins, 300mg/L casamino acid, 2mg/L 2, 4-D, 250mg/L cefotaxime

(added after autoclaving), 8g/L agar. The pH was adjusted to 5.8.

For hpt gene selection: to the above selection medium 30g/L sucrose was added and

autoclaved. After autoclaving the medium was cooled to 60 °C and then 50 mg/L

hygromycin was added.

Regeneration medium (MSKN)

MS salts, MS vitamins, 2mg/L kinetin, 1mg/L NAA, 300mg/L casamino acid, 50mg/L

cefotaxime, 30g/L sucrose, 10g/L sorbitol, 2.5g/L gelrite. The pH was adjusted to 5.8

and the medium was autoclaved.

Rooting medium (MSO)

MS salts, MS vitamins, 30g/L sucrose, 2.5g/L gelrite. The pH was adjusted to 5.8 and

the medium was autoclaved.

4.8.4. METHOD

Page 115: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxv

Preculturing of Agrobacterium strain

Day 1: A loop full culture was taken from a glycerol stock culture of

Agrobacterium transformed with pCAMBIA 1301-HCC-scFv and streaked

on to AB minimal medium containing 20 mg/ml rifampicin and 50mg/ml

kanamycin.

The plates were incubated in the dark at 28°C for 2 days.

Preculturing of calli

Day1: Embryogenic calli of 1.3-3mm diameter size calli was selected from

4 to 5 weeks old culture and transferred into fresh MS 2, 4 D medium

(approx 50 Calli /dish).

The plates were incubated at 28°C in the dark for 3 days.

Culturing of Agrobacterium strain

Day 3: Three to four well grown Agrobacterium colonies were transferred to

50 ml of AAM medium (Table 4.8.1) containing 20mg/ml of rifampicin and

50 mg/ml of Kanamycin in a 250ml conical flask

The conical flask was kept in a shaker at 250 rpm for 20 to 30 hours at

28°C.

Transformation

1. Day 4: Acetosyringone of 200 µM was added to the Agrobacterium suspension

and kept in shaker at 250 rpm for 2 hours at 28°C.

2. The bacterial culture was transferred into a 50 ml centrifuge tube and

centrifuged at 3,500g for 30 minutes at 10°C.

3. To the pellet 20 ml of Mg So4 was added and the OD measurement of the

Agrobacterium suspension was measured at 600nm.

Page 116: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxvi

4. The above bacterial suspension was centrifuged at 3,500g for 30 minutes at

10°C and the supernatant was discarded.

5. The pellet was resuspended in a small volume of liquid N6-AS in filtration

medium by pipetting up and down.

6. The OD of the final volume of agrobacterial suspension was adjusted to 1 with

N6-AS medium.

7. The bacterial suspension was transferred into a sterile Petri plate and then the

precultured embryogenic calli was put into the bacterial suspension.

8. The Petri plate was placed open in a vacuum desiccator for 10 minutes.

9. After another 20 minutes, the bacterial suspension was pipetted out from the

embrogenic calli.

10. Excess Agrobacterium-infection medium was absorbed from the calli using

sterile filter papers.

11. The calli was then transferred to N6-AS solid co-cultivation medium and the

plates were incubated at 28°C for 3 days under dark condition.

Calli Proliferation

After 3 days, the calli was transferred on MS-2, 4-D medium with 250mg/L

cefotaxime for proliferation of the calli.

The calli was incubated in the dark at 25°C for 10 days.

Selection

1. The calli was first selected on a medium containing antibiotic (250mg/l

cefotaxime) and selective agent (50mg/ml hygromycin B) and kept in the dark

for 2 weeks at 25 °C.

2. The survining healthy embryogenic calli was transferred from the first selection

media into the second selection media containing antibiotic (250mg/l

Page 117: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxvii

cefotaxime) and selective agent (50mg/ml hygromycin B) and kept in the dark

for 2 weeks at 25°C.

3. The survining embryogenic calli was further subcultured on a third selection

medium containing antibiotic (250mg/l cefotaxime) and selective agent

(50mg/ml hygromycin) and kept in the dark for 2 weeks at 25 °C.

Regeneration

1. The healthy surviving embryogenic calli was transferred into MSKN

regeneration medium and the cultures were kept in the dark for 20 days.

2. The emerging shoots were harvested and transferred again to a fresh

regeneration medium (MSKN) in a Petri dish. The Petri dish was placed in the

light at 27°C with 16 hours photoperiod (110µmol/m2/s) for 5 days.

3. The shoots which grew well were transferred into 25 ml conical flask

containing MSKN regeneration medium. The flask was placed in the light at

27°C with 16 hours photoperiod (110µmol/m2/s) for 10 to 20 days.

4. The healthy shoots were transferred into the rooting medium (MSO).

5. The percentage (%) of callus regeneration frequency (CRF) was calculated as

Equation (4.8.1)

Page 118: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxviii

4.9. EXTRACTION OF TOTAL GENOMIC DNA FROM PLANT TISSUES

FOR MOLECULAR ANALYSIS

4.9.1. OBJECTIVE

The plant genomic DNA was isolated by CTAB method which can be used

for molecular analysis to confirm the presence of the transformed gene.

4.9.2. PRINCIPLE

The extraction of genomic DNA from plant tissues requires the digestion of

cell wall to release the cellular components (Rogers 1994). The nucleases released

from the cellular components had to be inactivated and the genomic DNA has to be

separated from the cellular debris. The cellular contents are released by grinding the

tissue in liquid nitrogen with a motor and pestle. The liquid nitrogen also prevents

nuclease activity. The plant tissue contain large amount of carbohydrates and therefore

DNA is isolated using either SDS or CTAB (Cetyltrimethylammonium bromide).

CTAB protocol was developed by Murray and Thompson (1980) which is used for the

extraction and purification of DNA from plants and it is suitable for the elimination of

polysaccharides and polyphenolic compounds which affect the DNA purity and

quality.

The plant cells are lysed with the ionic detergent CTAB. The CTAB forms

stable and soluble complex with DNA (CTAB-DNA complex) under high salt

concentration. When the salt concentration was reduced to below 0.4 M NaCl causes

Page 119: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxix

the CTAB-nucleic acid complex to precipitate leaving the polysaccharides, phenolics

and other contaminants in the supernatant. Thus the extraction buffer with 2% CTAB

takes care of the release of genomic DNA from the cell content. During extraction

different concentration of CTAB is used which helps in the removal of

polysaccharides. Addition of chloroform: isoamyl alcohol denatures and degrades

protein and helps in the removal of CTAB. NaCl concentration in CTAB solutions is

reduced in successive steps, which aid in DNA precipitation by salting out principle.

EDTA and PVP act as chelating agents. They prevent binding of phenolic compounds

to DNA. Mg+ is a cofactor for nucleases. EDTA binds to Mg+ and prevents nuclease

activity. High salt TE dissolves DNA and CTAB. In 95% ethanol precipitation, DNA

does not dissolve in ethanol and gets precipitated but CTAB gets dissolved and is

removed with the supernantant. 70% ethanol washes and removes the traces of salt and

CTAB.

The isolated genomic DNA was further purified using the PCR clean up kit

and then the amount of DNA was estimated using Biophotometer. DNA had maximum

absorption at 260nm. Optical density of 1 at 260nm corresponds to 50 µg/ml of double

stranded DNA. The ration of O.D 260/280 provides the purity of the DNA. A pure

DNA will have a ratio of 1.8. Ratio less than 1.8 indicates probably the presence of

proteins or other UV absorption. Ratio higher than 2.0 indicates that the sample may

be contaminated with chloroform or phenol and should be precipitated again with

ethanol.

4.9.3. MATERIALS

DNA EXTARCTION

2X CTAB buffer (100ml)

2%CTAB (Cetyltrimethylammonium bromide) (w/v)-(Sigma)

100mM Tris, pH 8.0

Page 120: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxx

20mM EDTA, pH8.0

1.4 M NaCl

1% PVP

CTAB of 2g was dissolved in 70 ml of water. NaCl of 8.18g was added and dissolved.

To the above 10ml of Tris (1M, pH8.0) and 4ml of EDTA (0.5 M, pH 8.0) was added.

The final volume was made up to 99ml with autoclaved Millipore water. The buffer

was autoclaved and then 1g of PVP was added and dissolved.

5% CTAB (25ml)

5% CTAB

0.35M NaCl

CTAB of 1.25g was dissolved in 20 ml autoclaved Millipore water and then 0.5g of

NaCl was added and dissolved. The final volume was made up to 25ml with

autoclaved Millipore water and autoclaved.

CTAB precipitation buffer (100ml)

1%CTAB

50mM Tris, pH 8.0

10mM EDTA, pH8.0

CTAB of 1 g was dissolved in 90ml of autoclaved Millipore water and then 5ml of

Tris (1M, pH8.0) and 2ml of EDTA (0.5 M, pH 8.0) was added. The final volume was

made up to 100ml and autoclaved.

High salt TE buffer (50ml)

10 mM Tris, pH 8.0

1.0 mM EDTA, pH8.0

1 M NaCl

Page 121: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxi

NaCl of 2.922g was dissolved in 40 ml of Millipore water. To this 500 l of Tris (1M,

pH 8.0) and 100 l of EDTA (0.5 M, pH 8.0) was added and the volume was made up

to 50 ml and autoclaved.

0.1X TE (100ml)

1mM Tris pH8.0

0.1mM EDTA pH8.0

100 l of Tris (1M, pH 8.0) and 50 l of EDTA (0.5 M, pH 8.0) was added and the final

volume was made up to 100ml with Millipore water and autoclaved.

PURIFICATION OF DNA FOR PCR

Gel elution Kit (Real Genomics, Hi Yield TM

Gel/PCR DNA MINI KIT-

YDF100/YDF300)

100 mini preps/kit

DF Buffer: 80ml

W1 Buffer: 45ml

Wash Buffer (concentrated):25ml*

Elution Buffer: 6ml

DF Column: 100pcs

2ml Collection Tube: 100pcs

*Initially 100ml of ethanol (96-100%) was added to 25 ml of Wash buffer prior to use

1.5 ml eppendorf tubes

DNase-free pipette tips

4.9.4. METHOD

4.9.4.1. DNA EXTRACTION

Page 122: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxii

1. To one gram of the plant material, liquid nitrogen was added and grinded into fine

powder with chilled sterile motor and pestle.

2. Approximately 100mg of the grinded material was transferred into chilled 2ml

microfuge tubes. The content was transferred using chilled spatula and 500 µl of

hot (65°C) CTAB extraction buffer was added.

3. The tubes were mixed well by inversion and equal volume (700 µl) of ice cold

chloroform: isoamyl alcohol (24:1) was added.

4. The content was mixed well by inversion to form an emulsion and centrifuged for

10 minutes at 13,000rpm in a micofuge. The top aqueous phase was transferred

into new microfuge tubes using cut tips.

5. The volume was measured and 1/5th

volume of 5%CTAB solution was added and

mixed well by gently inverting the tubes.

6. Equal volume of chloroform: isoamyl alcohol (24:1) was added and mixed

thoroughly to form an emulsion and centrifuged at 13,000 rpm for 10 minutes.

7. The aqueous phase was transferred into fresh 1.5ml microfuge tubes using cut tips

and equal volume of CTAB precipitation buffer was added and mixed gently by

inversion.

8. The tubes were centrifuged at 10,000rpm for 1 minute. The supernatant was

discarded and the pellet was dissolved in 200 µl of high salt TE buffer by gently

tapping the tube.

9. If the DNA was not dissolved, the tubes were kept in water bath at 65 °C for 10

minutes and the content was centrifuged at 13,000rpm for 10 minutes and the

supernatant was transferred into a fresh microfuge tube. High salt TE buffer of 100

µl was added again, if the pellet was not dissolved. Then the contents were

centrifuged at 13,000 for 10 minutes and the supernatant were pooled together.

10. To the supernatant 2.5 volumes of ice cold 95% ethanol was added to the solution

and mixed gently by inversion and centrifuged for 15minutes at 13,000rpm at 4°C.

Page 123: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxiii

11. The supernatant was discarded and to the pellet 70% ethanol was added up to the

original volume and centrifuged at 13,000rpm for 10 minutes at 4°C.

12. The supernatant was discarded and the pellet was dried. The pellet was then

dissolved in 50 µl of 0.1X TE or sterile water.

4.9.4.2. PURIFICATION OF DNA FOR PCR

Sample preparation

Extracted DNA of 100 µl was transferred into 1.5 ml microcentrifuge tube.

DF buffer of 5 volumes were added to 1 volume of the sample and mixed by

vortexing.

DNA Binding

DF column was placed in a 2 ml collection tube.

The sample mixture from step 2 was added into DF Column.

The column was centrifuged at full speed approximately 13, 000 rpm for 30

seconds.

The flow through was discarded and the DF Column was placed back into the 2

ml collection Tube. When the sample was more, this step was repeated.

Wash

W1 Buffer of 600 µl was added into the DF Column and allowed to stand

for 1 minute.

The column was centrifuged at full speed approximately13, 000 rpm for 30

seconds.

Page 124: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxiv

The flow through was discarded and the DF column was placed back into

the 2 ml collection Tube.

The column was centrifuged at full speed approximately13, 000 rpm for 3

minutes to dry the column matrix.

DNA Elution

The dried DF column was transferred into a new 1.5ml microcentrifuge

tube.

Elution buffer or TE of 20 µl to 50 µl was added into the center of the

column matrix.

The elution buffer was allowed to stand for 2 minutes until the buffer is

absorbed by the matrix.

The column was centrifuged at full speed for 2 minutes to elute the purified

DNA

The eluted DNA of 2 µl was diluted with 48 µl of sterile water and the

quantity was measured using biophotometer.

4.9.4.3. ESTIMATION OF DNA

To measure the amount of DNA, the purified samples were diluted using

sterile water and measured using Biophotometer.Sterile water was used as blank.

The biophotometer displays the amount of DNA present in the samples

Page 125: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxv

4.10. MOLECULAR ANALYSIS OF PUTATIVE TRANSFORMED PLANTS

BY POLYMERASE CHAIN REACTION (PCR)

4.10.1. OBJECTIVE

Molecular analysis of transformed plant was carried out by detecting the

presence of CaMV35S promoter gene to confirm gene transformation.

4.10.2. PRINCIPLE

Page 126: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxvi

The most common strategy employed for screening transgene is PCR-based

detection of transgene followed by gel electrophoresis and compared with the markers.

PCR is a technique used to amplify a specific DNA sequence into millions of copies.

The enzyme Taq polymerase (94Kb) isolated from Thermus aquaticus is used in this

technique which is thermostable and resistant to denaturation by heat treatment and

active at 75°C to 80 °C. This enzyme is used to amplify minute quantities of transgene

DNA to a detectable level on 1% agarose gel. PCR based detection strategy is

extremely sensitive.

A primer is a short single stranded oligonucleotide which when attached to

a single stranded template molecule acts as a starting point for complementary strand

synthesis directed by DNA polymerase enzyme. PCR amplification involves two

oligonucleotide primers that flank the DNA sequence to be amplified. The primers

hybridize to the opposite strand of the target sequence such that the DNA synthesis by

the polymerase enzyme proceeds across the region between the primers effectively

doubling the amount of that DNA sequence. Since the extension products are also

complementary and capable of binding to the primers, the cycle is repeated after

denaturation step. By repeated cycles of denaturation, priming and extension there is a

rapid exponential accumulation of the specific target fragment. Thus after 10 cycles

the target sequence would be amplified 1000 folds.

Steps involved in PCR

The technique involves repeated cycles of DNA synthesis which is based on three

simple steps for amplification reactions (Mullis 1987). The three steps involved are

Denaturation of the template DNA into single strands

Annealing of primers

Extension of the new DNA strands

Page 127: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxvii

The three major steps are repeated for 30 to 40 cycles. This is carried out on an

automated thermo cycler, which can heat and cool the tubes containing the reaction

mixture in a very short time.

DNA Denaturation

During the denaturation, the double strand DNA is denatured into a single

stranded DNA and the enzymatic reaction are halted due to the high temperature. DNA

denaturation also eliminates the secondary structures which may interrupt DNA

amplification. This step is carried out from 94 C to 95 C for 1 minute. The

temperature depends upon various factors like nature of DNA, G-C content, secondary

structures etc. An initial denaturation of template DNA was carried out at 94 C to

95 C for 5 to 10 minutes for complete strand separation at the first PCR step.

Primer Annealing

The primers that are added to the reaction mixture keep moving due to

Brownian movement. Ionic bonds are constantly formed and broken between the

single stranded primer and the single stranded template. The stability of the bonds

increases when specific primers bind to the single stranded template and on this double

stranded region formed by the hybridization of the primer and specific nucleotide

sequence, the polymerase enzyme attaches and starts copying the template. Once few

bases are added to the template DNA, the ionic bond is so strong between the template

and the primer that it does not break. The annealing temperatures should be 5 C less

than the melting temperature Tm. The Tm of primers containing 18 to 24 base pairs

can be determined as in Equation (4.10.1). Higher annealing temperature result in no

amplification and lower annealing temperature results in non specific amplification.

Primer Extension

Page 128: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxviii

Primer extension results in synthesis of new strands usually carried out at

72 C. This is the ideal working temperature for the taq polymerase enzyme. The

primers, where the few bases are added to the strand, already have a strong ionic

attraction to the template DNA than the forces breaking these attractions. Primers that

are non specific for the template strand does not result in extension of the strand. For

every 1Kb target DNA, 1 minute is recommended.

b) Factors affecting amplification

Efficient amplification of target sequences depends on individual reaction compound,

time and temperature conditions of PCR reactions.

Sample volume

Sample volume of about 20 l, 50 l, 100 l volumes are recommended for PCR as it

assesses the adequate thermal equilibrium of reaction mixtures.

Template DNA

The purity and concentration of the template DNA affects the product yield

and the specificity of the PCR. Higher amount of template DNA may result in non

specific amplification or it will inhibit PCR amplification. A number of contaminants

such as SDS, phenol and other reagents used in template DNA preparation can inhibit

PCR.

Primers

It is one of the most important factors affecting the quality of PCR. The

primers usually used at equal concentration in the range of 0.1 to 1 M. Primers should

be free of complementary sequence at the 3’ end as it results in primer dimmer

formation which in turn reduces product yield. Typically 40 to 60 % of GC content is

recommended to avoid the formation of primer dimmer.

Page 129: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxix

Deoxy Nucleotide Tri Phosphates (dNTP’s)

Deoxy Nucleotide Tri Phosphate (dNTP) is the major source of phosphate

group in the reaction mixture. The final concentration of dNTP’s in the standard

amplification reaction mixture should be 200 M.

Taq DNA polymerase enzyme

Taq DNA polymerase lacks 3’5’ exonuclease activity but has 5’3’

exonuclease activity and 5’3’ polymerase activity. For most amplification 1.5– 2.0

units of enzyme is recommended. But higher enzyme concentration leads to non

specific amplification.

4.10.3. MATERIALS

The PCR ingredients and Primers were procured from Eppendorf, Germany and

Operon technologies, Germany. The sequence details of the primer for the detection of

CaMV35S promoter gene is provided below.

S.No. Primer Sequence

1

2

35s FP

35s RP

5’GCTCCTACAAATGCCATCA 3’

5’GATAGTGGGATTGTGCGTCA 3’

Sequence details of primers of CaMV35S

Negative control 1-Reaction mixture without the DNA sample

Page 130: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxx

Negative control 2- DNA isolated from regenerated plant from untransformed callus

Positive control- Restricted, eluted and purified pCAMBIA 1301(linearized plasmid-

eluted and purified using HiYield Gel/PCR DNA Mini kit (YDF100/YDF300)

4.10.4. METHOD

The following Reaction mixture was added in each tube in the same order

(Table 4.10.1).

S. No Reaction mixture Concentration Volume

1 10x Reaction buffer 1X 2.0 µl

2 MgCl2 5mM 2.0 µl

3 dNTP's 0.2mM 0.4 µl

4 Forward Primer 0.25mM 0.5 µl

5 Reverse Primer 0.25mM 0.5 µl

6 MilliQ water - 11.3 µl

7 DNA 20ng/ml 3.0 µl

8 Taq polymerase 1.5U 0.3 µl

Table 4.10.1: Reaction mixture for PCR setup

A total reaction volume of 20µl reaction was set.

The Taq Polymerase was added last to the tube, vortex and spinned briefly,

and then the reaction mixture was carefully layered with 8.0µl of sterile

mineral oil on top of the reaction to prevent evaporation.

These reactions were set up in the sterile hood to avoid any extraneous DNA

contamination from the environment.

Page 131: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxxi

The thermal cycler was programmed with times and temperatures for

denaturing, annealing and extending.

Initial denaturation was carried out at 94 C for 13minutes. The cycles were

carried out with the following characteristics.

a. Denaturation 94 C for 30s

b. Annealing 54 C for 30s

c. Extension 72 C for 30s

d. No. of cycles 40

Final extension of 7 min was set for extending temperature. Then at 4oC

until PCR machine was shut off.

When program was completed, the tubes containing the reaction mixture

were taken out and 5 l of gel loading dye was added.

The final PCR product was run on a 1% agarose gel with markers to

determine size of product.

Page 132: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxxii

CHAPTER – 5

RESULTS AND

DISCUSSIONS

Page 133: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxxiii

CHAPTER 5

RESULTS AND DISCUSSIONS

5.1. GENE CLONING USING VECTOR NTI SOFTWARE

The software analysis of cloning was successfully carried out using vector

NTI software 11.5 advanced versions. From the software analysis, it was clear that the

pCAMBIA 1301 and the modified gene of interest attached with the NcoI and pmlI

gene sequence at 5’ and 3’end, can be restricted with NcoI and PmlI restriction

enzymes for cloning. Both the plasmid and gene of interest (HCC-scFv) restricted with

the enzyme NcoI and PmlI are ligated to form a clone using this software. The results

of the cloning experiment are shown from Figures 5.1.1 to Figure 5.1.2. The results

from the software analysis (Figure 5.1.7 and Figure 5.1.8) clearly illustrate the

recombinant map and gene sequence which will be obtained after the cloning

experiments.

The Figure 5.1.1 shows the result of anti-HCC scFv antibody gene

sequence downloaded from the Gen bank which was used in vector NTI software for

cloning analysis. The gene map of the downloaded anti-HCC scFv antibody obtained

from the software analysis was show in the Figure 5.1.2. Similarly the results of

modified anti-HCC scFv antibody gene sequence and map are shown in Figure 5.1.3

and Figure 5.1.4 respectively. The results of the gene sequence and the map of the

plasmid vector pCAMBIA 1301 are shown in Figure 5.1.5 and Figure 5.1.6.

Page 134: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxxiv

The cloning results obtained from the software analysis using the gene sequence of the

plasmid vector pCAMBIA 1301and the modified anti-HCC scFv antibody are shown

in Figure 5.1.7 and Figure 5.1.8.

Page 135: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxxv

5.1.1. Gene sequence of anti-HCC scFv antibody gene: AY686498

Figure 5.1.1: Analysis of Gene sequence of HCC-scFv: AY686498 downloaded

from the Gen Bank using vector NTI software

5.1.2. Gene Map of anti-HCC scFv antibody gene: AY68649

Page 136: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxxvi

Figure 5.1.2: Analysis of Gene Map of HCC-scFv: AY686498 downloaded

from the Gen Bank using vector NTI software

5.1.3. Modified gene sequence of anti-HCC scFv antibody gene: AY686498 with

NcoI and PmlI restriction sites

Figure 5.1.3: Modified Gene sequence of HCC-scFv: AY686498 downloaded from

the Gen Bank using vector NTI software

Page 137: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxxvii

5.1.4. Modified gene Map of anti-HCC scFv antibody gene: AY686498 with NcoI

and PmlI restriction sites

Figure 5.1.4: Modified Gene Map of HCC-scFv: AY686498 downloaded from the

Gen Bank using vector NTI software

5.1.5. Gene sequence of pCAMBIA 1301-T-DNA vector

Page 138: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxxviii

Page 139: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxxxix

Page 140: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxl

Page 141: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxli

Page 142: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxlii

Page 143: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxliii

Page 144: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxliv

Figure 5.1.5: Analysis of Gene sequence of pCAMBIA 1301 downloaded from the

Gen Bank using vector NTI software

Page 145: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxlv

5.1.6. Gene map of pCAMBIA 1301-T-DNA vector

Figure 5.1.6: Analysis of Gene Map of pCAMBIA 1301 downloaded from the Gen

Bank using vector NTI software

Page 146: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxlvi

5.1.7. Cloning of anti HCC-scFv into pCAMBIA 1301

Page 147: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxlvii

y

Figure 5.1.7: Gene Map: Cloning of pCAMBIA 1301 and HCC-scFv to form

pCAMBIA 1301-HCC-scFv using vector NTI software

5.1.8. Gene sequence of cloned HCC-scFv into pCAMBIA 1301

Page 148: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxlviii

Page 149: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxlix

Page 150: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cl

Page 151: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cli

Page 152: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clii

Page 153: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cliii

Page 154: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cliv

Page 155: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clv

Figure 5.1.8: Gene Sequence: Cloning of pCAMBIA 1301 and HCC-scFv to form

pCAMBIA 1301-HCC-scFvusing vector NTI software

5.2. SYNTHESIS OF HCC-scFv GENE-GENEART

From the results of vector NTI software analysis, the sequence was

submitted to Geneart for gene synthesis. The gene was synthesized, sequenced and

confirmed for the identity of sequence. The percentage of identity of the synthesized

sequence was found to be 100%. The synthesized gene was cloned into pMA-T

containing ampicillin resistance marker and it was received in lyophilized form. These

plasmids were transformed into E. coli DH5α by calcium chloride transformation. The

images from Figure 5.2.1 to Figure 5.2.6 show the results of HCC-scFv gene

synthesis.

The results of the G+C content of the gene sequence analyzed using Gene

Optimizer® was seen in a 40 bp window centered at the indicated nucleotide position.

The average GC content was found to be 54% shown in the Figure 5.2.1, which

indicates the stability of the gene sequence. The Figure 5.2.2 shows the gene sequence

of both the strand used for the synthesis of the gene. The synthesized gene was

sequenced using gene sequencer and the results are shown in Figure 5.2.3. The gene

sequence obtained from the sequencer were compared with the given sequence

(submitted to Geneart) and found to be 100% similar (Figure 5.2.4). The results of

gene sequence and map of the anti-HCC-scFv cloned into pMA-T plasmid vector were

shown in Figure 5.2.5 and Figure 5.2.6.

Page 156: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clvi

5.2.1. Gene sequence analyzed with GeneOptimizer®

The analysis of GC content of the sequence using GeneOptimizer is shown

in the Figure 5.2.1, a 40bp window centered at the indicated nucleotide position. The

average GC content was about 54%.

Page 157: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clvii

Figure 5.2.1: Plot showing G+C content

5.2.2. Gene sequence analysis showing both the strand to be synthesized

Page 158: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clviii

Page 159: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clix

Page 160: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clx

Figure 5.2.2: Gene sequence of both the strands for gene synthesis.

Page 161: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxi

5.2.3. Analysis of synthesized Gene sequence using Gene sequencer

Page 162: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxii

Page 163: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxiii

Page 164: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxiv

Figure 5.2.3: Gene sequenced from the synthesized gene.

5.2.4. Comparitative analysis of the provided gene sequence with the

synthesized gene sequence

Page 165: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxv

Page 166: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxvi

Page 167: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxvii

Figure 5.2.4: Sequence identity of the synthesized gene sequence and the given

sequence.

5.2.5. Gene sequence of synthesized gene (HCC-scFv) cloned into pMA-T with

restriction sites.

Figure 5.2.5: Gene sequence of HCC-scFv gene cloned into plasmid pMA-T (pMA-T-

HCC-scFv) with NcoI and PmlI restriction sites.

Page 168: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxviii

5.2.6. Gene construct of HCC-scFv cloned into pMA-T with restriction enzyme

Page 169: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxix

Figure 5.2.6: Gene Map of pMA-T containing HCC-scFv with various restriction

sites.

5.3. Transformation of plasmid pCAMBIA 1301 and pMA-T into E. coli DH5α

and screening of transformed E. coli DH5α strain

The plasmid pCAMBIA 1301 and pMA-T was successfully transformed

into E. coli (DH5α) by calcium chloride transformation. In case of pCAMBIA 1301

the transformed cells were initially screened on kanamycin antibiotic LB medium. The

transformation efficiency was good which was calculated from the number of colonies

obtained on the plate. The plasmid which was transformed into E. coli (DH5α) carried

kanamycin resistant gene kan and grew on the LB medium containing kanamycin

sulfate (an aminoglycoside antibiotic) shown in the Figure 5.3.1 and Table 5.3.1. The

plates were compared with the controls and the presence of Lac Z gene was also

confirmed using X-gal and IPTG (Figure 5.3.2 and Table 5.3.2). The Lac Z gene

produces β-galactosidase, a stable enzyme which can be easily assayed using the

chromogenic substrate X-gal (5-bromo-4-chloro-indolyl- β-D-galactoside or

galactopyranoside). The X-gal is colourless and when the galactoside moiety is

cleaved off by β-galactosidase, 5-bromo-4-chloro-indolyl is formed and in the

presence of air they form 5-bromo-4-chloro-indigo (blue colour). This colour

formation was observed on the plates indicating that the strains have been transformed

with pCAMBIA 1301 containing lac Z gene.

Page 170: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxx

Similar to pCAMBIA 1301, the plasmid PMA-T (HCC-scFv) was also

transformed into E. coli (DH5α) by calcium chloride transformation and screened on

amplicillin antibiotic containing LB media. The Figure 5.3.3 and Table 5.3.3 shows

the transformation of pMA-T into E. coli (DH5α). The pMA-T contain ampicillin

resistance gene (amp) that codes for β lactamase, an enzyme that cleaves the β lactam

ring of penicillin and related antibiotics. This enzyme hydrolyzes the amide bond of

the β lactam ring to produce penicilloic acid, which does not show any antibiotic

activity. The E. coli strains transformed with pMA-T produces β lactamase enzyme

and it was secreated into the periplasmic space of the bacterium, where the enzyme

hydrolyzes the β lactam ring and detoxified the ampicillin in the LB media and grew

on the plate.

5.3.1. Transformation of pCAMBIA 1301 and Screening of competent cells

transformed with pCAMBIA 1301 on LB medium (kanamycin)

S.No of Plates

Competent

cells

Concentration

of

antibiotics

E. coli cells

screened for

pCAMBIA

1301

Result

A (positive

control)

200µl

No antibiotics

Untransformed

E. coli DH5α

cells

Lawn of E.

coli DH5α

B (negative

control)

200µl

Kanamycin

(50 µg/ml)

Untransformed

E. coli DH5α

cells

No growth

was

observed

C (experiment 1)

200µl

Kanamycin

(50 µg/ml)

Transformed E.

coli DH5α cells

with pCAMBIA

1301

Transforme

d colonies

grew on the

plate

D (experiment 2)

200µl

Kanamycin

(50 µg/ml)

Transformed E.

coli DH5α cells

with pCAMBIA

1301

Transforme

d colonies

grew on the

plate

Page 171: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxi

Table 5.3.1: Screening of the transformed E. coli containing pCAMBIA 1301 on

LB medium containing kanamycin.

Page 172: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxii

Figure 5.3.1: Transformation of pCAMBIA1301 into E. coli DH5α and screening

of the transformed cells on LB medium containing Kanamycin antibiotic.

5.3.2. Screening of transformed E. coli DH5α with pCAMBIA 1301 on LB

medium (kanamycin+X-gal +IPTG)

2

• Transformed cells were screened on antibiotic LB

medium ( - kan, +kan)

A. Untransformed cells-

on LB medium without

Kanamycin

B. Untransformed cells-

on LB medium with

Kanamycin

C and D. Transformed

cells - on LB medium

with Kanamycin

A B

C D

Page 173: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxiii

S. No of Plates

Concentration

of

Antibiotics in

LB medium

E. coli screend for

lac Z gene in

pCAMBIA 1301

Result

A (positive

control)

No antibiotics

Untransformed E.

coli DH5α cells

No bluish green

colonies were

observed

B (negative

control)

Kanamycin

(50 µg/ml)

+

X-gal+IPTG

Untransformed E.

coli DH5α cells

No growth was

observed on the

plate

C (experiment 1)

Kanamycin

(50 µg/ml)

+

X-gal+IPTG

Transformed E. coli

DH5α cells with

pCAMBIA 1301

screened on

kanamycin

Bluish green

colonies were

observed

D (experiment 2)

Kanamycin

(50 µg/ml)

+

X-gal+IPTG

Transformed E. coli

DH5α cells with

pCAMBIA 1301

screened on

kanamycin

Bluish green

colonies were

observed

Table 5.3.2: Screening of the transformed E. coli containing pCAMBIA 1301 on

LB medium containing -kanamycin+X-gal +IPTG.

Page 174: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxiv

Figure 5.3.2: Screening of the E. coli DH5α transformed with pCAMBIA1301 on

LB medium containing Kanamycin, X-gal and IPTG.

X-gal confirmation of transformed E.coli DH5α

• A- non transformed

E.coli DH5α

(white colonies)

• B- no growth

• C- transformed E.coli

DH5α ( blue colonies)

• D- transformed E.coli

DH5α ( blue colonies)

A B

C D

Page 175: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxv

5.3.3. Transformation of pMA-T:

Plate no Competent

cells

Antibiotics E. coli cells

screened for HCC-

scFv-pMA-T

Result

1 (positive

control)

Competent

cells

200µl

No

antibiotics

No plasmid Lawn of E.

coli DH5α

2 (negative

control)

Competent

cells

200µl

Ampicillin

(100µg/ml)

No plasmid Plate

showed no

growth

3 (experiment 1) Competent

cells

200µl

Ampicillin

(100µg/ml)

Plasmid (HCC-

scFv-pMA-T)

Transforme

d colonies

grew on the

plate

4 (experiment 2) Competent

cells

200µl

Ampicillin

(100µg/ml)

Plasmid (HCC-

scFv-pMA-T)

Transforme

d colonies

grew on the

plate

Table 5.3.3: Screening of the transformed E. coli containing pMA-T-HCC-scFv

on LB medium containing ampicillin.

Page 176: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxvi

Figure 5.3.3: Transformation of pMA-T-HCC-scFv into E. coli DH5α and

screening of the transformed cells on LB medium containing ampicillin antibiotic.

E. coli DH5α transformed with pMA-T-HCC-scFv

Page 177: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxvii

.

5.4. Isolation of plasmid pCAMBIA 1301 and pMA-T from transformed E. coli

DH5α and confirmation by Gel electrophoresis

The plasmid pCAMBIA 1301 and pMA-T was isolated from the

transformed E. coli DH5α and confirmed by gel electrophoresis. The image of the

DNA banding pattern in agrose gel clearly indicates the presence of the plasmid

pCAMBIA 1301 (Figure 5.4.1). The bands formed by pCAMBIA 1301 and pMA-T

on 0.8% agarose gel were shown in the Figure 5.4.2. These bands indicate that the

plasmids have been transformed into the E. coli DH5α cells and the colonies selected

on antibiotic media were found to be transformed with the respective plasmids.

Page 178: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxviii

5.4.1. Gel image of pCAMBIA 1301

ISOLATION OF TRANSFORMED PLASMID PCambia 1301 FROM

E.coli DH5α

• Lane 1-No sample

• Lane 2-5- pCambia 1301

• Lane 6-10-untransformed E.coli DH5α

1 2 3 4 5 6 7 8 9 10 1 2 3 4 5 6 7 8 9 10

Nicked plasmidSupercoiled plasmid

Page 179: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxix

Figure 5.4.1: Plasmid isolated from transformed E. coli DH5α containing

pCAMBIA 1301.

5.4 2. Gel image of pCAMBIA 1301 and pMA-T (HCC-scFv)

Plasmid isolation from E.coli DH5α transformed with pCambia1301 and pMA-T

• Lane 1-marker

• Lane 2&3 -pCambia 1301 without RNase treatment, Lane 4&5- pCambia 1301 treated with RNase

• Lane 6,7-pMA-T(HCC-scFv) without RNase treatment,Lane 8,9-pMA-T (HCC-scFv) treated with RNase

1 2 3 4 5 6 7 8 9 10 1 2 3 4 5 6 7 8 9 10

10Kb

8Kb

6Kb

5Kb

4Kb

3Kb

2Kb

1Kb

Page 180: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxx

Figure 5.4.2: Plasmid isolated from transformed E. coli DH5α (pCAMBIA 1301

and pMA-T-HCC-scFv).

5.5. CLONING OF HCC-scFv FROM pMA-T PLASMID INTO pCAMBIA 1301

5.5.1. Restriction digestion of pCAMBIA 1301 and pMA-T for cloning

The plasmid pCAMBIA1301 and pMA-T (HCC-scFv) was restricted with

NcoI and PmlI restriction enzymes and confirmed on 0.8% agarose gel. From the gel

images it was clear that the restriction digestion of pCAMBIA 1301 by NcoI and PmlI

restriction enzymes formed two bands, shown in the Figure 5.5.1 from lane 2 to 5.

The size of one fragment was about 10kb and another fragment was about 2Kb. The

fragment size of 10Kb was excised from the gel and eluted using gel elution kit for

ligation reaction. This fragment contains the CaMV 35S promoter, hpt marker gene

and Kanamycin antibiotic marker. The 2Kb fragment contain the gus reporter gene,

which was not used in this experiment.

Similarly from the gel images of the restriction digestion of pMA-T (HCC-

scFv) by NcoI and PmlI restriction enzymes, two bands were formed as shown in the

Figure 5.5.1 from lane 6 to 9. One fragment was about 1Kb and another fragment was

about 2.5 Kb. The fragment size of 1Kb was excised from the gel and eluted using gel

Page 181: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxxi

elution kit for ligation reaction. This excised fragment contains the gene of interest

(HCC-scFv).

During ligation reaction the 1 Kb fragment of HCC-scFv was ligated with

the 10Kb fragment of pCAMBIA 1301. The 2.5 Kb fragment contains the remaining

plasmid of pMA-T vector. In this experiment pCAMBIA 1301 and pMA-T was

successfully restrict digested with NcoI and PmlI restriction enzymes.

Page 182: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxxii

Figure 5.5.1: Restriction digestion of pCAMBIA 1301 and pMA-T (HCC-scFv)

with NcoI and PmlI restrcyion enzymes

5.5.2. Ligation of HCC-scFv and pCAMBIA 1301

Restriction digestion of pCambia 1301 and pMA-T(HCC-scFv) with NcoI and PmlI

• Lane 1-Marker1Kb

• Lane 2-5- pCambia 1301 restricted with NcoI and PmlI

• Lane 6-9-pMA-T (Hcc-scFv) restricted with NcoI and PmlI

1 2 3 4 5 6 7 8 9 10

10Kb

8Kb5Kb

1Kb

2Kb

3Kb

4Kb

1 2 3 4 5 6 7 8 9 10

Page 183: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxxiii

5.5.2.1. Ligation and Transformation of the sample

The fragment size of 9.8020Kb from pCAMBIA 1301 and 0.946Kb of the

Insert DNA was eluted from 0.8% agarose gel by gel elution kit. The eluted linearized

vector DNA- (pCAMBIA 1301-9.802 kb) and the Insert DNA - ( 3:1 molar ratio

excess over vector)-(HCC-scFv-0.946 kb) was efficiently ligated at 22°C (+ 1°C)

using 5X Rapid ligation kit containing T4 DNA Ligase (5U/µl). The ligated samples

were successfully transformed into E. coli DH5α by calcium chloride transformation.

The colonies of E. coli DH5α transformed with ligated vector and DNA insert was

selected on the LB medium containing Kanamycin and the results are shown in the

Figure 5.5.2.1.

5.5.2.2. Confirmation of the ligated clone

The transformed E. coli selected on the kanamycin antibiotic plates were

subcultured by streaking. The plasmid DNA was isolated from the transformed E. coli

cells to confirm the presence of cloned plasmid DNA by alkaline lysis method. The

isolated plasmid was restricted with the same restriction enzymes NcoI and PmlI and

confirmed on 0.8% agarose gel. The agarose gel image in the Figure 5.5.2.2 clearly

shows, the presence of two fragments (9.802 kb and 0.945 kb) in lane 4 confirming

the presence of cloned DNA (pCAMBIA 1301- HCC-scFv).

Page 184: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxxiv

Transformation of ligated samples

• Transformed plate 5 (experiment 1) • Transformed colonies were streaked on to the LB

plate containing antibiotic

Figure 5.5.2.1: Transformation of the ligated pCAMBIA 1301 and pMA-T (HCC-

scFv) into E. coli DH5α and screened on LB medium containing Kanamycin.

Page 185: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxxv

Figure 5.5.2.2: Confirmation of the transformed E. coli DH5α containing ligated

pCAMBIA 1301 and pMA-T (HCC-scFv)

Confirmation of the ligated clone

• Lane1: 1 Kb marker

• Lane 2: restricted plasmid (pCambia 1301)- eluted from the gel

• Lane 3: restricted HCC-scFv fragment- eluted from the gel

• Lane 4: restricted pCambia 1301-HCC-scFv(isolated from the transformed ligated sample)

1 2 3 4

10Kb

8Kb

6Kb5Kb

4Kb

3Kb

2Kb

1Kb

Page 186: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxxvi

5.6. Transformation of pCAMBIA 1301-scFv from E. coli DH5 α into

Agrobacterium tumefaciens EHA105

The plasmid containing the gene of interest (pCAMBIA 1301-HCC-scFv)

was successfully transformed into Agrobacterium tumefaciens EHA 105. The donor E.

coli DH5α strain harboring Ti plasmid (pCAMBIA1301-HCC-scFv) was transformed

into the recipient strain Agrobacterium tumefaciens EHA 105 with the help of conjugal

helper E. coli HB101 strain harboring pRK2013. The Ti plasmid was mobilized from

donor E. coli DH5α strain into the recipient strain Agrobacterium tumefaciens EHA

105 due to the mobilization function of pRK2013. Agrobacterium tumefaciens EHA

105 harboring the transformed gene was selected on AB minimal medium containing

kanamycin and rifampicin antibiotic. The Ti plasmid contain kanamycin resistant gene

which was used as a marker to screen the transconjugants. Only the transformed

Agrobacterium grew on kanamycin antibiotic selection medium. In this experiment the

plasmid pCAMBIA1301-HCC-scFv from the E. coli DH5α was transformed into

Agrobacterium tumefaciens EHA 105 by triparental mating. The results of triparental

mating are shown in the Figure 5.6.1 and Figure 5.6.2. These transformed

Agrobacterium tumefaciens EHA 105 were used for Agrobacterium-mediated

transformation, to transfer gene into IR64 rice variety.

Page 187: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxxvii

Figure 5.6.1: Transformation of pCAMBIA 1301-HCC-scFv into A.tumefaciens EHA

105 by triparental mating and screening on AB minimal medium containing

Kanamycin antibiotic

TRANSFORMATION OF pCAMBIA 1301-HCC-scFv INTO AGROBACTERIUM

EHA 105 BY TRIPARENTAL MATING

• Agrobacterium EHA 105 selected on

rifampicin medium

• Transformed Agrobacterium EHA 105

selected on AB minimal medium with

Rifampicin and kanamycin antibiotic

Page 188: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxxviii

Transformation of pCambia 1301-HCC-scFv into Agrobacterium

tumefaciens EHA 105 by Triparental mating

E.coli DH5α-

(pCambia 1301-

HCC-scFv)

E.coli HB101

(pRK2013)

Agrobacterium tumefaciens EHA 105

TRIPARENTAL

MATING

Transformed A.tumefaciens EHA 105

Page 189: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

clxxxix

Figure 5.6.2: Transformation of pCAMBIA 1301-HCC-scFv into A.tumefaciens EHA

105 by triparental mating.

5.7. Callus Induction of IR64 rice variety for transformation

White soft embryogenic callus developed on the surface of the rice scutellar

region after 2 to 3 weeks. The high concentration of 2, 4-D (2mg/L) induced the

formation of callus (undifferentiated mass of cells) and inhibited the formation of

roots. The seeds obtained from ICAR and callus obtained after 2 to 3 weeks are shown

in the Figure 5.7.1.

The phenomenon of the reversion of mature cells to the meristematic state

leading to the formation of callus is called dedifferentiation. These dedifferentiated

cells have the capacity to form a whole plant, a phenomenon described as

redifferentiation. These two phenomenons of dedifferentiation and redifferentiation are

described as cellular totipotency, which is seen only in plants and not in animal cells.

These dedifferentiated cells (callus) can be transformed with the gene of interest by

Agrobactetium – mediated transformation and they can be regenerated.

In this experiment callus has been successfully induced from IR64 rice

variety. The callus induced from IR64 rice variety can be used for Agrobacterium

mediated transformation to transfer the HCC-scFv gene.

Page 190: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxc

Page 191: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxci

Figure 5.7.1: Callus induction of IR4 using 2, 4 D-MS medium

5.8. Transformation of pCAMBIA 1301-HCC-scFv from Agrobacterium

tumefaciens EHA 105 into callus by Agrobacterium- mediated transformation

Callus induction of Orzae sativa –Indica rice variety- IR 64

callus

IR 64 obtained

from ICAR,

Cuttack,India

Callus induction on 2,4 D

MS medium

Callus obtained on 2,4 D MS

medium

Page 192: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxcii

The callus which was infected with Agrobacterium was selected on

first selection medium using cefotaxime. The surviving calli were further seleceted on

second selection medium containing cefotaxime and hygromycin B. The image of

third round of selection using cefotaxime and hygromycin B is shown in the Figure

5.8.1. The healthy surviving embryogenic calli was transferred into MSKN

regeneration medium. The image of regeneration has been shown in the Figure 5.8.1.

Cefotaxime was used to prevent contamination and kill bacteria. The callus

which was transformed with the Agrobacterium gene carrying pCAMBIA1301-HCC-

scFV containing the hpt gene which codes for hygromycin phosphotransferase

breakdown the aminocyclitol antibiotic hygromycin B in the substrate and grow on the

media. In the absence of transformation of the gene, the callus cannot break the

aminocyclitol antibiotic hygromycin B in the substrate due to lack of hpt gene and

thus, the callus does not survive on the media.

In this experiment, the gene of interest (pCAMBIA1301-HCC-scFV) was

successfully transformed from Agrobacterium tumefaciens EHA 105 into callus of IR

64 and it was successfully regenerated.

Page 193: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxciii

Figure 5.8.1: Transformation of pCAMBIA 1301-HCC-scFv from A.tumefaciens

EHA 105 into IR64 and selection of the transformed callus on Hygromycin.

5.9. Extraction of total genomic DNA from plant tissues for Molecular Analysis

Transformation of pCambia 1301-HCC-scFv from Agrobaerium tumefaciens

EHA 105 into callus of IR 64 and regeneration

• Third round of calli selection on

cefotaximine and hygromycin

• Regeneration of callus

Page 194: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxciv

The total genomic DNA from transformed and untransformed leaf samples

were extracted using CTAB method. A white precipitate was obtained which was

dissolved in sterile water and stored at -20°C. The amount of DNA obtained was

measured using Biophotometer. The organic extraction is a conventional technique

and the solvents are used to extract the contaminants from the cell lysate. The cells

were lysed by CTAB-detergent and then mixed with phenol, chloroform and isoamyl

alcohol. The concentration of salt and the pH used during the extraction must be

correct to ensure that contaminats are separated into the organic aqueous and the DNA

remains in the aqueous phase. DNA is precipitated and recovered by alcohol

precipitation. This technique is time consuming and the isolated DNA may contain

residual phenol or chloroform which might inhibit the downstream processing of the

PCR. Therefore purification of the DNA is required for further downstream

processing.

The isolated DNA was purified using the PCR clean up kit and the amount

of DNA obtained was measured using Biophotometer. Purification of the sample was

carried out to remove any residual components left behind after DNA isolation. The

purified samples were used for the detection and confirmation of CaMV35S promoter

gene sequence using PCR.

Page 195: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxcv

5.10. Molecular analysis of Putative transformed plants by Polymerase Chain

Reaction (PCR)

The sequence of the forward primer (FP-19bp long- CaMV35S) and the

reverse primer (RP-20bp long- CaMV35S) are specific for CaMV35S promoter gene

sequence, present upstream of the cloned pCAMBIA 1301-HCC-scFv gene. These

primers would bind specifically to the CaMV35S promoter gene sequence region of

the template DNA based on the homology and would help in the amplification of the

amplicon (195bp). As the result of amplification a minute concentration of template

DNA would get amplified to 1000 folds at the end of 30 cycles.

From the agarose gel image A (Figure 5.10.1), negative control 1 did not

show any band indicating the purity of water and the reaction mixture. The negative

control 2 did not show any band indicating the absence of the CaMV35S gene in

untransformed control plant. The positive control in lane 4 and 5 showed a thick band

indicating the presence of CaMV35S promoter gene and proves that the reaction

conditions were perfect. The lanes from 6 to 10 showed thick band indicating the

presence of CaMV35S in the transformed and regenerated plant. All the samples

showed amplification of CaMV35S.

Similar results were seen in the Agarose gel image B (Figure 5.10.1), Lane

10 showed the marker band, Lane 6 and 7 showed the amplification of the positive

control, Lane 8 and 9 did not show any amplification indicating untransformed

regenerated plant and Lane 1 to 5 showed the amplification of CaMV35S promoter

gene indicating that the plant has been transformed with T-DNA of pCAMBIA 1301

containing the gene of interest (HCC-scFv).

Page 196: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxcvi

Figure 5.10.1: Molecular analysis of the transformed plant (CaMV35S) by

polymerase chain reaction

PCR confirmation of CaMV35 gene in regenerated leaves

• Lane 1- marker (100bp)

• Lane 2-Negative control 1

• Lane 3-Negative control 2

• Lane 4&5-Positive control

• Lane 6-10- Samples-amplified CaMV 35S

• Lane 1-5- transformed callus regenerated-amplified CaMV 35S

• Lane 6,7-Positive control

• Lane 8,9- Negative control 3-untransformed callus regenerated

• Lane 10-marker(100bp)

1 2 3 4 5 6 7 8 9 10 1 2 3 4 5 6 7 8 9 10

195bp200 bp

100 bp

1000 bp

900 bp

800 bp

700 bp

600 bp

300 bp

400 bp500 bp

1000 bp

100 bp

A B

Page 197: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxcvii

.

CHAPTER – 6

SUMMARY

Page 198: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxcviii

CHAPTER 6

SUMMARY

The cloning of plasmid pCAMBIA 1301 and HCC-scFv (gene of interest) was

initially carried out using vector NTI software. The 5’ end and 3’ end of the

gene of interest (HCC-scFv) was modified by attaching with the gene sequence

of signal peptide (Rice α amylase) at 5’ end, KDEL retension signal peptide at

3’ end and the two adaptors NcoI (5’end) and pmlI (3’ end) were attached to the

gene to facilitate cloning. Cloning was initially carried out in vector NTI

software to confirm if the gene sequence could be inserted into the chosen T-

DNA plasmid – pCAMBIA 1301.

The gene of interest HCC-scFv from gene bank accession no: AY686498 had to

be modified to clone the gene into plant expression system based on the results

of the vector NTI software. The 5’ end and 3’ end were modified by adding

gene sequence of rice α amylase signal peptide at 5’end at 1-60 bp positions in

AY686498 and endoplasmic reticulum retention signal peptide KDEL at 3’end

to retain the protein molecule inside the endoplasmic reticulum and to avoid

protein degradation in the cytoplasm by proteolytic enzymes. To overcome the

Page 199: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cxcix

limitation of plant N-glycosylation, the proteins were retained in the

endoplasmic reticulum.

The ends were further modified by attaching the gene sequence of NcoI

adaptors and Pml adaptors to obtain restriction site present in the plasmid

pCAMBIA1301. In the modified AY686498 gene sequence the 24th

code c

(cytosine) i.e. cca coding for theronine was replaced by cga which also codes

for theronine. The reason for changing the coding sequence: ccatgg was

because that the gene sequence will be restricted by NcoI, when changed to

cgatgg the restriction site is altered but the amino acid sequence will remain the

same. This helped to clone the gene into pCAMBIA 1301 at NcoI and PmlI

restriction sites were the entire cDNA strand will remain unrestricted, when

restriced with NcoI and pmlI restriction enzymes.

The gene was cloned into pCAMBIA 1301 and transformed from E. coli DH5α

into Agrobacterium tumefaciens EHA 105 which was further co-cultivated with

the callus induced by 2, 4-D. The transformed regenerated plant was screened

for the presence of CaMV35S promoter. The amplification of CaMV35S from

the transformed plant indicates the presence of the HCC-scFv which was cloned

downstream of CaMV35S promoter.

Page 200: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cc

CHAPTER – 7

Page 201: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cci

CONCLUSION AND

SCOPE OF FUTURE

WORK

CHAPTER 7

CONCLUSION AND SCOPE OF FUTURE WORK

7.1. CONCLUSION

Page 202: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccii

The plasmid pCAMBIA 1301 and HCC-scfv (gene of interest) was successfully

cloned using vector NTI software after modifying the 5’ and 3’ ends of HCC-

scFv gene sequence.

The HCC-scFv gene sequence analysed for cloning, in vector NTI software was

successfully synthesized with the modified ends, to clone and express the gene

in the plant expression system.

The plasmid pCAMBIA 1301 (vector) and pMA-T (containing HCC-scFv)

obtained were successfully transformed into E. coli (DH5α) by calcium chloride

transformation. The transformed strains were confirmed by isolating the

plasmid and running them on the agarose gel.

The pCAMBIA 1301 (T-DNA vector) and pMA-T (containing the gene of

interest –HCC-scFv) were restricted with NcoI and PmlI restriction enzymes

and confirmed on 0.8% Agarose.

The band of particular size of the plasmid and the insert was successfully

eluted, ligated, transformed and confirmed on the agarose gel. From this

experiment it was confirmed that the gene of interest HCC-scFv was cloned

into the plasmid pCAMBIA 1301 and the cloned plasmid was denoted as

pCAMBIA 1301-HCC-scFv.

The recombinant Ti plasmid containing the gene of interest (pCAMBIA 1301-

HCC-scFv). was mobilized from E. coli DH5α into Agrobacterium tumefaciens

EHA 105

Page 203: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cciii

Callus has been successfully induced using 2, 4-D on MS medium containing

organic salts, inorganic salts, vitamins and carbon sources.

The Agrobacterium tumefaciens EHA 105 containing pCAMBIA1301-HCC-

scFv was successfully transformed into IR 64 callus and it was successfully

regenerated.

The DNA was isolated from the plant tissue by CTAB method and it was

further purified by using PCR clean up kit.

From the PCR technique it is clear that the T-DNA of pCAMBIA 1301

containing CaMV35S has been transformed into plant along with the gene of

interest (HCC-scFv). As HCC-scFv gene was cloned after CaMV35S promoter,

the amplification of CaMV35S indicates the presence of the HCC-scFv.

7.2. APPLICATION

The HCC-scFv transformed into IR64 can be used for the diagnosis and

immunotherapy of Heptocellular carcinoma (HCC).

The gene of HCC-scFv fragment cloned into IR64 can specifically bind to the

receptors of the Heptocellular carcinoma (HCC) cells.

The protein expressed from the cloned IR64 containing HCC-scFv gene

fragment can be isolated from the transformed plants which can be fused with

pro-drug, anti-cancer agents or radio nucleides. This fused HCC-scFv can

specifically bind to hepatocellular carcinoma receptors and release the

Page 204: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

cciv

particular drug specifically to the Heptocellular carcinoma (HCC) cells and kills

them.

The HCC-scFv fragment can be isolated from the transformed IR64 rice variety

which can also be used for the diagnosis of Heptocellular carcinoma (HCC) in

the earlier stages of cancer.

The method used in this research work can be used to produce various other

scFv fragments to target against other cancer receptors for diagnostic purposes

and immunotheraphy in IR64 rice variety.

7.3. SCOPE FOR FUTURE WORK

The HCC-scFv protein has to be extracted from the transformed IR64 rice

variety by immobilized metal affinity chromatoghraphy using IMAC

purification kit (5X his tag (fused with the gene) or 6X his tag (present on the

plasmid).

The presence of the protein has to be confirmed by western blotting.

The isolated protein (human monoclonal HCC-scFv) confirmed by western

blotting, can be fused with the drugs targeted specifically to HCC.

The drug fused protein has to be checked for its specificity on HepG2 cell line.

Toxicity assessment of the human monoclonal HCC-scFv fused with the drug

has to be carried out for its application.

Page 205: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccv

APPENDICES

Page 206: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccvi

APPENDICES

Appendix 1

Equation 4.5.2.1

Equation 4.7.1

Equation 4.8.1

Equation 4.10.1

Tm = 4 C (No. of G+C) + 2 C (No. of A+T)

ng of insert = ng of vector X Size of insert X insert ratio

--------------------------------

Size of vector X Vector ratio

Callus Induction frequency % = Total number of seeds that produced callus x 100

Total number of seeds plated

CRF (%) = Total number of callus that produced plants

---------------------------------------------------- X 100

Total number of callus plated

Page 207: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccvii

Appendix 2

Page 208: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccviii

Appendix 3

Page 209: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccix

Page 210: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccx

Page 211: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxi

Page 212: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxii

REFERENCES

Page 213: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxiii

REFERENCE

Adkins S and Burmeister M. (1996) “Visualization of DNA in agarose gels and

educational demonstrations”, Anal Biochem., Vol 240 No.10, pp 17-23.

Alamillo J.M, Monger W, Sola I, Garcia B, Perrin Y, Bestagno M, Burrone O.R,

Sabella P, Plana-Durán J, Enjuanes L, Lomonossoff G.P and García J.A. (2006) “

Use of virus vectors for the expression in plants of active full-length and single chain

anti-coronavirus antibodies”, Biotechnol J., Vol 1, pp 1103-11.

Arcalis E, Marcel S, Altmann F, Kolarich D, Drakakaki G, Fischer R, Christou P and

Stoger E. (2004) “Unexpected deposition patterns of recombinant proteins in post-

endoplasmic reticulum compartments of wheat endosperm” Plant Physiol., Vol 136,

pp 3457-3466.

Austin S, Bingham E.T, Koegel R.G, Mathews D.E, Shahan M.N, Straub R.J and

Burgess R.R. (1994) “An overview of feasibility study for the production of industrial

enzymes in transgenic alfalfa”, Ann NY Acad Sci., Vol 721, pp 234-244.

Bakker H, Bardor M, Molthoff J.W, Gomord V, Elbers I, Stevens L.H, Jordi W,

Lommen A, Faye L, Lerouge P, and Bosch D. (2001) “Galactose-extended glycans

of antibodies produced by transgenic plants”, Proc. Natl. Acad. Sci. USA., Vol 98, pp

2899-2904.

Bakker H, Rouwendal G.J, Karnoup A.S, Florack D.E, Stoopen G.M, Helsper J.P,van

Ree R, van Die I and Bosch D. (2006) “An antibody produced in tobacco expressing a

hybrid beta-1, 4-galactosyltransferase is essentially devoid of plant carbohydrate

epitopes”, Proc Natl Acad Sci USA., Vol 103, pp 7577-82.

Bardor M, Faveeuw C, Fitchette A.C, Gilbert D, Galas L, Trottein F, Faye L and

Lerouge P. (2003) “Immunoreactivity in mammals of two typical plant glyco-epitopes,

core alpha (1,3)-fucose and core xylose”, Glycobiology., Vol 13, pp 427-434.

Page 214: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxiv

Berger M, Shankar V and Vafai A. (2002) “Therapeutic applications of monoclonal

antibodies”, Am. J. Med. Sci., Vol 324, pp 14 -30.

Birnboim H.C and Doly J.A. (1979) “A rapid alkaline extraction procedure for

screening recombinant plasmid DNA”, Nucl. Acid. Res. Vol 7, pp 1513-23.

Borisjuk N.V, Borisjuk L.G, Logendra S, Petersen F, Gleba Y and Raskin I. (1999)

“Production of recombinant proteins in plant root exudates”, Nat Biotechnol., Vol 17,

pp 466-469.

Bosch F.X, Ribes J, Diaz M and Cleries R. (2004) “Primary liver cancer: worldwide

incidenceand trends”, Gastroenterology., Vol 127, pp 5-16.

Bouquin T, Thomsen M, Nielsen L.K, Green T.H, Mundy J and Hanefeld Dziegiel M.

(2002) “Human anti-rhesus D IgG1 antibody produced in transgenic plants”,

Transgenic Res. Vol 11, pp 115-122.

Bruyns A.M, De Jaeger G, De Neve M, De Wilde C, van Montagu M and Depicker A.

(1996) “Bacterial and plant-produced scFv proteins have similar antigen-binding

properties”, FEBS letters., Vol 386, pp 5-10.

Cabanes-Macheteau M, Fitchette-Laine A.C, Loutelier-Bourhis C, Lange C, Vine N.D,

Ma J.K, Lerouge P and Faye L.(1999) “N-Glycosylation of a mouse IgG expressed in

transgenic tobacco plants”, Glycobiology., Vol 9, pp 365-372.

Chadd H.E and Chamow S.M. (2001) “Therapeutic antibody expression technology.” ,

Curr. Opin. Biotechnol., Vol 12, pp 188-194.

Chari R.V. (1998) “Targeted delivery of chemotherapeutics: tumor-activated prodrug

therapy”, Adv.Drug Deliv.Rev., Vol 31, pp 89–104.

Conrad U and Fiedler U. (1998) “Compartment-specific accumulation of recombinant

immunoglobulins in plant cells, an essential tool for antibody production and

immunomodulation of physiological functions and pathogen activity”, Plant Mol Biol.,

Vol 38, pp 101–109.

Cox K.M, Sterling J.D, Regan J.T, Gasdaska J.R, Frantz K.K, Peele C.G, Black A,

Passmore D, Moldovan Loomis C, Srinivasan M, Cuison S, Cardarelli P.M and

Dickey L.F. (2006) “Glycan optimization of a human monoclonal antibody in the

aquatic plant Lemna minor”, Nat Biotechnol., Vol 24, pp 1591-1597.

Dagert M and Ehrlich S. (1979) “Prolonged incubation in calcium chloride improves

the competence of Escherichia coli cells”, Gene.,Vol 6, N0.1, pp 23–28.

Page 215: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxv

Daniell H and Dhingra A. (2002) “Multigene engineering: dawn of an exciting new era

in biotechnology”, Curr Opin Biotechnol., Vol 13, pp 136-141.

Daniell H, Streatfield S.J and Wycoff K. (2001) “Medical molecular farming:

production of antibodies, biopharmaceuticals and edible vaccines in plants”, Trends

Plant Sci., Vol 6, pp 219-226.

Dass S, Vital E.M and Emery P. (2006) “Rituximab: novel B-cell depletion therapy for

the treatment of rheumatoid arthritis”, Expert Opin. Pharmacother., Vol 7, No.18, pp

2559 -2570.

De Cosa B, Moar W, Lee S.B, Miller M and Daniell H. (2001) “Overexpression of the

Bt cry2Aa2 operon in chloroplasts leads to formation of insecticidal crystals”, Nat

Biotechnol., Vol 19, pp 71-74.

De Frammond, A. J., K. A. Barton, and M.-D. Chilton. (1983) “Mini-Ti-: a new vector

strategy for plant genetic engineering”, Bio/Technology, Vol 5, pp 262-269.

De Neve M, De Loose M, Jacobs A, Van Houdt H, Kaulza B, Weidle U, Van Montagu

M and Depicker A. (1993) “Assembly of an antibody and its derived antibody

fragment in Nicotiana and Arabidopsis”, Transgenic Res., Vol 2, pp 227-237.

De Wilde C, De Neve M, De Rycke R, Bruyns AM, De Jaeger G, van Montagu M,

Depicker A and Engler G. (1996) “Intact antigen-binding MAK33 antibody and Fab

fragment accumulate in intercellular spaces of Arabidopsis thaliana”, Plant science.,

Vol 114, pp 233-241.

Ditta, G, Schmidhauser T, Yakobson E., Lu P, Liang X.W, Finlay D.R, Guiney D and

Helinski, D.R. (1985) “Plasmids related to the broad range vector pRK 290 useful for

gene cloning and for monitoring gene expression”, Plasmid., Vol 13, pp 149-153.

Downing W.H, Galpin J.D, Clemens S, Lauzon S.M, Samuels A.L, Pidkowich M.S,

Clarke L.A and Kermode A.R. (2006) “Synthesis of enzymatically active human

alpha-L-iduronidase in Arabidopsis cgl (complex glycan-deficient) seeds” Plant

Biotechnol J., Vol 4, pp 169-182.

Drake P.M, Chargelegue D.M, Vine N.D, van Dolleweerd C.J, Obregon P and Ma J.K.

(2003) “Rhizosecretion of a monoclonal antibody protein complex from transgenic

tobacco roots”, Plant Mol Biol., Vol 52, pp 233-241.

Page 216: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxvi

During K, Hippe S, Kreutzaler F and Schell J. (1990) “Synthesis and self-assembly of

a functional monoclonal antibody in transgenic Nicotiana tobacum”, Plant Mol.

Biol.,Vol 15, pp 281-293.

Ehsani P, Meunier A, Nato F, Jafari A, Nato A and Lafaye P. (2003) “Expression of

anti human IL-4 and IL-6 scFvs in transgenic tobacco plants”, Plant Mol Biol., Vol 52,

pp 17-29.

Elbers I.J, Stoopen G.M, Bakker. H and Stevens LH. (2001) “Influence of growth

conditions and developmental stage on N-glycan heterogeneity of transgenic

immunoglobulin G and endogenous proteins in tobacco leaves”, Plant Physiol., Vol

126, No.3, pp 1314-22.

Farran I, Sanchez-Serrano J.J, Medina J.F, Prieto J and Mingo-Castel A.M. (2002)

“Targeted expression of human serum albumin to potato tubers”, Transgenic Res., Vol

11, pp 337-346.

Fattovich G, Stroffolini T, Zagni I. and Donato F. (2004) “Hepatocellular carcinoma in

cirrhosis: incidence and risk factors”, Gastroenterology., Vol 127, pp 35-50.

Faye L and Daniell H. (2006) “Novel pathway for glycoprotein import into

chloroplasts”, Plant Biotechnol J. Vol 4, pp 275-279.

Faye L, Boulaflous A, Benchabane M, Gomord V and Michaud D. (2005) “Protein

modifications in the plant secretory pathway: current status and practical implications

in molecular pharming ” , Vaccine., Vol 23, pp 1770-1778.

Fiedler U and Conrad U. (1995) “High-level production and long-term storage of

engineered antibodies in transgenic tobacco seeds”, Biotechnology., Vol 13, pp 1090-

3.

Fischer R and Emans N. (2000) “Molecular farming of pharmaceutical proteins”,

Transgenic Res., Vol 9, pp 279-299.

Fischer R, Schumann D, Zimmermann S, Drossard J, Sack M and Schillberg S. (1999)

“Expression and characterization of bispecific single-chain Fv fragments produced in

transgenic plants”, Eur J Biochem., Vol 262, pp 810-816.

Fischer R, Twyman R.M and Schillberg S. (2003) “Production of antibodies in plants

and their use for global health”, Vaccine., Vol 21, pp 820-825.

Page 217: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxvii

Frey A.D, Karg S.R and Kallio P.T. (2009) “Expression of rat beta(1, 4)-N-

acetylglucosaminyltransferase III in Nicotiana tabacum remodels the plant-specific N-

glycosylation”, Plant Biotechnol J., Vol 7, pp 33-48.

Fujiyama K, Misaki R, Sakai Y, Omasa T and Seki T. (2009) “Change in

glycosylation pattern with extension of endoplasmic reticulum retention signal

sequence of mouse antibody produced by suspension-cultured tobacco BY2 cells”, J

Biosci Bioeng., Vol 107, pp 165-72.

Fujiyama K, Palacpac N.Q, Sakai H, Kimura Y, Shinmyo A, Yoshida T and Seki T.

(2001) “In vivo conversion of a glycan to human compatible type by transformed

tobacco cells”, Biochem Biophys Res Commun., Vol 289, pp 553-557.

Gaume A, Komarnytsky S, Borisjuk N and Raskin I. (2003) “Rhizosecretion of

recombinant proteins from plant hairy roots”, Plant Cell Rep., Vol 21, pp 1188-1193.

Giddings G, Allison G, Brooks D and Carter A. (2000) “Transgenic plants as factories

for biopharmaceuticals”, Nat. Biotechnol., Vol 18, pp 1151-1155.

Goff S.A, Ricke D, Lan T.H, Presting G, Wang R, Dunn M, Glazebrook J, Sessions A,

Oeller P and Varma H. (2002) “A draft sequence of the rice genome (Oryza sativa L.

ssp.japonica)”, Science., Vol 296, pp 92–100.

Gomord V and Faye L. (2004) “Posttranslational modification of therapeutic proteins

in plants”, Curr Opin Plant Biol., Vol 7, pp 171-181.

Gomord V, Chamberlain P, Jefferis R and Faye L. (2005) “Biopharmaceutical

production in plants: problems, solutions and opportunities”, Trends Biotechnol., Vol

23, pp 559-565.

Gomord V, Denmat L.A, Fitchette-Laine A.C, Satiat-Jeunemaitre B, Hawes C and

Faye L. (1997) “The C-terminal HDEL sequence is sufficient for retention of secretory

proteins in the endoplasmic reticulum (ER) but promotes vacuolar targeting of proteins

that escape the ER”, Plant J., Vol 11, pp 313-25.

Gomord V, Sourrouille C, Fitchette A.C, Bardor M, Pagnt S, Lerouge P and Faye L.

(2004) “Production and glycosylation of plant-made pharmaceuticals: the antibodies as

a challenge”, Plant Biotechnol J., Vol 2, pp 83-100.

Gomord V, Wee E and Faye L. (1999) “Protein retention and localization in the

endoplasmic reticulum and the golgi apparatus”, Biochimie., Vol 81, pp 607-618.

Page 218: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxviii

Hamilton C. M., Lee H., LiP.-L., Cook D. M., Piper K. R., von Bodman S. B., Lanka

E., Ream W. and Farrand S. K. (2000) “TraG from RP4 and TraG and VirD4 from Ti

Plasmids Confer Relaxosome Specificity to the Conjugal Transfer System of pTiC58.

J”, Bacteriol, Vol 182, pp 1541-1548

Haq T.A, Mason H.S, Clements J.D and Arntzen C.J. (1995) “Oral immunization with

a recombinant bacterial antigen produced in transgenic plants”, Science., Vol 268, pp

714-6.

Hein M.B, Tang Y, McLeod D.A, Janda K.D and Hiatt A. (1991) “Evaluation of

immunoglobulins from plant cells”, Biotechnol. Prog., Vol 7, pp 455-461.

Hellens R.P, Edwards E.A, Leyland N.R, Bean S and Mullineaux P.M. (2000)

“pGreen: a versatile and flexible binary Ti vector for Agrobacterium-mediated plant

transformation”, Plant Mol Biol., Vol 42, pp 819-32.

Hellwig S, Drossard J, Twyman R.M and Fischer R. (2004) “Plant cell cultures for the

production of recombinant proteins”, Nat Biotechnol., Vol 22, pp 1415-1422.

Hiatt A, Cafferkey R and Bowdish K. (1989) “Production of antibodies in transgenic

plants”, Nature., Vol 342, pp 76-78.

Hiatt A. (1990) “Antibodies produced in plants”, Nature., Vol 344, pp 469-470.

Hiei Y, Ohta S, Komari T and Kumashiro T. (1994) “Efficient transformation of rice

(Oryza sativa L.,) mediated by Agrobacterium and sequence analysis of the boundaries

of the T-DNA”, Plant J., Vol 6, pp 271-282.

Hiroaki Inouea, Hiroshi Nojimab and Hiroto Okayama. (1990) “High efficiency

transformation of Escherichia coli with plasmids”, Gene Volume 96, No1, pp 23-28.

Hoekema, A., P. R. Hirsh, P. J. J. Hooykaas, and R. A. Schilperoort. (1983) “A binary

plant vector strategy based on separation of vir-and T-region of the Agrobacterium

tumefaciens Ti-plasmid”, Nature, Vol 303, pp 179-180.

Hood E.E, Love R, Lane J, Bray J, Clough R, Pappu K, Drees C, Hood K.R, Yoon S,

Ahmad A and Howard J.A. (2007) “Subcellular targeting is a key condition for high-

level accumulation of cellulase protein in transgenic maize seed”, Plant Biotechnol J.,

Vol 5, No. 6, pp 709-719.

Hood, E. E., G. L. Helmer, R. T. Fraley, and M.-D. Chilton. (1986) “The

hypervirulence of Agrobacterium tumefaciens A281 is encoded in a region of

pTiBo542 outside of T-DNA. J”, Bacteriol., Vol 168, pp 1291-1301

Page 219: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxix

Hood, E. E., S. B. Gelvin, L. S. Melchers, and A. Hoekema. (1993) “New

Agrobacterium helper plasmids for gene transfer to plants”, Transgenic Res., Vol 2, pp

208-218.

Humphrey B.D, Huang N and Klasing K.C. (2002) “Rice expressing lactoferrin and

lysozyme has antibiotic-like properties when fed to chicks”, J Nutr., Vol 132, pp 1214-

1218.

Ish-Horowicz D and Burke J.F. (1981) “Rapid and efficient cosmid cloning”, Nucleic

Acids Research, Vol 9 No 13, pp 2898-2989.

Jarvis D.L. (2003) “Developing baculovirus-insect cell expression systems for

humanized recombinant glycoprotein production”, Virology., Vol 310, pp 1-7.

Jin C, Bencurova M, Borth N, Ferko B, Jensen-Jarolim E, Altmann F and Hantusch

B. (2006) “Immunoglobulin G specifically binding plant N-glycans with high affinity

could be generated in rabbits but not in mice”, Glycobiology., Vol 16, pp 349-357.

Kapusta J, Modelska A, Figlerowicz M, Pniewski T, Letellier M, Lisowa O, Yusibov

V, Koprowski H, Plucienniczak A and Legocki A.B. (1999) “A plant-derived edible

vaccine against hepatitis B virus”, FASEB J., Vol 13, pp 1796-1799.

Karabi Datta and Swapan Datta. (2006) “Methods in Molecular Biology, vol. 343

Agrobacterium Protocols, 2/e, volume1, chapter 16. Indica rice (Oryza sativa, BR29

and IR64)”, Edited by Kan Wang Human press Inc., Totowa, NJ.

kathuria S, Sriraman R, Nath R, Sack M, Pal R, Artsaenko O, Talwar G.P, Fischer R

and Finnern R. (2002) “Efficacy of plant-produced recombinant antibodies against

HCG”, Hum reprod., Vol 17, pp 2054-2061.

Kathuria S.R, Nath R, Pal R, Sigh O, Fischer R, Lohiya N.K and Talwar G.P. (2002)

“Functional recombinant antibodies against human chorionic gonadotropin expressed

in plants”, Curr.sci., Vol 82, pp 1452-1457.

Kelm S and Schauer R. (1997) “Sialic acids in molecular and cellular interactions”, Int

Rev Cytol., Vol 175, pp 137-240.

Khoudi H, Laberge S, Ferullo J.M, Bazin R, Darveau A, Castonguay Y, Allard G,

Lemieux R and Vezina L.P. (1999) “Production of a diagnostic monoclonal antibody

in perennial alfalfa plants”, Biotechnol Bioeng., Vol 64, pp 135-143.

Page 220: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxx

Kikkert J.R, Vidal J.R and Reisch B.I. (2005) “Stable transformation of plant cells by

particle bombardment/biolistics”, Methods Mol Biol., Vol 286, pp 61-78.

Kilpatrick J, Cockburn B and Whitelam G. (1995) “The expression of recombinant

proteins in crop plants”, Outlook Agri., Vol 24, pp 207-211.

Kim S.J, Park Y and Hong H.J. (2005) “Antibody engineering for the development of

therapeutic antibodies”, Mol. Cells., Vol 20, No.1, pp 17 -29.

Ko K, Tekoah Y, Rudd P.M, Harvey D.J, Dwek R.A, Spitsin S, Halon C.A, Rupprecht

C,Dietzschold B, Golovkin M and Koprowski H. (2003) “Function and glycosylation

of plant-derived antiviral monoclonal antibody”, Proc Natl Acad Sci USA., Vol 100,

pp 8013-8018.

Komari T, Hiei Y, Saito Y, Murai N and Kumashiro T. (1996) “Vectors carrying two

separate T-DNAs for co-transformation of higher plants mediated by Agrobacterium

tumefaciens and segregation of transformants free from selection markers”, Plant J.,

Vol 10, pp 165-174.

Komarnytsky S, Borisjuk N, Yakoby N, Garvey A and Raskin I. (2006) “Cosecretion

of protease inhibitor stabilizes antibodies produced by plant roots”, Plant Physiol., Vol

141, pp 1185-1193.

Koncz, C. and J. Schell. (1986) “The promoter of TL-DNA gene 5 controls the tissue-

specific expression of chimaeric genes carried by a novel type of Agrobacterium

binary vector. Mol. Gen”, Genet, Vol 204, pp 383-396.

Koprivova A, Stemmer C, Altmann F, Hoffmann A, Kopriva S, Gorr G, Reski R and

Decker E.L. (2004) “Targeted knockouts of Physcomitrella lacking plant-specific

immunogenic N-glycans”, Plant Biotechnol. J., Vol 2, pp 517-523.

Krauss J. (2003) “Recombinant antibodies for the diagnosis and treatment of cancer”,

Mol. Biotechnol., Vol 25, pp 1-17.

Larrick J.W, Yu L, Naftzger C, Jaiswal S and Wycoff K. (2001) “Production of

secretory IgA antibodies in plants”, Biomol Eng., Vol 18, pp 87-94.

Lazo, G. R., P. A. Stein, and R. A. Ludwig. (1991) “A DNA transformation-competent

Arabidopsis genomic library in Agrobacterium”, Bio/Technology Vol 9, pp 963-967.

Lehnman I.R. (1974) “DNA Ligase: Structure, Mechanism, and Function”, Science.,

Vol 186, No.4166, pp 790–7.

Page 221: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxi

Lerouge P, Cabanes-Macheteau M, Rayon C, Fischette-Laine A.C, Gomord V and

Faye L. (1998) “N-glycoprotein biosynthesis in plants: recent developments and future

trends”, Plant Mol Biol., Vol 38, pp 31-48.

Li B, Huang W and Bass T. (2003) “Shoot production per responsive leaf explant

increases exponentially with explant organogenic potential in Nicotiana species”, Plant

Cell Rep., Vol 22, pp 231-238.

Liu S.Y, Eary J.F, Petersdorf S.H, Martin P.J, Maloney D.G, Appelbaum F.R,

Matthews D.C, Bush S.A, Durack L.D, Fisher D.R, Gooley T.A, Bernstein I.D and

Press O.W. (1998) “Follow-up of relapsed B-cell lymphoma patients treated with

iodine-131-labeled anti-CD20 antibody and autologous stem-cell rescue”,

J.Clin.Oncol., Vol 16, pp 3270-3278.

Ma J.K, Hiatt A, Hein M, Vine N.D, Wang F, Stabila P, van Dolleweerd C, Mostov

K and Lehner T. (1995) “Generation and assembly of secretory antibodies in plants”,

Science., Vol 268, pp 716-9.

Ma J.K, Drake P.M and Christou P. (2003) “The production of recombinant

pharmaceutical proteins in plants”, Nat Rev Genet., Vol 4, pp 794-805.

Ma J.K.C and Hein M.B. (1996) “Antibody production and engineering in plants. In:

Collins GB, Shepherd RJ (eds). Engineering plants for commercial products and

applications”, New York Academy of Sciences, New York., Vol 792, pp 72-81.

Ma J.K.C, Lehner T, Stabila P, Fux CI and Hiatt A. (1994) “Assembly of monoclonal

antibodies with IgG1 and IgA heavy chain domains in transgenic tobacco plants”, Eur.

J.Immunol., Vol 24, pp 131-138.

Ma K.C.J and Hein BM. (1995) “Plant antibodies for Immunotherapy”, Plant physiol.,

Vol 109, pp 341-346.

Ma K.C.J, Hikmat Y.B, Wycoff K, Vine D.N, Chargelegue D,Yu L, Hein B.M and

Lehner T. (1998) “Characterization of a recombinant plant monoclonal secretory

antibody and preventive Immunotherapy in humans”, Nature Medicine., Vol 4, pp

601-606.

Mayfield S.P, Franklin S.E and Lerner R.A. (2003) “Expression and assembly of a

fully active antibody in algae”, Proc Natl Acad Sci USA., Vol 100, pp 438-442.

Page 222: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxii

McCormick A, Kumagal M.H, Hanley K, Turpen T.H, Hakim I, Grill L.K, Tuse D,

Levy S and Levy R. (1999) “Rapid production of specific vaccines for lymphoma by

expression of the tumor-derived single-chain Fv epitopes in tobacco plants”, Proc,

Natl.Acad, Sci, U.S.A., Vol 96, pp 703-708.

Mendez M.J, Green L.L and Corvalan J.R. (1997) “Functional transplant of megabase

human immunoglobulin loci recapitulates human antibody response in mice”, Nat.

Genet., Vol 15 No.2, pp 146 -156.

Misaki R, Fujiyama K and Seki T. (2006) “Expression of human CMP-N-

acetylneuraminic acid synthetase and CMP-sialic acid transporter in tobacco

suspension-cultured cell”, Biochem Biophys Res Commun., Vol 339, pp 1184-9.

Misaki R, Kimura Y, Palacpac N.Q, Yoshida S, Fujiyama K and Seki T. (2003) “Plant

cultured cells expressing human beta1,4-galactosyltransferase secrete glycoproteins

with gala-ctose-extended N-linked glycans”, Glycobiology., Vol 13, pp 199-205.

Morrow K.J. (2001) “Biopharm manufacturing capacities”, Genet Eng News., Vol 21,

pp 1-85.

Mullis K.B. and Faloona F.A. (1987) “Specific synthesis of DNA in vitro via a

polymerase-catalyzed chain reaction”, Methods Enzymol , Vol 155, pp 335-50.

Murray M.G. and Thompson W.F. (1980) “Rapid isolation of high molecular weight

plant DNA”, Nucleic Acid Res., Vol 8, pp 19-20.

Negrouk V, Eisner G, Lee H.I, Han K, Taylor D and Wong H.C. (2005) “Highly

efficient transient expression of functional recombinant antibodies in lettuce”, Plant

Science., Vol 169, pp 433-438.

Ooms, G., P. J. J. Hooykaas, G. Moolenaar, and R. A. Schilperoort. (1981) “Crown

gall plant tumors of abnormal morphology, induced by Agrobacterium tumefaciens

carrying mutated octopine Ti plasmids; analysis of T-DNA functions”, Gene Vol 14,

pp 33-50.

Osbourn J, Jermutus L and Duncan A. (2003) “Current methods for the generation of

human antibodies for the treatment of autoimmune disease”, Drug Discov. Today., Vol

8, pp 845-851.

Paccalet T, Bardor M, Rihouey C, Delmas F, Chevalier C, D'Aoust M.A, Faye L,

Vezina L, Gomord V and Lerouge P.et al. (2007) “Engineering of a sialic acid

Page 223: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxiii

synthesis pathway in transgenic plants by expression of bacterial Neu5Ac-synthesizing

enzymes”, Plant Biotechnol J., Vol 5, pp 16-25.

Pagny S, Denmat-Ouisse L.A, Gomord V and Faye L. (2003) “Fusion with HDEL

protects cell wall invertase from early degradation when N-glycosylation is inhibited”,

Plant Cell Physiol., Vol 44, pp 173-182.

Pai L.H and Pastan I. (1998) “Clinical trials with Pseudomonas exotoxin

immunotoxins”, Curr.Top.Microbiol.Immunol., Vol 234, pp 83–96.

Palacpac N.Q, Yoshida S, Sakai H, Kimura Y, Fujiyama K, Yoshida T and Seki T.

(1999) “Stable expression of human beta1, 4-galactosyltransferase in plant cells

modifies N-linked glycosylation patterns”, Proc Natl Acad Sci USA., Vol 96, pp 4692-

7.

Palanichelvam, K., P. Oger, S. J. Clough, C. Cha, A. F. Bent, and S. K. Farrand.

(2000) “A second T-region of the soybean-supervirulent chrysopine-type Ti plasmid

pTiChry5, and construction of a fully disarmed vir helper plasmid. ” , Mol. Plant-

Microbe Interact,Vol 13, pp1081-1091.

Park M, Kim S.J, Vitale A and Hwang I. (2004) “Identification of the protein storage

vacuole and protein targeting to the vacuole in leaf cells of three plant species”, Plant

Physiol., Vol 134, pp 625-639.

Park M, Lee D, Lee G.J and Hwang I. (2005) “AtRMR1 functions as a cargo receptor

for protein trafficking to the protein storage vacuole”, J Cell Biol., Vol 170, pp 757-

767.

Parkin D.M, Bray F, Ferlay J and Pisani P. (2001) “Estimating the world cancer

burden”, Globocan 2000. Int J Cancer., Vol 94 No.2, pp 153-6.

Peeters K, De Wilde C, De Jaeger G, Angenon G and Depicker A. (2001) “Production

of antibodies and antibody fragments in plants”, Vaccine., Vol 19, pp 2756-2761.

Petruccelli S, Otegui M.S, Lareu F, Tran Dinh O, Fitchette A-C, Circosta A, Rumbo

M, Bardor M, Carcamo R, Gomord V and 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

Biotechnol J., Vol 4, pp 511-527.

Pisani P, Bray F and Parkin D.M. (2002) “Estimates of the world-wide prevalence of

cancer for 25 sites in the adult population”, Int J Cancer., Vol 97, No.1, pp 72-81.

Page 224: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxiv

Porta C and Lomonossoff G.P. (1996) “Use of viral replicons for the expression of

genes in plants”, Mol Biotechnol., Vol 5, pp 209-21.

Pueyo J.J, Chrispeels M.J and Herman E.M. (1995) “Degradation of transport-

competent destabilized phaseolin with a signal for retention in the endoplasmic

reticulum occurs in the vacuole”, Planta., Vol 196, pp 586-596.

Pujol M, Ramırez N.I, Ayala M, Gavilondo J.V, Valdes R, Rodriguez M, Brito J,

Padilla S, Gomez L, Reyes B, Peral R, Perez M, Marcelo J.L, Mila L, Sanchez R.F,

Rolando P, Cremata J.A, Enriquez G, Mendoza O, Ortega M and Borroto C. (2005)

“An integral approach towards a practical application for a plant-made monoclonal

antibody in vaccine purification”, Vaccine., Vol 23, pp 1833-1837.

Ramirez N, Ayala M, Lorenzo D, Palenzuela D, Herrera L, Doreste V, Perez M,

Gavilond J.V and Oramas P. (2002) “Expression of a single-chain Fv antibody

fragment specific for the hepatitis B surface antigen in transgenic tobacco plants”,

Transgenic Res., Vol 11, pp 61-64.

Roger Hellens, Philip Mullineaux and Harry Klee. (2000) “A guide to Agrobacterium

binary Ti vectors”, Trends in Plant Science, Vol 5 No.10, pp 446-451.

Rogers, S.O and A.J.Bendich. (1994), “Extraction of total cellular DNA from plants,

algae and fungi.” In S.B. Gelvin and R.A.Schilperoort Eds. Plant Molecular Biology

Manual. Kluwer Academic Publishers, London.

Saint-Jore-Dupas C, Faye L and Gomord V. (2007) “From planta to pharma with

glycosylation in the toolbox”, Trends Biotechnol., Vol 25, pp 317-23.

Schneebaum S, Troitsa A, Avital S, Haddad R, Kashtan H, Gitstein G, Baratz M,

Brazovsky E, Papo J and Skornick Y. (2000) “Identification of lymph node

metastases in recurrent colorectal cancer”, Recent Results Cancer Res., Vol 157, pp

281-292.

Semenyuk E.G, Stremovskii O.A, Orlova I.V, Balandin T.G, Nosov A.M, Buryanov

Ya I, Deyev S.M and Petrov R.V. (2002) “Biosynthesis of the scFv antibody to human

Ferritin in plant and bacterial Producers”, Molecular biology., Vol 37, pp 780-786.

Seon J.H, Szarka S and Molone M.M. (2002) “A unique strategy for recovering

recombinant proteins from molecular farming: affinity capture on engineered

oilbodies”, J Plant Biotechnol., Vol 4, pp 95-101.

Page 225: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxv

Seveno M, Bardor M, Paccalet T, Gomord V, Lerouge P and Faye L. “Glycoprotein

sialylation in plants?”, Nat Biotechnol., Vol 22, pp 1351-3.

Shah M.M, Fujiyama K, Flynn C.R and Joshi L. (2003) “Sialylated endogenous

glycoconjugates in plant cells”, Nat Biotechnol., Vol 21, pp 1470-1.

Smith M.D and Glick BR. (2000) “The production of antibodies in plants: An idea

whose time has come”, Biotechnol. Adv., Vol 18, pp 85-89.

Sojikul P, Buehner N and Mason H.S. (2003) “A plant signal peptide-hepatitis B

surface antigen fusion protein with enhanced stability and immunogenicity expressed

in plant cells”, Proc Natl Acad Sci USA., Vol 100, pp 2209-2214.

Sparrow P.A.C, Irwin J.A, Dale P.J, Twyman R.M and Ma J.K.C. (2007) “Pharma-

Planta: road testing the developing regulatory guidelines for plant-made

pharmaceuticals”, Transgenic Res., Vol 16, pp 147–161.

Sriraman R, Bardor M, Sack M, Vaquero C, Faye L, Fischer R, Finnern R and

Lerouge P. (2004) “Recombinant anti-hCG antibodies retained in the endoplasmic

reticulum of transformed plants lack core-xylose and core-alpha(1,3)-fucose residues”,

Plant Biotechnol J., Vol 2 No.4, pp 279-287.

Staub J.M, Garcia B, Graves J, Hajdukiewicz P.T, Hunter P, Nehra N, Paradkar V,

Schlittler M, Carroll J.A, Spatola L, Ward D, Ye G and Russell D.A. (2000) “High-

yield production of a human therapeutic protein in tobacco chloroplasts”, Nat

Biotechnol., Vol 18, pp 333-338.

Stein K.E and Webber K.O. (2001) “The regulation of biologic products derived from

bioengineered plants”, Curr Opin Biotechnol., Vol 12, pp 308-311.

Stoger E, Sack M, Fischer R and Christou P. (2002) “Plantibodies: applications,

advantages and bottlenecks”, Curr Opin Biotechnol., Vol 13, pp 161-166.

Stoger E, Sack M, Perrin Y, Vaquero C, Torres E, Twyman R.M, Christou P and

Fischer R. (2002) “Practical considerations for pharmaceutical antibody production in

different crop systems”, Mol Breed., Vol 9, pp 149–158.

Stoger E, Vaquero C, Torres E, Sack M, Nicholson L, Drossard J, Williams S, Keen

D, Perrin Y, Christou P and Fischer R. (2000) “Cereal crops as viable production and

storage systems for pharmaceutical scFv antibodies”, Plant Mol Biol., Vol 42, pp

583–590.

Page 226: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxvi

Strasser R, Altmann F, Mach L, Glossl J and Steinkellner H. (2004) “Generation of

Arabidopsis thaliana plants with complex N-glycans lacking beta 1, 2-linked xylose

and core alpha 1, 3-linked fucose”, FEBS Lett., Vol 561, pp 132-6.

Strasser R, Stadlmann J, Schahs M, Stiegler G, Quendler H, Mach L,Glossl J,

Weterings K, Pabst M and Steinkellner H. (2008) “Generation of glyco-engineered

Nicotiana benthamiana for the production of monoclonal antibodies with a

homogeneous human-like N-glycan structure”, Plant Biotechnol J., Vol 6, pp 392-402.

Strasser R, Stadlmann J, Svoboda B, Altmann F, Glossl J and Mach L. “Molecular

basis of N-acetylglucosaminyltransferase I deficiency in Arabidopsis thaliana plants

lacking complex N-glycans”, Biochem J., Vol 387, pp 385-391.

Streatfield S.J. (2006) “Approaches to achieve high-level heterologous protein

production in plants”, Plant Biotechnol J., Vol 5, pp 2-15.

Tabe L.M, Wardley-Richardson T, Ceriotti A, Aryan A, McNabb W, Moore A and

Higgins T.J. (1995) “A biotechnological approach to improving the nutritive value of

alfalfa”, J Anim Sci., Vol 73, pp 2752-2759.

Takaiwa F, Takagi H, Hirose S and Wakasa Y. (2007) “Endosperm tissue is good

production platform for artificial recombinant proteins in transgenic rice”, Plant

Biotechnol J., Vol 5, pp 84–92.

Thor Straten P, Guldberg P, Schrama D andersen M.H, Moerch U, Seremet T, Siedel

C, Reisfeld R.A and Becker J.C. (2001) “In situ cytokine therapy: redistribution of

clonally expanded T cells”, Eur.J.Immunol., Vol 31, pp 250–258.

Torres E, Vaquero C, Nicholson L, Sack M, Stoger E, Drossard J, Christou P, Fischer

R and Perrin Y. (1999) “Rice cell culture as an alternative production system for the

functional diagnostic and therapeutical antibodies”, Transgenic Res., Vol 8, pp 441-

449.

Triguero A, Cabrera G, Cremata J.A, Yuen C.T, Wheeler J and Ramirez N.I. (2005)

“Plant-derived mouse IgG monoclonal antibody fused to KDEL endoplasmic

reticulum-retention signal is N-glycosylated homogeneously throughout the plant with

mostly high mannose-type N-glycans”, Plant Biotechnol J., Vol 3, pp 449-457.

Twyman R.M, Stoger E, Schillberg S, Christou P and Fischer R. (2003) “Molecular

farming in plants: host systems and expression technology”, Trends Biotechnol., Vol

21, pp 570-578.

Page 227: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxvii

Usha R, Rohll J, Spall V, Shanks M, Maule A, Johnson J and Lomonossoff G. (1993)

“Expression of an animal virus antigenic site on the surface of a plant virus particle”,

Virology., Vol 197, pp 366-374.

Valdes R, Gomez L, Padilla S, Brito J, Reyes B, Alvarez T, Mendoza O, Herrera O,

Ferro W, Pujol M, Leal V, Linares M, Hevia Y, Garcia C, Mila L, Garcia O, Sanchez

R, Acosta A, Geada D, Paez R,Vega J.L and Borroto C. (2003) “Large-scale

purification of an antibody directed against hepatitis B surface antigen from transgenic

tobacco plants”, Biochemical and Biophysical Research communications., Vol 308, pp

94-100.

Van Droogenbroeck B, De Wilde K and Depicker A. (2008) “Production of antibody

fragments in Arabidopsis seeds”, Methods Mol Biol., Vol 483, pp 89-101.

Van Engelen F.A, Schouten A, Molthoff J.W, Roosien J, Salinas J, Dirkse W.G,

Schots A, Bakker J, Gommers F.J and Jongsma M.A. (1994) “Coordinate expression

of antibody subunit genes yields high levels of functional antibodies in roots of

transgenic tobacco”, Plant Mol Biol., Vol 26, pp 1701-1710.

Van Rooijen G.J and Moloney M.M. (1995) “Structural requirements of oleosin

domains for subcellular targeting to the oil body”, Plant Physiol., Vol 109, pp 1353-

1361.

Vaquero C, Sack M, Schuster F, Finnem R, Drossard I, Schumann D, Reimann A and

Fischer R. (2002) “A carcinoembryonic antigen-specific diabody produced in

tobacco”, FASEB J., Vol 16, pp 408-410.

Verch T, Yusibov V and Koprowski H. (1998) “Expression and assembly of a full-

length monoclonal antibody in plants using a plant virus vector”, J Immunol Methods.,

Vol 220, pp 69-75.

Vitale A and Pedrazzini E. (2005) “Recombinant pharmaceuticals from plants: the

plant endo membrane system as bioreactor”, Mol Interv., Vol 5, pp 216-225.

Von Schaewen A, Sturm A, O'Neill J and Chrispeels M.J. (1993) “Isolation of a

mutant Arabidopsis plant that lacks N-acetyl glucosaminyl transferase I and is unable

to synthesize Golgi-modified complex N-linked glycans”, Plant Physiol., Vol 102, pp

1109-18.

Voss A, Nierbach M, Hain R, Hirsch H.J, Liao Y, Kreuzaler F and Fischer R. (1995)

“Reduced virus infectivity in N. tabacum secreting a TMV-specific full size antibody”,

Mol. Breed., Vol 1, pp 15-26.

Page 228: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxviii

Walsh G and Jefferis R. (2006) “Post-translational modifications in the context of

therapeutic proteins”, Nat Biotechnol., Vol 24, pp 1241-1252.

Wandelt C.I, Khan M.R, Craig S, Schroeder H.E, Spencer D and Higgins T.J. (1992)

“Vicilin with carboxy-terminal KDEL is retained in the endoplasmic reticulum and

accumulates to high levels in the leaves of transgenic plants”, Plant J., Vol 2, pp 181-

92.

Waters V.L. (1999) “conjugative transfer in the dissemination of beta-lactam and

aminoglycoside resistance”, Front.Biosci., Vol 4, pp 433-456.

Wee E.G, Sherrier D.J, Prime T.A and Dupree P. (1998) “Targeting of active

sialyltransferase to the plant Golgi apparatus”, Plant Cell., Vol 10, pp 1759-68.

Weiss B and Richardson C.C, (1967). "Enzymatic Breakage and Joining of

Deoxyribonucleic Acid, I. Repair of Single-Strand Breaks in DNA by an Enzyme

System from Escherichia coli Infected with T4 Bacteriophage", Proceedings of the

National Academy of Sciences, Vol 57 No.4, pp 1021–8.

White P.R. (1939a) “Potentially unlimited growth of excised plant callusin an

artificial nutrient”, Am. J. Bot., Vol 26, pp 59–64.

White P.R. (1939b) “Controlled differentiation in a plant tissue culture.Bull”, Torrey

Bot. Club, Vol 66,pp 507–513.

Wright A and Morrison S.L. (1998) “Effect of C2-associated carbohydrate structure on

Ig effector function: studies with chimeric mouse-human IgG1 antibodies in

glycosylation mutants of Chinese hamster ovary cells”, J Immunol., Vol 160, pp 3393-

402.

Yamagata H and Tanaka K. (1986) “The site of synthesis and accumulation of rice

storage proteins”, Plant Cell Physiol., Vol 27, pp 135–145.

Yang D, Guo F, Liu B, Huang N and Watkins S.C. (2003) “Expression and

localization of human lysozyme in the endosperm of transgenic rice”, Planta., Vol 216

No.4, pp 597-603.

Yang L, Tada Y, Yamamoto M.P, Zhao H, Yoshikawa M and Takaiwa F. (2006) “A

transgenic rice seed accumulating an anti-hypertensive peptide reduces the blood

pressure of spontaneously hypertensive rats”, FEBS Lett., Vol 580, pp 3315-3320.

Page 229: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxix

Yoder J.I and Goldsbrough A.P. (1994) “Transformation systems for generating

marker-free transgenic plants”, Bio/Technology., Vol 12, pp 263-267.

Yuan Q, Hu W, Pestka J.J, He S.Y and Hart L.P. (2000) “Expression of a functional

antizearalenone single-chain Fv antibody in transgenic Arabidopsis plants”, Appl

Environ Microbiol., Vol 66 No.8, pp 3499-505.

Zeitlin L, Olmsted S.S, Moench T.R, Co M.S, Martinell B.J, Paradkar V.M, Russell

D.R, Queen C, Cone R.A and Whaley K.J. (1998) “A humanized monoclonal

antibody produced in transgenic plants for immunoprotection of the vagina against

genital herpes.” Nat Biotechnol., Vol 16, pp 1361-4.

Zeleny R, Kolarich D, Strasser R and Altmann F. (2006) “Sialic acid concentrations in

plants are in the range of inadvertent contamination”, Planta., Vol 224, pp 222-7.

Page 230: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxx

LIST OF

PUBLICATIONS

LIST OF PUBLICATIONS

Page 231: CLONING, TRANSFORMATION AND EXPRESSION OF cDNA

ccxxxi

(Peer-Reviewed Publications- International journals)

Aadarsh Prasanna, Deepa.V, Balakrishna Murthy.P, M.Deecaraman, Sridhar.R

and Dhandapani P. Insoluble phosphate solubilization by Bacterial strains

isolated from Rice rhizosphere soils from Southern India. International journal of

soil science., 2011, 6(2), 134-141. (Impact factor 0.4)

Aadarsh Prasanna, P. Balakrishna Murthy, M. Deecaraman and R. Sridhar.

Optimization of Callus Induction and Regeneration in Swarna masoori rice

cultivar. Research Journal of Agriculture and Biological Sciences, 6(6): 917-922,

2010. (Impact factor 0.3)

Deepa.V, Aadarsh Prasanna, Balakrishna Murthy.P, M.Deecaraman, Sridhar.R

Efficient Phosphate solubilization by fungal strains isolated from Rice-

rhizosphere soils for the phosphorus release. Research journal of Agriculture

and Biological Science, 6(4): 487-492, 2010. (Impact factor 0.3)

T.N. Sathya, Aadarsh Prasanna, Deepa.V, Balakrishna Murthy.P. Moringa

oleifera lam. Leaves prevent Cyclophosphamide-induced micronucleus and DNA

damage in mice. International Journal of phytomedicine, 2 (2010) 147-154. (

Impact factor 0.1)