72
This thesis comprises 30 ECTS credits and is a compulsory part in the Master of Science with a Major in Resource Recovery - Industrial Biotechnology, 120 ECTS credits No. 2/2012 Construction of hpRNA expression vector for silencing a gene in Rhizopus oryzae Kiran Kumar Penmatsa Bharat Balu

Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

Embed Size (px)

Citation preview

Page 1: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

This thesis comprises 30 ECTS credits and is a compulsory part in the Master of Science

with a Major in Resource Recovery - Industrial Biotechnology, 120 ECTS credits

No. 2/2012

Construction of

hpRNA expression vector

for silencing a gene in

Rhizopus oryzae

Kiran Kumar Penmatsa

Bharat Balu

Page 2: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

ii

Construction of hpRNA expression vector for silencing LDHA gene in Rhizopus oryzae

Kiran Kumar Penmatsa, [email protected]

Bharat Balu, [email protected]

Master thesis

Subject Category: Technology

University of Borås

School of Engineering

SE-501 90 BORÅS

Telephone +46 033 435 4640

Examiner: Elisabeth Feuk Lagerstedt

Supervisor,name: Elisabeth Feuk Lagerstedt

Supervisor,address: School of Engineering, University of Borås

SE-50190, Borås

Date: 2012-08-28

Keywords: RNA interference, hpRNA, Silencing vector construction, Rhizopus

oryzae, Ethanol, Lactic acid

Page 3: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

iii

Acknowledgement

It’s a great privilege to express our deep gratitude to our supervisor Dr. Elisabeth Feuk-

Lagerstedt for her continuous guidance, enthusiastic encouragement and patience during the

project.

This project would not be completed on time without the support of the PhD students and

personal in the Department of Biotechnology, University of Borås. We would like to thank

Dr. Patrik Lennartsson for his valuable suggestions which have helped us in deciding the

alternative methods for solving technical problems encountered. We also want to thank Päivi

Ylitervo for her support in handling equipment in the laboratory and Johan Westman for his

supportive ideas. Kristina Laurila had been a great support for us by assisting in the lab during

the project, a hearty thanks to her.

We would also like to thank Dr. Peter Therning for all his support during thesis work.

Page 4: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

iv

Page 5: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

v

Page 6: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

vi

Abstract

Depending on the previous research on LDHA gene silencing in Rhizopus oryzae CCUG

28959 through introduction of siRNA, a integral vector was constructed by inserting two

copies of LDHA gene (by PCR cloning) in a fashion that it can express hpRNA in the

transformed fungi, which will trigger the post transcriptional degradation of targeted mRNA

through RNA degradation pathway which is known to be quelling in fungi.

The vector was successfully designed with the LDHA gene, transformed in to the host

organism, and also transferred to its progeny. This helps in maintaining stability of the

transformed cell lines. This created vector will be advantageous at this point when compared

to the use of siRNA for gene silencing, which is not a stable way. In the future, this vector can

be used for down regulating other genes of interest in R. oryzae and can also be used for

studying its effect on other metabolic pathways.

In this study, Hygromycin resistance to the R. oryzae CCUG 28959 was shown at levels up to

1000 µg/ml, which has not been reported previously.

Keywords: RNA interference, hpRNA, Silencing vector construction, Rhizopus oryzae,

Ethanol, Lactic acid

Page 7: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

vii

Page 8: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

viii

Abreviations:

adj hpRNA – adjacent hairpin RNA

AMT – agrobacterium mediated transformation

bp – base pairs

BT – Biolistic transformation

C.elegans – Caenorhabditis elegans

CBP – Consolidated bio processing

CCUG – Culture collection, University of Göteborg

cDNA – Complimentary DNA

CUT – cutinase

dsRNA – double stranded RNA

EDTA - EthyleneDiamineTetraAcetic acid

EP - electroporation

FMA – Fumaric Malic acid producing strain

GFP – Green fluoroscence protein

hpRNA – Hair pin RNA

kb – kilo base pairs

LA – Lactate producing strain

LB medium – Luria Bertani medium

LDHA – Lactate dehydrogenase gene A

miRNA – micro RNA

M.oryzae – Magnoparthy oryzae

ORF – open reading frame

PCR – Polymerase chain reaction

PEG – Polyethylene glycol

Page 9: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

vi

PMT – protoplast mediated transformation

pS1 – p silent 1

pSD1 – p silent dual 1

PtrpC – tryptophan gene promoter

qPCR – Quantitative PCR

R.oryzae – Rhizopus oryzae

RdRP – RNA dependent RNA polymerase

RISC – RNA induced silencing complex

RT PCR – Real time PCR

SDS – Sodium dodicyl sulphate

siRNA – Small interference RNA

SOC – Super optimal broth

Taq – thermus aquaticus

Tm – melting temperature

TtrpC – tryptophan gene terminator

UV – ultra violet

Page 10: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

vii

Contents

1. Introduction .................................................................................................................................. 1 2. Background ................................................................................................................................... 3

2.1 RNA interference and its application in Fungi: ....................................................................... 5 2.2 Silencing Mechanisms other than post transcriptional control: ............................................... 7 2.3 Uses and draw backs of RNA silencing: .................................................................................. 8 2.4 Studies in RNA silencing at the University of Borås ............................................................... 9

3. Aim of this study ........................................................................................................................... 9 4. Literature Review ......................................................................................................................... 9

4.1 RNA silencing vectors: ............................................................................................................ 9 4.1.1 pCAP59i and pCAP59/ADE2i: ................................................................................... 10 4.1.2 pHANNIBAL and pHELLSGATE (intron splices hpRNA expressing vectors) :....... 11 4.1.3 Vectors for finding better RNA species for triggering RNA interference: .................. 12 4.1.4 P-Silent1: ..................................................................................................................... 12 4.1.5 PSV-IRT (with inverted repeat transcription silencing constructs): ............................ 14 4.1.6 pFANTAi4 ................................................................................................................... 15 4.1.7 p Silent Dual (pSD1): .................................................................................................. 16 4.1.8 pTROYA: .................................................................................................................... 17

5. Rhizopus oryzae: .......................................................................................................................... 18 5.1 Classification: ........................................................................................................................ 18 5.2 Morphology: .......................................................................................................................... 19 5.3 Cell wall composition: ........................................................................................................... 21 5.4 Protoplast formation: ............................................................................................................. 21 5.5 Propagation: ........................................................................................................................... 22 5.6 Plasmids in Fungi:.................................................................................................................. 24 5.7 Transformation:...................................................................................................................... 25

5.7.1 Protoplast Mediated Transformation using PEG/CaCl2: ............................................. 26 6. Methods and Materials .............................................................................................................. 26

6.1 Genomic DNA extraction from R. oryzae: (Hoskins. L, 1998) ............................................. 26 6.2 Extraction of Plasmids from paper discs: .............................................................................. 27 6.3 Transformation of Competent E.coli cells using CaCl2 (Sambrook and Russell, 2001

modified) ........................................................................................................................................... 28 6.4 Vector construction: ............................................................................................................... 28

6.4.1 Primer designing: ......................................................................................................... 28 6.4.2 Gene sequence to be amplified: ................................................................................... 29

6.5 PCR: ....................................................................................................................................... 30 6.5.1 Reaction mixture concentrations used (Fisher, 2010): ................................................ 30 6.5.2 Thermal cycling parameters: ....................................................................................... 30

6.6 Agarose gel electrophoresis:(Lewis, 2001) ............................................................................ 31 6.7 Purification of PCR product: ................................................................................................. 31 6.8 Plasmid DNA Extraction from E.coli using Alkaline Lysis Method (Winstanley and Rapley,

2000) 31 6.9 Restriction digestion and Ligation: ........................................................................................ 33

6.9.1 Double digestion of PCR product and Plasmid: .......................................................... 33 6.9.2 Inactivation of Restriction enzymes that are not heat sensitive:(uregina) ................... 33 6.9.3 Ligation: ....................................................................................................................... 34

6.10 Protoplast Preparation of Rhizopus oryzae by Using Yatalase (Jahromi and Gheinani,

2011 modified) .................................................................................................................................. 34 6.11 Transformation of Rhizopus oryzae by the Calcium Chloride/Polyethylene .................... 35 6.12 Sub culturing of transformed R. oryzae ............................................................................ 36

Page 11: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

viii

6.13 Primer design for amplifying LDHA gene along with cutinase intron: ............................ 36 7. Results and Discussion ............................................................................................................... 38

7.1 LDHA gene amplification through PCR: ............................................................................... 38 7.2 Psilent 1 with one LDHA gene inserted in it: ........................................................................ 39 7.3 Complete vector after inserting second LDHA gene in to the vector: ................................... 40 7.4 Protoplast formation from R. oryzae: ..................................................................................... 41 7.5 Hygromycin resistance for Rhizopus oryzae: ........................................................................ 41 7.6 Confirmation of transformed R.oryzae and demonstration of the stability in the progeny: ... 42

8. Conclusion ................................................................................................................................... 43 8.1 Draw backs of present work: ................................................................................................. 44

9. Further research ......................................................................................................................... 44 9.1 Optimization and analysis of Fermentation with transformed Rhizopus oryzae: ................... 44 9.2 Changing the inserted LDHA gene sequence in the vector: .................................................. 44 9.3 Improving the silencing vector by inserting eGFP gene: ....................................................... 45

10. References ............................................................................................................................. 46 A1: Preparations ................................................................................................................................... 1

Preparation 1: LB (Luria-Bertani) medium (100 ml) (bioprotocols.info) ........................................... 1 Preparation 2: LB agar plates with ampicillin (bioprotocols.info) ...................................................... 1 Preparation 3: Salt solutions (100 ml each) ........................................................................................ 1 Preparation 4: Vitamin solution (Jahromi and Gheinani, 2011 modified) ......................................... 2 Preparation 5: Trace metal solution (Jahromi and Gheinani, 2011 modified) .................................... 2 Preparation 6: Semi synthetic medium for cultivation of Zygomycetes Fungi (1000 ml) .................. 3 Preparation 7: Potato Dextrose Agar plates ........................................................................................ 4 Preparation 8: Yatalase Digestion Buffer (Takara) ............................................................................. 4 Preparation 9: STC Buffer .................................................................................................................. 5 Preparation 10: PEG solution (25ml) .................................................................................................. 5 Preparation 11: 10X TBE Buffer (bioprotocols.info) ......................................................................... 5 Preparation 12: Spore solution (Jahromi and Gheinani, 2011 modified) ............................................ 6

Appendix:

A1. Preparations

Preparation 1: LB (Luria Bertani medium)

Preparation 2: LB agar plates with ampicillin

Preparation 3: Salt solutions

Preparation 4: Vitamin solution

Preparation 5: Trace metal solution

Preparation 6: Semin synthetic medium for cultivaion of Zygomycetes Fungi

Preparation 7: Potato Dextrose Agar plates

Preparation 8: Yatalase Digestion Buffer

Preparation 9: STC Buffer

Preparation 10: PEG solution

Preparation 11: 10x TBE Buffer

Preparation 12: Spore solution

A2. List of equipment used

A3. List of Chemicals used

Page 12: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

1

1. Introduction

The briskly growing demand for bio fuels is due to the realization that these are the only

substitutes for the present fossil fuels which covers more than 95% of the world’s

transportation energy requirement in the near future. Present Bio fuels, Ethanol and Bio diesel

have an advantage of being integrated in to the present established huge petroleum

infrastructure. This makes them a solution for the present as well as future generations for

national and global fuel insecurity (Suzzane and Flavin, 2007). First generation bio fuels can

be considered as those derived directly from sugars, starch and vegetable oils. Second

generation bio fuels are those derived from sustainable feed stocks (non food materials) such

as wheat straw and they mainly do not have impact on global food reserves. Second

generation Bio fuels have been developed due to the limitations of global food reserves being

diverted to fuels (Tye et al., 2011, Taylor et al., 2009, Hayes, 2009, Ueng and Gong, 1982,

Wu et al., 1986, Zhang et al., 1995). Third generation bio fuels are derived from algae which

are grown in natural water bodies or artificial environments. Third generation bio fuels

overcome the limitation of first and second generation bio fuels mainly the land, fresh water

and food security risks (Sergeeva et al., 2008). Much research has been done in developing

economical ways for this bio fuel production. In the second generation bio fuels, the carbon

source is not available for the microbes in the simplest form. Direct enzymatic hydrolysis of

streams from agricultural, industrial or house hold waste is not so efficient due to high

stability of the raw material and not easily accessible for enzymes and microbes. Waste

streams have complex mixtures of sugars. Many micro organisms that were used for ethanol

production from food based substrates are capable of using only hexose sugars. Different

methods are in practice for the pre-treatment process such as milling, acid hydrolysis,

irradiation, hot water treatment and alkaline hydrolysis (figure 2.1). Each method has its own

advantages and draw backs. Methods such as use of acids, hot water, ammonia, and lime for

pre-treatment are capital intensive. Whereas methods like biological pre-treatment are very

slow (Taherzadeh and Karimi, 2008). The choice of pre treatment method can be done

according to the available resources. Bio refinery is the possible way of using the Biomass

that is available abundantly in the global market as cheap substrate.

Page 13: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

2

Figure2.1: The various pre-treatment and subsequent conversion technologies possible for the

treatment of lignocellulosics (Hayes, 2009)

In a new approach, cellulase production, cellulose hydrolysis and fermentation combined in to

a single process known to be as Consolidated Bio Processing (CBP). This method seems to be

a better alternative than all existing methods for bio fuel production from biological wastes.

This approach is mainly done in two ways. Engineering naturally occurring cellulolytic

microorganisms to have high ethanol yield and productivity known to be CBPI organisms and

engineering non cellulolytic microbes to produce heterologous cellulase system known to be

CBPII organisms (Lynd et al., 2005, Amore and Faraco, 2012). Ethanologic bacteria like

Zymomonas mobilis, Escherichia coli and Klebsiella oxytoca, as well as the yeasts

Saccharomyces cerevisiae, Pachysolen tannophilus, Pichia stipitis, and Candida shehatae

were modified to express various genes for cellulolytic enzymes (Hasunuma and Kondo,

2011, la Grange et al., 2010, Elkins et al., 2010, Amore and Faraco, 2012). Most of the genes

coding for the heterologous cellulolytic enzymes were of eukaryotic origin and they have

many difficulties in being produced and secreted in active form in prokaryotic organisms.

Also the amount of enzymes produced by these organisms was not sufficient for complete

utilization of cellulose. This makes it difficult to work with CBPII organisms. The working

model for CBPI organisms was diagrammatically represented in figure 2.2.

The cellulolytic thermophilic bacteria Geobacillus thermoglucosidasius, Thermoaner-

obacterium saccharolyticum and Thermoanerobacter mathranii have been described as

potential microbes in the category of CBPI organisms (Taylor et al., 2009). When comparing

the best productive cellulolytic bacteria with the fungi like Trichoderma reesei that can

produce high amounts of cellulases (100g of cellulases / liter broth) for complete

saccharification of lignocelluloses, these bacteria seem to be less effecient (Schuster and

Schmoll, 2010). Some other filamentous fungi belonging to the genera Neurospora,

Aspergillus, Trichoderma, Monilia, Rhizopus, Paecilomyces and Fusarium have the

capability of directly converting celloloses to ethanol with other byproducts like acetic acid,

fumaric acid and lactic acid (Singh et al., 1992, Meussen et al., Stevenson and Weimer, 2002).

Page 14: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

3

Figure 2.2: A conceptual whole-cell biocatalyst for producing liquid transportation fuels (Elkins et al.,

2010).

To have more productivity of ethanol by using the fungal species that come under CBPI, the

metabolic pathways which are responsible for the reduction of Ethanol production are to be

controlled. It was reported in several studies that most of the organisms have the similar

pathways for the production of ethanol. Pyruvate was produced by glycolysis and it is the

main intermediate from which the metabolic pathways split to give different products

according to the microbe’s environment. Most of the time, there will be production of one or

more bi products varying in amounts. It was demonstrated that the carbon flux will be

distributed between the byproducts in varying levels and that determines the final yield of the

individual bi product. By controlling the enzymes responsible for the undesirable products,

the yield and productivity of desired ones can be increased. It was also demonstrated that

complete inactivation of genes responsible for undesirable products may cease the cell

growth. In Saccharomyces cerevisiae, by inserting Lactate Dehydrogenase (LDH) gene (for

lactic acid production) and disrupting Pyruvate decarboxylase gene (for ethanol production),

there was reduction in cell growth with little increase in Lactate production (Adachi et al.,

1998, Ishida et al., 2006).

2. Background

Among the fungi that were capable of producing ethanol from lignocelluloses, Rhizopus

oryzae (figure 2.1) has its own importance. It was reported of having the capability of

assimilating variety of sugars such as glucose, galactose, mannose and xylose (Edebo, 2000).

This zygomycetes species was extensively used for production of cellulolytic enzymes

(Amadioha, 1993, Archer and Wood, 1994), proteases (Tunga et al., 1999), lactic acid

(Skory, 2004b) and also ethanol (Abedinifar et al., 2009, Büyükkileci et al., 2006). Tolerance

to the inhibitors present in lingo cellulose acid hydrolyzates, ability to grow in high

temperatures, and valuable biomass content makes Rhizopus oryzae attractive (Karimi et al.,

2006, Taherzadeh et al., 2003, Millati et al., 2005, Ferreira. A.J et al., 2012).

Page 15: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

4

Figure 2.1: Rhizopus oryzae CCUG 28958

It was reported that the lactic acid was the major by product formed along with ethanol during

fermentation (Taherzadeh et al., 2003). As previously discussed, lactate production was

increased by deleting the pyruvate dehydrogenase gene, The gene coding for lactate

dehydrogenase (LDH) (enzyme for converting pyruvate to lactate) can also be deleted or

down regulated to increase ethanol production (Cai et al., 2011). LDH gene can also be

knocked down using post transcriptional modification of mRNA through RNA interference

known to be quelling in fungi (Gheinani et al., 2011). It was found out that two distinct lactate

dehydrogenase enzymes were present in rhizopus oryzae. One is NAD+ independent and the

other is dependent. The dependent one can catalyze the conversion of pyruvate to l-lactic acid.

Independent one has the additional capability of catalyzing opposite reaction, that is from l-

lactate to pyruvate. It was also found out that the second enzyme could not utilize D-lactate

(Pritchard, 1971). The LDH gene from Rhizopus oryzae was cloned in E.coli. The RNA was

used to make cDNA which served as a template for amplification with PCR primers referred

to as universal ldh primers (5´-ACACGCCCATCCGAGCAGG-3´ and 5´-

GCACAGGCACCA ATTCCATAAAAC-3´). These universal primers were designed to

anneal to regions that are identical in both ldhA and ldhB genes. Comparisons between cDNA

and genomic sequence revealed that there were no introns present in this gene. Even ldhB

gene also has no introns in it. ldhb gene was amplified from the cultures grown on lactic acid

but not from the cultures grown on Glucose medium (Skory, 2000). The increase of ethanol or

decrease of lactate could be possible only by controlling the gene expression. This can be

done by transforming the organisms which will result in the diversion of metabolic pathways

towards desired ethanol production. Diagrammatic representation of Glucose metabolic

pathway that occurs in microorganisms was shown in figure 2-2.

Page 16: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

5

Figure 2-2: Simplified overview of the main fermentation routes with glucose as carbon source.

The numbers indicate key enzymes in this pathway: 1, pyruvate decarboxylase (PDC); 2, alcohol dehydrogenase

(ADH); 3, lactate dehydrogenase (LDH); 4, pyruvate carboxylase (PYC); 5, malate dehydrogenase (MDH); 6,

fumarase (FUM). (Meussen et al., 2012)

2.1 RNA interference and its application in Fungi:

Molecular biotechnology has been advanced since the time DNA sequencing of various

organisms was done from the simplest organisms like prokaryotes to fully fledged multi

cellular eukaryotes. The complete genome sequencing of S.cereviciae in the year 1996 made

it possible to explore more functional properties of molecular genetics of this species. Since

then a lot of genomes of various fungi were sequenced due to their compactness. These

known genes help in gaining much knowledge regarding the reverse genetic approaches for

genetic control (Nakayashiki and Nguyen, 2008). Much effort was laid by researchers for

getting the functional information of various genes in all organisms. There are few methods

by which the function of a particular gene can be determined. This identification of genetic

functions will be easier in prokaryotes where there are fewer complications when compared to

Eukaryotes. The old and mostly followed method for knowing the gene function is Gene

Knock out. This is very laborious work since and also there should be some phenotypic effect

that could be determined. Recently gene knock down mechanism was discovered in the

eukaryotic organism Caenorhabditis elegans by double stranded RNA and they were

awarded Nobel Prize in the year 2006 for their discovery (Fire et al., 1998, Nobelprize.org,

2006). It was found out that the silencing of gene can be done not only through the knockout

of specific DNA but also through some RNA related approaches. This approach seems to be

beneficial when gene targeting approaches fail, multiple copies of a gene of interest are

Page 17: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

6

present in the genome or iso genes might compensate for the knockout of the deleted gene

(Meyer, 2008).

Three RNA-based methods antisense RNA, hammerhead ribozymes and RNA interference

have been shown to be valuable tools for gene silencing in eukaryotes. Antisense RNA

constructs were used in silencing genes in different organisms like A. Oryzae (Zheng et al.,

1998) and A. awamori (Moralejo et al., 2002). Many other genes were silenced successfully in

various filamentous fungi using antisense RNA constructs (Kitamoto et al., 1999, Moralejo et

al., 2002, Lombraña et al., 2004). But complete silencing of a gene was not yet achieved using

this approach till date. Complete silencing of particular gene may be lethal to the organisms

and this tool will be beneficial for this type of problems (Bautista et al., 2000). Hammerhead

ribozyme mediated silencing was another approach where the substrate recognition arms of a

ribozyme can be altered such that they are complimentary to the target mRNA to be degraded.

(Akashi et al., 2005). This approach for repressing a gene in filamentous fungi was

demonstrated by Müller and coworkers (Müller et al., 2006). In the RNA interference

approach, small RNA molecules direct the complexes that can degrade targeted mRNA

molecules thereby blocking the translation in to polypeptides. This RNA silencing may also

occur through RNA directed DNA methylation or heterochromatin formation and

translational repression (Schumann et al., 2010). Many species of small RNA were reported

for having silencing effect in plants. Some of them are small interference RNA (siRNA)

(Gheinani et al., 2011), micro RNA (mi RNA) (Molnar et al., 2007), natural anti sense RNA

(Zheng et al., 1998). All these small RNA’s are derived from double stranded or hair pin

RNA precursors (Eamens et al., 2008). This knock down mechanism or gene silencing is

termed as RNA interference in animals, co suppression in plants and quelling in fungi

(Schumann et al., 2010). RNA interference in fungi also does not cause complete suppression

of targeted gene similar to antisense RNA mediated silencing.

The protein complex named Dicer was responsible for the production of siRNA’s from

precursor double stranded RNA’s (Zamore et al., 2000). One of the strands from 20-25 long

small RNA will be unwound to form a passenger strand and guiding strand. The guiding

strand guides the RNA induced silencing complex (RISC) along with an AGO protein

towards the targeted mRNA. This finally leads to the cleavage and results in silencing of the

specific gene (Paroo et al., 2007, Hammond et al., 2001, Schumann et al., 2010).

Page 18: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

7

Figure 4.1.1.1: The RNA interference pathway. Long double-stranded RNA (dsRNA) or small hairpin

RNA (shRNA) is processed by Dicer to form a small interfering RNA (siRNA), which associates with

RNA-induced silencing protein complex (RISC) and mediates target sequence specificity for subsequent

mRNA cleavage (Rutz and Scheffold, 2004).

When coming to the gene silencing mechanisms in fungi, it was identified in many organisms

like N .cassa (Romano and Macino, 1992), A. nidulans (Khatri and Rajam, 2007), M.oryzae

(Kadotani et al., 2003), R.oryzae (Skory, 2004a), A.oryzae (Yamada et al., 2007), A.

parasiticus (McDonald et al., 2005) A.niger (Barnes et al., 2008). A prerequisite for this

method is that the fungal strain comprises components of the RNAi silencing machinery, such

as RNA-dependent RNA polymerase (RdRP), dicer-like proteins and the RNA-induced

silencing complex (RISC). In most of the studies, RNA interference in fungi was done to

study and control the pathogenic pathways over plants and animals. Different strategies were

employed for inducing silencing of various types of genes in different fungi like direct

delivery of siRNA, dsRNA or through expression of hpRNA constructs. Each method has its

own advantages and disadvantages. The efficiency of gene silencing varied significantly for

different methods (Kadotani et al., 2003).

2.2 Silencing Mechanisms other than post transcriptional control:

The small RNA’s that were produced from precursor RNA molecules help in silencing the

targeted genes in various ways. One of them was as described above where the small RNA’s

bind to targeted mRNA and leads to degradation, there by silencing the gene. The other way

is by inducing chromatin modifications within the target gene and thereby silencing its

transcription. In both types of silencing the same machinery like RNA-induced silencing

complex (RISC), various Argonaute family proteins are involved. If the guiding RNA with

mature RISC complex exactly matches the target mRNA, it leads to the degradation known as

―Slicing‖ of the mRNA and this is accomplished by Argonaute family proteins known as

―Slicer‖. If the guiding RNA is not much complimentary to target mRNA, then the RISC can

be directed into the nucleus where it recruits other proteins that modify the chromatin around

the promoter of the gene complementary to the guide RNA as shown in the figure below. The

proposed mechanism was that the regions of centromere were transcribed to produce RNAs

Page 19: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

8

that hybridize with other RNAs from the same region. The resulting dsRNAs are recognized

by Dicer and cleaved to produce siRNAs which finally directs the machinery to the

centromeres. Very small amount of dsRNA is sufficient for the induction of near complete

shutdown of targeted gene expression. The main enzyme that was known to be responsible for

this efficiency was RNA-dependent RNA polymerase (RdRP). This protein helps in

amplifying the inhibitory signal by generating dsRNAs after recruiting the mRNA by the

original siRNA. This protein was not yet identified in mammalian cells. But the slicing

mechanism was catalytic and each RISC complex could slice many mRNA molecules. The

other way is by interfering with translation mechanism of the target mRNA. All this shows

that the silencing mechanism was completely different in simple organisms and higher

eukaryotes (figure 2.2.1) (James D. Watson, 2008).

Figure 2.2.1: RNAi switches off the expression

of a gene when dsRNA molecules that have

homology to that gene are introduced, or made, in

the cell. This effect involves processing of the

dsRNA to make siRNA and miRNAs by the

enzyme Dicer. The siRNA and miRNAs direct a

complex called RISC (RNA-induced silencing

complex) to repress genes in three ways. It attacks

and digests mRNA that has homology with

siRNA; it interferes with translation of those

mRNAs; or it directs chromatin modifying

enzymes to the promoter that direct expression of

those mRNAs (James D. Watson, 2008).

2.3 Uses and draw backs of RNA silencing:

When compared to gene knock out methods, gene knock down strategies require information

of only small stretches of sequence information. So this can be implemented even in the

organisms with less genetic information. As silencing occurs at post transcriptional level,

multiple copies of gene located at multiple loci can be silenced. Certain genes coding for

proteolytic enzymes that belong to a common family could be silenced simultaneously by

selecting the conserved region for constructing the vector. This gives a great benefit when

comparing to gene knock out methods where each gene needs to be treated separately. To

control the gene silencing, inducible promoters can be employed. In the similar fashion,

strong promoters help in increasing the efficiency of gene silencing (Weld et al., 2006,

Page 20: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

9

Schumann et al., 2010). There were also different levels of RNA interference observed in the

transformants. This can be employed in knowing the minimum drug dosage levels required to

change or inhibit a particular phenotypic effect. This cannot be known in knock out models.

Also the genes that are essential for a cell survival can be knocked down up to certain levels

without having lethal effect on cells growth (Liu et al., 2002, Kadotani et al., 2003).

The efficiency of gene silencing was not stable and constant in all the transformants. It is

difficult to select the transformants with high level of gene silencing or having similar levels

of silencing (Mouyna et al., 2004). This can be solved by co suppressing the gene that has

phenotypic effect. Reporter gene such as eGFP (coding for green fluorescence protein) comes

under this category. In this approach, the reporter gene expressing strain will be used for

studying silencing effect and can be transformed with silencing construct that can silence

gene of interest along with reporter gene (Fitzgerald et al., 2004, Krajaejun et al., 2007). Only

phenotypically distinct transformants can be selected after transformation and this is also not

very precise. An other major problem with this gene silencing approach is off targeting effect.

As only small sequence of RNA was being used, there may be undesired complementary

targets that have a chance to get silenced along with desired ones (Nguyen et al., 2008,

Gheinani et al., 2011).

2.4 Studies in RNA silencing at the University of Borås

Research at the University of Borås has earlier shown that it is possible to silence the LDHA

gene in Rhizopus oryzae CCUG 28959 by siRNA and thereby decreasing the ethanol

production (Gheinani et al., 2011). The drawback with this silencing method is that there will

be no permanent change to the organism and the silencing effect will not be inherited by its

progeny. For such a change the genome has to be altered for example by transforming an

integral silencing vector.

3. Aim of this study

The purpose of this study was to construct a stable integral hp RNA expression vector for

permanent silencing of genes in Rhizopus oryzae

4. Literature Review

4.1 RNA silencing vectors:

The first silencing construct used for silencing in pathogenic fungi Cryptococcus neoformans

was reported by Hong Liu et al. The plasmid constructs pCAP59i and pCAP59/ADE2i were

used to silence CAP59 and ADE2 genes (Liu et al., 2002). High through put versatile

silencing vectors pHANNIBAL and pHELLSGATE were created for inducing silencing in

plants (Wesley et al., 2001). Till date, many groups have used hpRNA with intron for RNA

interference. P-Silent1 vector was developed from pBSTRP-PT (Kadotani et al., 2003) which

was similar to pHANNIBAL and pHELLSGATE for silencing in ascomycete fungi

(Nakayashiki et al., 2005). Shafran et al have introduced the Gateway technology into pSilent-

1 and established the high-throughput RNAi vector, pTroya (Shafran et al., 2008). Similarly,

Page 21: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

10

pFANTAi4, which uses the Gateway technology and a green fluorescent protein (GFP)

sentinel system, has been developed for the human pathogenic fungus Blastomyces

dermatitidis (Krajaejun et al., 2007). Using eGFP gene as a marker to measure the efficiency

of silencing, P Silent Dual1was developed by modifying Psilent1 which has advantages of

both the vectors pFANTAi4 and P silent1(Nguyen et al., 2008).

Brief description of construction and application of silencing vectors mentioned above

follows:

4.1.1 pCAP59i and pCAP59/ADE2i:

The first gene CAP59 codes for a protein essential for synthesis of a capsule essential for

virulence and second one ADE2 for phosphoribosyl amino imidazole carboxylase. Silencing

of first gene gave colonies with different hydrophobicity than wild type and second gene

resulted in pink colored colonies due to accumulation of adenine biosynthetic intermediates

and this worked as a phenotypic marker for identifying the transformants having silencing

effect.

For construction of a vector that was capable of expressing hpRNA, Liu and coworkers used

two duplicate sequences of 500 bp CAP59 gene in opposite direction with a spacer sequence

of 250 bp from eGFP gene. This hair pin construct was preceded by cryptococcal actin

promoter and followed by GAL7 terminator (figure 4.1.1.1).

Figure 4.1.1.1: Plasmid design. pCAP59i was used to interfere with CAP59 expression, and

pCAP59/ADE2i was used for tandem interference with both CAP59 and ADE2 (Liu et al., 2002).

C.neoformans cells of serotype D were transformed by electroporation with linear pCAP59i

and grown on appropriate media for the synthesis of capsule. This transformants were

compared with wild type and knock out mutants of CAP59 gene. They also had closer

examination of transformants under light microscope. Closer clump formation was observed

in transformants which was similar in knock out mutants. India ink staining as well as

monoclonal antibody specific tests were performed for testing the presence of capsule. All

these tests resulted in common conclusion that the gene was silenced. The construct was

modified by deleting the promoter sequence and checked for interference effect. This showed

no modified phenotype which concludes that the expression of hairpin construct was essential

for RNA interference. Quantitative RT-PCR method was done to check the levels of CAP59

mRNA and the results were in accordance to expected levels. Similarly ADE2 gene was

silenced by inserting the gene along with CAP59. Simultaneous silencing of genes was

observed even though there was difference in their levels for individual genes. Several levels

of silencing were observed which hypothesizes that the efficiency of silencing was not similar

in all the transformants. This may depend on the variation in the level of transcription of the

hair pin construct. This variation may be due to modifications in the transformed construct

Page 22: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

11

inside the host. To improve the stability of construct and reduce modifications, cryptococcal

telomere sequences were incorporated in to pCAP59i (pCAP59i-tel) (Liu et al., 2002).

This proved that dsRNA have significant role in interference mechanisms in fungi.

4.1.2 pHANNIBAL and pHELLSGATE (intron splices hpRNA expressing vectors) :

Two types of constructs that can express adjacent hpRNA (adj-hpRNA) and hpRNA were

tested to compare the best construct for silencing. The results showed that hpRNA construct

was the best one which has more efficiency compared to siRNA and adj-hpRNA for

silencing. For constructing hpRNA expressing vectors, there is a need to have two copies of

target gene in an inverted repeat orientation and it involves more steps.

Intron spliced hp RNA constructs were designed by inserting functional intron in the place of

spacer gene in between the two arms to increase the stability of inverted repeat DNA in

E.coli. These construct with intron showed more efficiency than the constructs without intron.

pHANNIBAL was constructed with all the optimized properties and this gave a better

silencing in plants (figure 4.1.2.1) (Wesley et al., 2001).

Figure 4.1.2.1: PCR products from the target gene are cloned into the polylinkers of pHANNIBAL

conventionally; restriction sites added by the primers ensure the correct orientation of the resulting sense

and anti-sense arms (Wesley et al., 2001).

A high throughput vector pHELLSGATE was constructed which involves less number of

steps. It was designed such that single PCR product with appropriate primer pairs was

sufficient to incorporate the gene of target simultaneously to form two arms of hpRNA

construct. A gene sequence ccdB which is lethal for E. coli DH5α was included on either side

of the arms to ensure the correct formation of arms with the gene of interest (figure 4.1.2.2)

(Wesley et al., 2001).

Figure 4.1.2.2: The attB1 and attB2 sequences on a single PCR product facilitate the

recombination of one sense-orientated and one anti-sense orientated molecule into each

molecule of pHELLSGATE when incubated with BP clonase (Wesley et al., 2001).

Page 23: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

12

4.1.3 Vectors for finding better RNA species for triggering RNA interference:

Various constructs were employed to determine the efficient RNA species that can trigger

silencing mechanisms in filamentous fungi. Different constructs were designed to express

eGFP sense, anti sense, sense-antisense, sense-sense, anti sense- anti sense strands and these

were transformed into eGFP expressing transformants. Silencing efficiency was measured

according to the reduced fluorescence. It was found out that constructs with hpRNA

expressing cassette have highest silencing efficiency among them. This shows that hpRNA

expressing will be the better trigger for silencing.

For eGFP gene expression, a plasmid pEGFP75 was used for transformation. Later pEGFP-A,

pEGFP-SA, pEGFP-SS, pEGFP-AA (figure 4.1.3.1) were used to express various silencing

constructs to test the efficiency.

Figure 4.1.3.1: Figure: Diagram of

transformation plasmids used in this

study. Arrows indicate the orientation

of the enhanced green fluorescence

protein gene. Segments used as probes

in Southern Blot analysis are

indicated by thick bars. A HincII

fragment of the beta-glucuronidase

(GUS) gene was used as a spacer. All

the constructs were under the control

of the Aspergillus nidulans trpC

promoter (PtrpC) and terminator

(TtrpC) (Kadotani et al., 2003).

Lower mRNA levels of eGFP gene were detected in the transformants with less fluorescence.

Three different sizes of siRNA’s were detected ranging from 19-23 as reported in animals and

plants and this was correlated with the silencing efficiency. These results also give strong

evidence that RNA interference in fungi in many cases is similar to that in animals and plants

(Kadotani et al., 2003).

4.1.4 P-Silent1:

A silencing vector capable of expressing hpRNA with an intron in between the two arms in

fungi was constructed. The efficiency of this construct was tested in rice blast fungus M.

Oryzae by testing the silencing of eGFP gene. This vector was also used to silence two

endogenous genes which showed various levels of silencing.

For optimizing the length of intron sequence for better silencing effect, three different introns

of various lengths were tested using the vector pBSTRP-PT (Kadotani et al., 2003). trpC

promoter and terminator were used for the construction of vector. Two copies of eGFP gene

were inserted in the opposite orientation to form hpRNA. Among different constructs with

Page 24: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

13

introns used, construct having CUT gene of length 142 bp as spacer showed better efficiency

of silencing.

P-Silent1 vector was constructed by including multiple cloning sites with unique restriction

sites for inserting PCR amplified sense and antisense strands with appropriate set of primers

for gene of interest with CUT gene as intron in between (figure 4.1.4.1).

Figure 4.1.4.1: Schematic representation

of the silencing vector pSilent-1, and the

sequences of multi-cloning sites and

splice junctions in the vector.

(A) A map of pSilent-1. Ampr,

ampicillin-resistant gene; Hygr,

hygromycin- resistant gene; IT, intron 2

of the cutinase (CUT) gene from M.

oryzae; PtrpC, A. nidulans trpC

promoter; and TtrpC, A. nidulans trpC

terminator.

(B) Restriction sites are underlined.

Asterisks indicate

unique sites in the silencing vector. Italic

nucleotides indicate sequence from the

cutinase gene and bold letters represent

5_ and 3_ splice sites (Nakayashiki et al.,

2005)

Measurement of fluorescence, hydrophobicity test and RT PCR analysis showed significant

silencing effect with varied levels of silencing in the transformants. A study was done by

doing southern blotting analysis on the transformants that lost the silencing effect. This

showed that the loss of silencing effect was due to the loss of intact eGFP hpRNA expressing

construct and that was not due to epigenetic reversion. This epigenetic reversion was due to

rearrangements that occur after transformation in filamentous fungi. By maintaining required

growth conditions can help in reducing this effect. This was applicable to pSilent1 as it

contains hygromycin resistance gene. It was also showed that the conidiophores of strongly

silenced transformants also have the silencing effect. This shows that silencing initiated in the

stable transformants can be transferred to asexual progenies and also pSilent1 can be used as

an efficient vector for silencing gene of interest in ascomycete fungi (Nakayashiki et al.,

2005). The complete sequence of p Silent1 vector was also available with the accession

number AB303070.2.

Page 25: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

14

4.1.5 PSV-IRT (with inverted repeat transcription silencing constructs):

RNA interference was employed to down regulate a gene that was responsible for melanin

production in pathogenic fungi that belongs to Ophiostoma species that grows in trees. The

frequency of homologous recombination for this species was very low and this made the

determination of gene function very challenging. A silencing construct was made with

inverted repeat transcription sequence which can produce hpRNA and result in silencing of

targeted gene.

The plasmid pCAMIA-0380 was used for developing PSV binary vector. The promoter and

terminator used in the construction were of trpC origin and they were amplified from

pCB1004 and pAN7-1 (Kitpreechavanich et al., 2008) respectively through PCR

amplification. The spacer between sense and anti sense was from an oligonucleotide sequence

used in the amplification of antisense fragment. Hygromycin B and Ampicillin resistance

cassettes were also amplified and ligated into the vector by PCR reactions with appropriate

primers having restriction sites (figure 4.1.5.1).

Figure 4.1.5.1: IRT silencing constructs: (A) Schematic representation of pSV vector with IRT; shaded

box denotes the intron sequence, (B) RNAi targeted region of the PKS1 gene.

The NCBI GenBank accession numbers forPKS1 genes are: DQ372811 for O. piceae (AU55-3),

and AF411603 for O. floccosum (387 N). Op-PKS1-1 is located 1038–1336 bp from the start codon of the

AU55-3 PKS1 gene; Op-PKS1-2, 53–257 bp; Op-PKS1-3, 4076–4338 bp; Op-PKS1-4, 53–1336 bp; Op-

PKS1-5, 3669–4338 bp. White boxes indicate exon. B, BamHI; Bg, BglII; E, EcoRI; H, HindIII; P, PstI;

S, SacI; S2, SacII; Sa, SalI; Sp, SpeI (Tanguay et al., 2006).

Agrobactrium mediated transformation was used for transformation and the transformants

were selected by growing in medium having 300 μg/ml of hygromycin B.

Southern blotting and RT-PCR analysis were done to know the interference effect.

Transformants with high levels of silencing shown very low pigmentation and the mRNA

levels were also very low and this reveals the post transcriptional down regulation of target

gene. The silenced transformants had other physiological and morphological differences when

Page 26: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

15

compared to wild type like production of soluble pigmentation when grown on agar medium

and also no spore formation. This concludes that untargeted genes were silenced with the

silencing construct used. Copy number and the position of the coding sequence of the target

gene used for construction also affected the silencing efficiency and this was analyzed by

constructing various vectors with different gene lengths and sizes. The transformants were

grown for two months on sterile sapwood and tested for the reversion rate. The conidia

collected showed no reversion and this suggests a very stable method for silencing genes in

fungi.

The group also suggested that the same interference construct can be employed to silence the

gene between the closely related species having similar gene sequence (Tanguay et al., 2006).

4.1.6 pFANTAi4

Using Gateway® Cloning Technology from Invitrogen, a vector was designed where it

includes the target gene along with the eGFP gene. This helps in getting the system where

simultaneous silencing of both the genes was achieved and the green fluorescence acts as an

indicator for knowing the level of silencing. In the construction similar to pHELLSGATE,

two cassettes with two copies of ccdB genes each were inserted in either of the arms in

inverted orientation next to inverted copies of GFP gene. H. capsulatum H2AB promoter was

employed upstream to the hp RNA construct for expression (figure 4.1.6.1).

Figure 4.1.6.1: Map of the destination vector

(pFANTAi4) demonstrating two copies of GFP

(470 bp) that were cloned next to two copies of a

Gateway cassette, in convergent orientation. The

Gateway cassette will be replaced by an attB-

flanked target gene PCR product to construct a

GFP-target gene RNAi vector. Two enzymatic

recombination steps

(BP and LR clonase reactions) are required for

RNAi vector construction

(see Materials and Methods for details). kanR,

kanamycin resistance; hygR, hygromycin

resistance; Term, terminator; LB and RB, left

border and right border (Krajaejun et al., 2007)

For ensuring the transcription of complete

construct, GFP gene copies were placed at

the ends of intended RNAi construct. It was

also shown that the length and location of the target sequence should be optimized empirically

for the maximal RNAi effect. Agrobacterium mediated transformation was employed for

delivering the silencing construct in to dimorphic

fungus B. dermatitidis. Various genes were tested for silencing efficiency and LACZ was one

of them. Sequence from 3’ end that was used in the construction shown more silencing

efficiency than the sequence from 5’ end. Different lengths of genes and loops were used to

find the better combination for gene silencing. It was observed that shorter loop and longer

gene sequence showed more efficiency among all combinations. When several lengths (0.9

kb, 1.5 kb and 3 kb) of LACZ gene were employed, the shorter ones showed more efficiency

Page 27: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

16

of silencing. From these observations, it was suggested that for any gene to be silenced,

optimal length and position of target gene to be used should be found out for having better

silencing construct (Krajaejun et al., 2007).

4.1.7 p Silent Dual (pSD1):

The construction of hpRNA expressing vector was limited to small and moderate scale as it

involves two steps of oriented cloning. Gateway® technology proves to be an alternative way

for this limitation. The vector was developed with opposite dual promoters and targeted gene

silencing can be done by a single step cloning process. In this construction, sense and

antisense strands of the target gene will be transcribed by two opposing RNA polymerase II

promoters.

eGFP gene was introduced along with the cassette to know the level of silencing of the target

gene. The two promoters used were of different origin and they were trpC and gdp promoters

(figure 4.1.7.1).

This construct was used for functional analysis of calcium signaling proteins in the genome of

rice blast fungus M. oryzae. Three vectors were used simultaneously to test the efficiency

namely pSD1 with eGFP gene (pSD1-GFP), pS1-CUT (p silent 1 vector with cutinase intron

expressing eGFP gene hpRNA) and empty pSD1 as negative control.

Figure 4.1.7.1: Schematic diagram of the RNA-silencing vector, pSilent-Dual1 (A), and the nucleic acids

sequence in the multiple cloning sites (B). Asterisks indicate unique sites in the vector. PtrpC, Aspergillus

nidulans trpC promoter; Pgpd, A. nidulans gpd promoter; Ampr, ampicillin-resistant gene (Nguyen et al.,

2008).

It was observed that pS1 CUT had more silencing efficiency than pSD1-GFP. mRNA levels

were also in consistent to this result. Calcium signaling analysis was done efficiently using

Page 28: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

17

pSD1 in M. oryzae and this silencing construct can be employed in other similar fungi. This

vector works under promoter that found to be effective in many fungal species. Single step

non-oriented cloning also helps in constructing a vector for multiple genes in less time. The

working model of pSilent-Dual1 was shown in the figure 4.1.7.2.

Figure 4.1.7.2: Schematic diagram of the high-throughput RNA-silencing system by pSilent-Dual1 (pSD1).

A pool of DNA fragments were amplified by PCR using specific sets of primers and genomic DNA, and

inserted into pSD1G (pSD1 with a fragment of the eGFP gene). Resulting recombinant plasmids were

screened by colony PCR, and each of them was introduced into a GFP-expressing Magnaporthe oryzae

strain. The M. oryzae transformants were grown in a 96-well plate, and GFP fluorescence of the colonies

was measured using a fluorescent plate reader. Northern blot or qPCR analysis was performed with M.

oryzae transformants showing low or no GFP fluorescence, and ones with a reduced mRNA level were

further subjected to phenotypic analyses (Nguyen et al., 2008)

The lower efficiency of pSD1 when compare to pS1 may be due to that the synthesized RNA

complimentary RNA strands have to be combined physically to form dsRNA. When coming

to pS1 transcript, the sense and anti sense strands have an intron in between. This enables

them to self fold to form hpRNA. pSD1 was modified by replacing gdp promoter with trpC

promoter. This also did not improve the silencing efficiency (Nguyen et al., 2008).

4.1.8 pTROYA:

The vector P silent 1 was used as the basis for constructing the vector pTROYA. They have

employed Invitrogen Gateway technology for getting high through put silencing construct. A

Gateway cassette was inserted in to XhoI site in the multiple cloning site. This cassette has

recombination sites, chloramphenicol resistance gene and ccdB gene. Similar cassette was

inserted at StuI site in the opposite direction. The construct was verified by restriction

mapping and partial sequencing around the cloning sites (Shafran et al., 2008).

Page 29: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

18

5. Rhizopus oryzae:

Rhizopus oryzae comes under the order Mucorales in the phylum Zygomycota. The main

difference between Rhizopus genus and others (Aspergillus, Fusarium and Penicillium) is its

ability to produce fumaric acid (Kitpreechavanich et al., 2008). In few parts of Asia, Rhizopus

species was being used in foods such as tempeh, peka, ragi, and loog-pang (Kitpreechavanich

et al., 2008). Some strains of this species are regarded as opportunistic human pathogens and

mostly responsible for mucor mycosis infections (Ibrahim et al., 2010).

5.1 Classification:

Taxonomic classification of Rhizopus (Zamore et al., 2000) :

Kingdom ---- Fungi (Myceteae)

Phylum ---- Zygomycota

Class ---- Zygomycetes

Order ---- Mucorales

Family ---- Mucoraceae

Genus ---- Rhizopus

Recent reclassification was done according to the organic acid producing capabilities. Two

classes were divided as Lactic acid producing (LA) and Fumaric – Malic acid producing

(FMA) and these were in accordance to the phylogenetic classification as well when analysed

with their rDNA ITS, lactate dehydrogenase B, actin, translation elongation factor-1a and

genome wide AFLP. R. oryzae has two genes for lactic acid production ldhA and ldhB. It was

found that FMA lack ldhA gene and is responsible for lactic acid production in lactic acid

producers. Abe and coworkers proposed that FMA producers of Rhizopus species can be

called as R. delemar and LA producers comes under R. oryzae (figure 5.1.1) (Tanguay et al.,

2006).

Page 30: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

19

Figure 5.1.1: Classification of the organisms of biotechnological interest within the fungal kingdom, with a

focus on Zygomycetes, and specifically the microorganism studied in the current work (Lennartsson, 2012).

Different strains of this species are being used for production of various commercially

valuable products such as l-lactic acid, Ethanol and Fumaric acid (Table 5.1.1).

Table 5.1.1: Literature data on the main fermentation end product production by Rhizopus oryzae strains

(Kitpreechavanich et al., 2008). Strain used in this study was marked in the table.

Product R.Oryzae Strain Yield

(g product/g sugar)

L-Lactic Acid NRRL 395

ATCC 52311

GY 18

0.87

0.88

0.81

Ethanol CCUG 28958

CCUG 22420

CCUG 18663 NRRL1501

NRRL2625

NRRL395

0.42

0.44

0.38

0.50

0.50

0.38

Fumaric acid Rhizopus arrhizus 2582

ATCC 20344

R. arrhizus NRRL1526

0.79

0.86

0.82

5.2 Morphology:

Rhizopus species have rhizoids which are root like anchors. These are connected by hyphae

called stolons and they penetrate in to the medium. The length and location of their rhizoids,

the diameter of sporangia, the shape of columellae, and the size, shape and surface texture of

sporangiospores and the maximum growth temperature differentiate Rhizopus genera from

others (Absidia, Mucor, Apophysomyces, Rhizomucor, Saksenaea, and Cunninghamella)

(Zamore et al., 2000). The growth of R. oryzae was as shown in the picture (figure 5.2.1)

when grown on solid media and liquid medium.

Figure 5.2.1: R.oryzae grown on Potato dextrose agar plates for 1 day and 2 days. Right end picture showing

hyphae grown in liquid semi defined medium for 2 days.

Page 31: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

20

Different growth morphologies were observed when Rhizopus oryzae grown in the medium

with varying pH, metal ion concentrations, or change in carbon and nitrogen sources. It was

observed that big clump formation resulted at pH 5 and pellets formation at lower pH. Mg2+

ion concentration also affected the morphology from clump to pellet and mycelium form. It

was also observed that the fungi have different growth morphologies with different types of

sugars in the medium like arabinose favored growth of mycelia fragments, mannose and

glucose favored clump formation, xylose and galactose resulted in fluffy pellets (Punt et al.,

1987). Zhou, Y and coworkers also had similar growth morphologies with varying factors of

pH and trace metal ion concentration during the fermentation of R. oryzae (Meussen et al.,

2012). Filamentous morphology of the strain used in this study was clearly shown in the

picture (Figure 5.2.2) below.

Figure 5.2.2: Microscopic picture of R.oryzae CCUG 28958 under light microscope at 20X, 40X and 100X

magnifications.

The filamentous morphology of many fungal species during fermentation processes implies

many rheological problems in fermenters. In contrast, the filamentous form is the better

morphology for high ethanol production. So if the morphology could be adjusted according to

media components or other parameters, this could give more reliable way in industrial level of

ethanol productions. Such type of approach was developed by Wei Liao and co workers. By

using potato dextrose broth, soy bean peptone and calcium carbonate size at appropriate

concentrations gives pellet morphology of varied sizes. Growth in the medium with no metal

ions resulted in less biomass when compared to medium with metal ions. This showed that

metal ions favor the formation of clumps as spores grow faster and hyphae tangle up

themselves. Avoiding metal ions until regeneration of spores favor pellet formation.

Supplementation of metal ions is very essential for fungal metabolism and avoiding them

completely, may be lethal for product formation (Meussen et al., 2012). Similar type of

studies was conducted by Canan Tari and group on obtaining the pellet morphology in

relation with various media compositions (Canan. T et al., 2011). Even the growth rate and

shear stress in the reactor was shown to be having influence on the morphology of

filamentous fungi. This is due to that the mass transfer, mixing, and heat transfer processes

alter the microbial metabolic processes there by influencing the growth morphology. It was

shown that pH and oxygen tension in the range 12-300 mm Hg does not have any effect on

morphology. It was also noted that hyphal extension rate was proportional to growth rate

(Treskatis et al., 1997).

Page 32: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

21

5.3 Cell wall composition:

Wide ranges of fungal cell walls consist of 80 to 90 % polysaccharides and the remain is of

proteins and lipids. The cell wall is a fabric of interwoven micro fibrils embedded in or

cemented by amorphous matrix substances. Chitin and Cellulose are the mostly found micro

fibrillar or skeletal components of fungi (Garcia, 1968). The main function of cell wall is to

protect the organism mainly from environmental stress and help to survive in harsh

conditions. Also has its importance in cell-environmental interactions like attachment of fungi

to nutrient surfaces. The fugal kingdom comprises of eight different types of cell wall

compositions such as Cellulose, Mannan, Chitin and Polygalactosamine-galactine containing

groups. Chitin-Chitosan combination was only observed in Zygomycetes cell wall, but the

exact role of Chitosan is not yet understood completely. As Chitosan replaces the glucan, it

serves the role of glucans in the cell wall of Zygomycetes fungi (Zamani, 2010). Chitin has a

very huge market and industrially extracted from crab and shrimp shells. After the advances

in the knowledge of various fungal species, much attention was drawn towards the fungal

fermentation as a source for Chitosan production (Wu et al., 2005).

5.4 Protoplast formation:

Protoplast preparation was essential for transforming filamentous fungi irrespective of

transformation method as they have strong layer of cell wall which will be a main obstacle for

transforming DNA to enter the cytoplasm. In general, different enzymes that have the ability

to degrade the cell wall can be employed. As the Rhizopus oryzae cell wall composition was

mainly Chitin, different types of enzymes and their compositions were reported in the

literature for the degradation of cell wall (Michielse et al., 2004, Zhou. X et al., 2008, Linhui.

L et al., 2010, Huan. S. A et al., 2004). There was no specific enzyme that can be used for

particular cell wall type. Various factors such as age of mycelia used, time of exposure of

hyphae to the enzyme mixture, osmotic stabilizer used temperature have their effect on

protoplast formation as well as regeneration efficiency. It was shown that for Ozonium Sp.

optimum enzymatic exposure time was around 3 hours. Prolonged exposure resulted in lysed

protoplast due to the damage of plasma membrane. The enzymatic reactions were faster at

high temperatures and 30°C was observed to be optimal temperature for obtaining highest

concentrations of protoplasts. The regeneration of protoplast was very slow when one and two

days old mycelium was used for protoplast preparation when compared to three days old

mycelium. Complete regeneration was observed after 60 hours. As these factors completely

depend on particular species, they have to be adjusted according to the organism used and

enzymes being used. Use of osmolytic stabilizers help in protecting the protoplasts from

disruption. Inorganic salts, sugars and sugar alcohols could be used as osmolytic stabilizers.

Inorganic salts were shown to be appropriate for filamentous fungi as stabilizers (Zhou. X et

al., 2008, Ohmasa et al., 1999). Various lytic enzymes such as Chitinase, Pectinase,

Lysozyme, Cellulase, Yatalase, were employed individually as well as in combinations. There

are many manufacturers available in the market that has their specific catalogues for

degrading particular type of cell walls like Novozymes (Balasubramanian et al., 2003,

Page 33: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

22

Hébraud and Fèvre, 1988). The microscopic view of protoplasts from R. oryzae was as shown

in the picture (Figure 5.4.1) below.

Figure 5.4.1: Protoplasts of Rhizopus oryzae formed after 3 hour treatment with Yatalase digestion buffer

5.5 Propagation:

The reproduction in fungi is complicated and most of them involve both sexual and asexual

modes of replication. The nomenclature of fungi was also done according to the mode of

replication and it was not clearly elaborated (Hawksworth, 2011).

Similar to other members of Mucorales, Rhizopus Oryzae propagates rapidly through

dispersion of hydrophobic asexual sporangiospores after maturation. Some fungi can form

zygospores through mating between positive and negative strains and propagate sexually

(Figure 5.5.1). Most of the time, R.oryzae is haploid and their germination of zygospores was

long and rate of germination was unpredictable (Broadinstitute, 2012, Taylor et al., 2000).

Page 34: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

23

Figure 5.5.1: Diagrammatic representation of sexual and asexual cycles of propagation in Rhizopus species

(Mathes, 2001).

Rhizopus species can also produce asexual zygospores named to be azygospores which look

similar to sexually formed zygospores (microbiolocigalconcepts). Azygospores are produced

in the absence of a mating partner (Kesington, 2009). Genetic analysis of Rhizopus species

had revealed some regions in the genome responsible for developing mating characters which

regulates mating system. It was reported that the mating experiments produced zygospores

but not progeny. The location of zygospores produced was in the form of straight line

between the two compatible strains (Blakeslee et al., 1927), but the distribution was observed

around the inoculation points in the plates by the Gryganskyi and group. The colour of the

zygospores of various strains of Rhizopus species varied from reddish brown to dark brown

and large central vacuoles were observed inside them. An equal ratio (1:1) in the availability

of each mating type was observed and also sporangiospores sizes were indistinguishable in

the population of R.oryzae and R.delemar. Most of the sporangiospores were of binucleate

and of varying range of sizes (Gryganskyi et al., 2010). The microscopic and electron

microscopic pictures of conjugated hyphae were shown very clearly in the picture (Figure

5.5.2) below.

Page 35: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

24

Figure 5.5.2: Zygospores of Rhizopus oryzae. A cross between CBS346.36 (+) and CBS110.17 (−) was

analyzed with light microscopy (A) and scanning electron microscopy (B). Zygospores (white arrow heads) are

attached to an asymmetric suspensor (black arrow heads). Scale = 20 µm. Asterisks indicate conjugated hyphae

during early stage of zygospore formation (Gryganskyi et al., 2010).

5.6 Plasmids in Fungi:

Plasmids were first discovered in bacteria and later their occurrence was studied in higher

organisms such as yeast, very few in fungi, rarely in plants and also in animals. Discovery of

2µm plasmid in yeast lead to the advancements in molecular and genetic research in

eukaryotes (Broach et al., 1982). Plasmids in yeast were found in nuclei where as fungal

plasmids occur in cytoplasm mostly in mitochondria. Plasmids can be in linear or circular

form. Most of them are linear and they can be determined on gels after the treatment with

proteases. This is because the 5’ ends of the most linear plasmids were protected by a

covalently inked protein. It is difficult to find the circular plasmids as they could not be

determined by normal ethedium bromide staining unless probed with suitable compound.

Extra stronger bands appear in some cases when probes were not used in the restriction

digests of the mitochondrial extracts which represent high copy number of plasmids

(Arganoza et al., 1994). Most of the natural linear plasmids found in fungi have common

features such as terminal inverted repeat specific for each plasmid, 5’ terminus protected by a

protein covalently bound to it, dual non overlapping open reading frames present within the

terminal repeats and finally the presence of intergenic region between ORF’s. The 5’ end

binding proteins was assumed to be coded by a part of DNA polymerase sequence. When

coming to the inheritance of these plasmids, during sexual cycle of propagation, maternal

parent plasmids are transferred to the progeny but not from the paternal parent in most of the

cases observed (May and Taylor, 1989). Various findings show that natural plasmids in

fungal community can be randomly transmitted among them (Arganoza et al., 1994, Yang and

Griffiths, 1993, Griffiths et al., 1990) . There is also chance of losing the plasmids during

sexual cycle and there are several factors that affect the transmission of plasmids (Debets et

al., 1995). The replication of linear plasmids was not clearly understood but it was assumed to

be similar to viral DNA replication where it also contains protected 5’ end and inverted

terminal sequence (Salas, 1988, Sakaguchi, 1990). The replication of circular plasmids was

thought to be similar as rolling circle replication of circular elements in prokaryotes. But

analysis of ORF of Mauriceville plasmids revealed the presence of reverse transcriptase

coding region that revealed ―Reverse transcriptase‖ based replication (Griffiths, 1995).

Neurospora species was studied extensively for knowing the integration mechanisms of

plasmids in to mitochondrial DNA (mtDNA) in fungi. Senescence plasmids, kalilo (Bertrand

et al., 1985) and maranhar (Court et al., 1991) from N. intermedia and N. crassa respectively

were used for this studies and it was found that their integration process in to mtDNA was

completely different form generally observed plasmids integration mechanisms found in

prokaryotic and eukaryotic systems. These plasmids contain flanks of long inverted repeats of

mtDNA where the plasmid integrates. Replication intermediate of kalilo plasmid might

crossover with mtDNA resulting in mtDNA getting flanked to plasmid. This was different in

the case of maranhar plasmid where complete cross over occurs (Griffiths, 1995).

Page 36: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

25

5.7 Transformation:

Different methods have been developed for transforming fungi. The efficiency of

transformation of filamentous fungi depends on the type of method being used. First

transformation of yeast Saccharomyces cerevisiae was done by making protoplast mediated

transformation and it was extended to many filamentous fungi. But the frequency was much

lower when compared to S.cerevisiae. In order to improve transformation of filamentous

fungi, progress has been made over the last decade that has resulted in the establishment of

alternative methods for fungal transformation such as electroporation, biolistic transformation

and agro bacterium mediated transformation (Meyer, 2008). The method that could be used

depends on the fate of the DNA after transformation (Ruiz-Díez, 2002). That is, when the

transformation is for targeted integration or deletion of particular gene, then Agro bacterium

Mediated Transformation (AMT) would be preferable method (Michielse et al., 2005). When

multiple copies of a particular gene has to be integrated at several loci, then Protoplast

Mediated Transformation (PMT) is the best way (Fincham, 1989, Meyer et al., 2003).

Different experiments resulted in a common conclusion that recombinant mechanisms have

preferential affinity toward single stranded DNA. The table below (Table 5.7.1) shows the

main principle, advantages and disadvantages of commonly used transformation methods in

fungi.

Table 5.7.1: A brief summary of advantages and disadvantages of different approaches of

transformation and their respective applications. PMT: Protoplast-mediated transformation;

AMT: Agrobacterium-mediated transformation; EP: Electroporation; BT: Biolistic transformation;

TR: transformation rate; TE: transformation efficiency (Fincham, 1989, Ruiz-Díez, 2002, Michielse et

al., 2005).

Method Principle Advantage Disadvantage

PMT Preparation of protoplast using cell wall

degrading enzymes. Uptake of DNA is

achieved by PEG and CaCl2.

Different cell types can be

used (spores, Hypal tissue,

germlings)

Particular batch of

enzymes alter TE.

AMT DNA transfer is achieved during co-

cultivation of A. tumefaciens with the

fungus.

Copy number of DNA

insertions is low. Improves

targeted integration.

Various parameters

during co-

cultivation affect

TR. More time-

consuming than the

other methods.

EP Reversible membrane permeabilisation

induced by local application of electric

pulses mediated DNA uptake.

Different cell types can be

used. Cheap and simple

method.

Often requires

protoplast

formation to render

cells competent.

BT Particles (tungsten, gold) are coated with

DNA and become accelerated at high

velocity into cells.

Recipient cells can retain

their cell walls (no pre-

treatment necessary)

Requires special

equipment.

Page 37: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

26

These transformation procedures can be applied according to the requirement. As mentioned

in the above table, each method has its own advantages and disadvantages.

5.7.1 Protoplast Mediated Transformation using PEG/CaCl2:

Due to simplicity in operation and technical convenience, PEG/CaCl2 method was the mostly

followed procedure for transforming filamentous fungi since last 30 years (Hinnen et al.,

1978, Case et al., 1979) in spite of many disadvantages when compared to other methods. The

main disadvantage was, difficult in obtaining high concentrations of viable protoplasts, lower

transformation efficiency and unstable transformants. High molecular weight Polyethylene

Glycol (PEG) that is being used in this method helps in formation of clumps and fusion of

protoplasts. It was assumed that Polyethylene Glycol (PEG) allows the DNA to stick to the

protoplasts there by facilitates its uptake (Liu and Friesen, 2012). But recent study shows that

PEG does not cause DNA to stick to cell surface. Osmotic stabilizer and pH adjusting buffer

that were being used in this method helps in maintaining protoplast viability, whereas the role

of PEG and Ca2+

was unclear (Kuwano et al., 2008).

6. Methods and Materials

Rhizopus oryzae is a filamentous fungi and it grows very easily on various media both in

submerged form or on solid surface. In the fermentation process with this organism,

submerged fermentation is implemented. Due to its filamentous nature, the hyphae easily

tangle around the surfaces available for it inside the fermenter. This leads to great agitation

problem in large scale.

6.1 Genomic DNA extraction from R. oryzae: (Hoskins. L, 1998)

Materials required:

1-2 days old culture of R.oryzae grown in liquid semi defined glucose medium

(Appendix),

Acid washed glass beads (400-500 microns),

Sterile 10 ml microfuge tubes, Vortex machine, Heavy Phase Lock GelTM

tubes (1.5

ml), sterile eppendorf tubes (1.5 ml),

Phenol: Chloroform: Isoamyl alcohol (24:24:1), 95% Ethanol, 70% Ethanol, TE buffer

(10mM Tris HCL, 0.1 mM EDTA pH 8.0),

Distilled water, 5M NaCL,

Lysis buffer (0.1M Tris HCL pH8.0, 50 mM EDTA, 1% SDS, all the components

dissolved in distilled water).

Page 38: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

27

Procedure:

About 1 gram of grown Rhizopus oryzae was taken in to a 10ml microfuge tube using

sterile scalpel and centrifuged at high speed for 2 minutes to remove medium. The

mycelium was washed with distilled water later.

The washed mycelium was resuspended in 1 ml of lysis buffer (see that the mycelium

is completely suspended) and vortexed briefly.

The suspension was added with 300 µg of acid washed glass beads and vortexed again

to break the cells through mechanical shearing and centrifuged for 2 minutes to reduce

the foam.

The lysed cells (supernatant) were transferred to a pre spinned Heavy Phase Lock

GelTM

tubes.

Equal amount of Phenol: Chloroform: Isoamyl alcohol (24:24:1) mixture (around 500

µl) was added and mixed gently by inverting.

The tubes were centrifuged for 5 minutes at 13500 rpm and the upper phase was

transferred to a new pre spinned Heavy Phase Lock GelTM

tube.

The upper phase was again added with 500 µl of Phenol: Chloroform: Isoamyl alcohol

(24:24:1) mixture and vortexed to get uniform solution.

The mixture was centrifuged for 5 minutes and upper phase was transferred to a sterile

eppendorf tubes (1.5 ml).

1 ml of 95% ethanol was added to the tube and spinned for 6 minutes at high speed to

precipitate the DNA. Ethanol was discarded carefully without disturbing the pellet.

Again the pellet was washed with 70% ethanol by vortexing and finally centrifuged to

discard ethanol. The pellet was dried on bench for about 10 minutes and re suspended

in 50µl sterile TE buffer.

6.2 Extraction of Plasmids from paper discs:

The plasmid P silent 1 (Nakayashiki et al., 2005) was obtained from the FGSC (Kansas City,

Missouri USA) (Center). The plasmid was sent on paper discs and they were stored at 4°C in

fridge.

A paper disc was taken in to a sterile eppendorf tube and added with 100 µl of TE buffer to

dissolve the plasmid DNA present on the paper disc. The disc was crushed thorougly using a

sterile plastic tip for few seconds and the tube was centrifuged. The supernatant was used for

transforming the competent E.coli cells as described in the next section.

The DNA concentration obtained was very less and the amount of DNA for transformation

was adjusted according to the requirement for getting considerable number of transformants.

Page 39: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

28

6.3 Transformation of Competent E.coli cells using CaCl2 (Sambrook and Russell, 2001

modified)

Material required:

DH5α overnight culture,

LB medium (preparation 1), LB agar plates with ampicillin (60 µg/ml) (Appendix,

preparation 2)

0.1 M ice cold CaCl2, ice

Transforming plasmid DNA,

Procedure:

5 ml overnight grown DH5α was diluted with 95 ml of fresh LB medium in a 250 ml

flask and grown on shaking incubator at 37°C until the OD600 reaches 0.2-0.4

The cells were collected by centrifuging the broth at 4300 rpm at 4°C for 5 minutes.

Supernatant was discarded the cells were resuspended in 12.5 ml of 0.1 M ice cold

CaCl2.

The cells were maintained on ice for 30 minutes and they were collected by spinning

at 4°C.

Again the cells were re suspended in 2.5 ml 0.1 M ice cold CaCl2.

100 µl of the competent cells were mixed with 0.5 µg DNA and left on ice for 30

minutes.

The cells were subjected to heat shock for 2 minutes at 42°C using preheated water

bath.

The cells were added with 900 µl of SOC medium and left undisturbed for gene

expression to take place for 30 minutes.

Finally the transformed cells were inoculated on to LB agar plate with ampicillin

(60µg/ml) and incubated at 37°C for overnight to get the colonies of transformants.

6.4 Vector construction:

The vector should express hpRNA construct containing LDHA gene sequence in the

organism.

6.4.1 Primer designing:

For solving the purpose described above, two sets of primers were designed where both of

them amplify the same part of LDHA gene but varying at flanking ends with different

restriction sites. These flanks serve the purpose of inserting the PCR product in to the p

Silent1 vector in right orientation.

Page 40: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

29

6.4.2 Gene sequence to be amplified:

From the previous studies on silencing LDHA gene in R. oryzae through siRNA delivery

(Gheinani et al., 2011), it was shown that a set of 25bp size siRNA that was homologous to a

portion of LDHA gene (shown in yellow) triggered silencing. Depending on their finding,

portion of LDHA gene was chosen to be amplified.

LDHA gene sequence of R. oryzae CCUG 28958 was taken and used for designing primers

that can amplify the region containing (198bp -222bp) represented in yellow.

1 atggtattac actcaaaggt cgccatcgtt ggagctggtg cagtaggagc ctccactgct

61 tatgcactta tgtttaaaaa catttgtaca gaaatcatta ttgtggatgt taatcctgac

121 atcgttcaag ctcaagtcct tgaccttgca gatgctgcca gtataagtca cacgcccatc

181 cgagcaggta gcgcagagga ggcagggcag gcagatattg ttgtcatcac ggccggtgcg

241 aaacaaaggg aaggtgagcc tcggacaaag ctcattgaac gaaacttcag agtgttgcaa

301 agtatcattg gtggcatgca acccattcga ccagacgcag tcatcttggt ggtagcaaat

361 ccagtcgata tcttgacaca cattgcaaag accctctctg gactgcctcc aaaccaggtc

421 attggctccg gtacctacct tgacacgacc cgtcttcgcg tccatcttgg cgatgtcttt

481 gatgtcaatc ctcaatcggt ccatgctttt gtcttgggtg aacatgggga ttcccagatg

541 atcgcttggg aggctgcttc gattggtggg cagccgttga caagtttccc ggaattcgca

601 aagctggata aaacagcaat ttcaaaagcg atatcaggta aagcgatgga gatcattcgt

661 ttgaaaggag ccacgtttta tggaattggt gcctgtgcag cggatttagt gcacactatc

721 atgttgaata ggaaatcagt acatccagtt tctgtttatg ttgaaaagta tggagccact

781 ttttctatgc ctgctaaact tggatggaga ggtgttgaac agatctatga agtaccactg

841 acggaagaag aagaagcgct gcttgtaaaa tctgtagaag cactgaaatc agttgaatat

901 tcatctacaa aagtcccaga aaaaaaagtt catgctactt ccttttctaa aagtagctgt

961 tga

The size of the PCR product should be in the range of 300 to 600 bp for better silencing effect

(Zhong et al., 2012).

PerlPrimer v1.1.21 (Marshall, 2011) was used for primer designing and the two primer sets

used were shown below (Table 6.4.2.1) along with their restriction sites at their 5’ ends. The

amplified product was around 400 bp and the region was indicated in green in the above

sequence.

Table 6.4.2.1: FP = Forward Primer, RP = Reverse Primer, 1=set1, 2 = Set2.

Label Sequence Restriction site present

FP1 5'-ATAA[CTCGAG]GGTATTACACTCAAAGG-3' xhoI

RP1 5’-AATT[AAGCTT]GACCTGGTTTGGAG-3’ hindIII

FP2 5'-AT[GGTACC]GGTATTACACTCAAAGG-3’ kpnI

RP2 5’-AATGT[AGATCT]GACCTGGTTTGGAG-3’ BglII

Page 41: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

30

6.5 PCR:

6.5.1 Reaction mixture concentrations used (Fisher, 2010):

Table 6.5.1.1: PCR reaction mixture composition

10 X Dream TaqTM

Green Buffer 5 µl

dNTP Mix, 2mM each 5 µl (final concentration 0.2mM of each)

Forward primer, 40 µM 1 µl (final concentration 0.8 µM)

Reverse primer, 40 µM 1 µl (final concentration 0.8 µM)

Template DNA 2 µl (10 pg -1 µg)

Dream TaqTM

Polymerase (1.25 u/ µl) 1 µl

DNase free water 35 µl

Total volume 50 µl

6.5.2 Thermal cycling parameters:

Table: 6.5.2.1: PCR conditions

Step Temperature Time

1 95°C (to make ssDNA) 2 minutes

2 95°C 30 seconds

3 Annealing temperature(<Tm)

47°C for set1, 48°C for set2, 43°C for set3 and 45°C for set4

45 seconds

4 72°C 1 minute

5 72°C (final elongation to fill in all nicks) 10 minutes

6 4°C (storage) 1-2 hours

Step 4 to 2 repeated for 35 times

Page 42: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

31

6.6 Agarose gel electrophoresis:(Lewis, 2001)

Materials required:

Agarose, 10X TBE buffer (Appencix, preparation 11), Ethedium bromide (EtBr),

Electrophoresis equipment, UV lamp

Procedure:

1 gram of agarose was mixed with 100 ml 1XTBE in a conical flask.

The agarose solution was heated in a microwave oven and added with 5 µl EtBr stock

solution (10mg/ml readymade). As EtBr was carcinogenic, it was handled carefully

using gloves and eye glasses.

The mixture was poured into a casting tray equipped with an appropriate comb and

left undisturbed until solidified.

The gel chamber was carefully turned such that the comb was near the negative

electrode so that DNA moves toward the positive electrode.

The chamber was filled with 1X TBE buffer after removing the comb (see note) until

it covers the gel.

The DNA was loaded in to the wells according to the well size and the electrodes were

connected to voltmeter (110V) for required duration.

After completion of the electrophoresis, the gel was looked under UV lamp and

photographed. The buffer was collected in to a separate container which was filtered

later using charcoal and filter paper.

Note: Care should be taken not to disturb the wells while taking out the comb. The gel may

deform if sufficient time was not provided.

6.7 Purification of PCR product:

Purification of PCR product was done to get rid of DNA polymerase present in the reaction

mixture. This was done using Wizard® SV Gel and PCR clean up System (bioprotocols.info)

and concentrated by distilled water and stored at -20°C for further use.

6.8 Plasmid DNA Extraction from E.coli using Alkaline Lysis Method (Winstanley and

Rapley, 2000)

Materials required:

LB- broth (Appendix, preparation 1)

Lysis solution : 200mM NaOH, 1%SDS, (should be prepared on the same day)

Resuspension solution: 50 mM Glucose, 50mM Tris-Hcl, pH8.0, 10mM EDTA, store

at 4°C

Page 43: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

32

Potassium acetate: 3 M potassium /5M acetate: for 100ml, 29.4g of potassium acetate

was dissolved in 88.5ml of distilled water and 11.5 ml of glacial acetic acid. (should

not be autoclaved)

70 % Ethanol

Isopropanol

Procedure:

3 ml of LB-broth was inoculated with E.coli carrying vector and incubated at 37°C

overnight with shaking.

The culture was transferred into two Eppendorf tubes and centrifuged at high speed

for 30 s in microfuge and the supernatant was discarded carefully without disturbing

the pellet using pipette

100 µl of re suspension solution was added to the tube and the pellet was resuspended

by shaking or vortexing.

200 µl of lysis solution was added and the tubes were inverted several times for

mixing. The tubes were left undisturbed for 2 minutes for the lysis to take place.

150 µl of neutralizing solution (potassium acetate) was added and mixed well by

inverting which gives a white precipitate (chromosomal DNA).

The tubes were centrifuged for 5 minutes in a microfuge at 13,500 rpm.

Two new sterile tubes were filled with 250µl isopropanol and labeled.

The tubes were carefully removed from the microfuge without disturbing the

precipitate and the supernatant was carefully transferred to isopropanol containing

tubes using 1 ml pipette.

The tubes were vortexed for 10 seconds and centrifuged in the microfuge for 30s at

high speed which precipitate the plasmid DNA as white pellet.

The supernatant was discarded and pellet was washed by adding 750 µl of 70%

ethanol and centrifuged at high speed for 30s.

The ethanol was discarded and centrifuged for 10 seconds to collect the remaining

ethanol at the bottom of the tubes. the remaining ethanol was carefully aspirated and

the tubes were allowed to air dry for 5min on the bench.

50 µl of TE buffer was added into the tubes to re suspend the pellets and left

undisturbed for few minutes and checked for concentration using Spectrophotometer

(Nano drop).

After the extraction of Plasmid Psilent1 from E.coli using the above procedure, PCR product

that was amplified using first set if primers was ligated in to the plasmid using restriction

digestion and ligation protocols described below.

Page 44: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

33

6.9 Restriction digestion and Ligation:

6.9.1 Double digestion of PCR product and Plasmid:

Double digestion of purified PCR product and plasmid were done in separate PCR tubes. This

enables them to acquire sticky ends for ligation. In the first cycle of construction, one LDHA

gene was inserted by double digestion with XhoI (Fisher, 2011a) and HindIII (Fischer, 2011).

The reaction mixture for double digestion was followed as given table below.

Table 6.9.1.1: Reaction concentrations

DNA (Plasmid or PCR product) 5µg

Enzyme 5µl

10X fast digest Buffer 5µl

Total volume (make up with distilled H2O) 50µl

The order of adding the contents was water, Digestion buffer, DNA and finally enzyme. The

reaction was carried out at 37°C in a thermal cycler for one hour. The enzymes were

inactivated by incubating at 80°C for 10 minutes.

6.9.2 Inactivation of Restriction enzymes that are not heat sensitive:(uregina)

Some of the restriction enzymes used in this study where not heat sensitive and so phenol

chloroform extraction was performed to separate them from the mixture.

Method:

Double the volume of Phenol: chloroform: Isoamyl alcohol (24: 24: 1) solution was

added to the digested mixture in a sterile eppendorf tube and vortexed briefly.

The solution was spinned at 10000 rpm for 5 minutes and the supernatant was

transferred to new eppendorf tube.

Double the volume of 100% isopropanol was added for precipitating the DNA and

spinned for 5 minutes at 10000 rpm.

The pellet was washed using 70% ethanol twice and finally dried for few minutes to

evaporate traces of ethanol and dissolved using sterile TE buffer (amount adjusted to

required concentration of DNA).

Page 45: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

34

6.9.3 Ligation:

Restricted products (Plasmid and PCR product) in separate tubes were ligated using T4 DNA

ligase enzyme using the following reaction conditions (Fisher, 2011b).

Table 6.9.3.1: Reaction concentrations

Linear plasmid 20-100 ng

PCR product 1:1 to 5:1 molar ratio over plasmid

10X T4 DNA ligase buffer 2 µl

T4 DNA ligase 1 unit

Final volume (make up with distilled H2O) 20 µl

The reaction was carried out at 22°C for 20 minutes in thermal cycler and the product was

stored at -20°C for transformation in to E.coli.

The ligated product was used for transforming the E.coli using the same protocol that was

mentioned previously.

Plasmid amplification was done by inoculating the transformants in the LB medium

containing 100 µg/ml ampicillin for overnight in a shaking incubator at 37°C.

The vector was extracted from the bacteria in the same way described previously and tested if

the LDHA gene was properly inserted by running in 1.7% Agarose gel electrophoresis. After

the conformation, insertion of second LDHA gene was done.

The second LDHA gene was amplified using second set of primers and inserted in to the

vector in the same way as inserting first LDHA gene and transformed in to competent E.coli

cells using CaCl2 method for transformation. The same procedure was repeated for extraction

of plasmid and the vector was tested for proper insertion of the second LDHA gene on

agarose gel.

Now the completely constructed vector was used for transforming the protoplast prepared

from wild Rhizopus oryzae as described below.

6.10 Protoplast Preparation of Rhizopus oryzae by Using Yatalase (Jahromi and

Gheinani, 2011 modified)

Materials Required:

Semi synthetic medium (Appendix, preparation 6)

Potato dextrose agar plate (Appendix, preparation 7)

Digestion buffer prepared by Yatalase (Appendix, preparation 8)

Page 46: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

35

Distilled water

Sterile metallic sieve and scalpels

Shaking incubator

Procedure:

Inoculation:

150 ml of semi-synthetic medium was transferred to a 500ml Erlenmeyer flask.

20 ml of water was added to a cultured potato dextrose agar plate (PDA) and 3ml of it

was added to the semi synthetic medium.

It was incubated at 30°C and 140rpm, for 3-4 days. (see note)

Protoplast preparation:

The mycelium was taken in to a sterilized sieve and around 1 gram of mycelium was

transferred to a 50 ml sterile plastic tube near flame using scalpel.

Yatalase digestion buffer was added to the tube with mycelium (10ml/g mycelium)

The tube was kept on a shaking incubator for 3hours at 30°C and 90rpm. Protoplast

formation was checked in between in a light microscope.

The digested mycelium was filtered through single layer of gauze cloth in to a new

tube near the flame and centrifuged in a swing bucket rotor at 4°C and 3500 rpm for 5

minutes.

The pellet was separated from supernatant with little solution left.

The pellet was vortexed and immediately used for transformation or can be stored at -

20°C for further use.

Note: The incubation time was very crucial which determines the frequency of protoplast

regeneration. Use of young cultures (1-2 days) for protoplast preparation may give less

frequency of protoplast regeneration which is not desirable.

6.11 Transformation of Rhizopus oryzae by the Calcium Chloride/Polyethylene

Glycol (CaCl2/PEG) method (Jahromi and Gheinani, 2011 modified)

Materials required:

Ice, PEG solution (from preparation 10), STC buffer (Appendix, preparation 9), PDA plates

(from preparation 7 *see note), linearized plasmid vector DNA by SpeI, Protoplasts

Procedure:

100 µl of protoplast solution of Rhizopus oryzae was transferred to 15ml sterile plastic

tube along with 100 µl plasmid DNA (conc. from 1 - 2 µg/ml) (silencing vector) and

mixed gently using pipette.

Page 47: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

36

The tube was incubated on ice for 15 minutes.

PEG solution was added to the tube in incremental order of 200 µl, 400 µl and 600 µl

and mixed gently by pipetting up and down each time.

The tube was left on ice for 10 minutes and then centrifuged for 3 minutes at 3700

rpm (or 2000 x g) at 4°C.

The supernatant was discarded and 1ml of STC buffer was added to re suspend the

protoplasts.

The whole protoplast solution was used for spreading on the prepared potato dextrose

agar plate incubated at 30°C.

The growth of transformed Rhizopus oryzae is visible only after 24 hours of

incubation.

*Note: Care should be taken to have the level of agar medium in the plates to be half of the

maximum volume.

6.12 Sub culturing of transformed R. oryzae

To test if the if a permanent change in the genome has been induced to R. oryzae the

transformed R. oryzae was sub cultured by letting them grow fully in 3 days and form spores

in about 6 days..

Materials required:

Distilled water, sterile plastic spreader, pipette, PDA plates (Appendix, preparation7), sterile

plastic tube, and cultured plates of transformed R.oryzae.

Procedure:

20 ml distilled water was used to dilute the formed spores in the culture plates and

they are slowly streaked using sterile plastic tube to suspend the spores.

The spore suspension was transferred carefully in to sterile plastic tube near the flame

and mixed well to get uniform solution.

100 µl of this spore suspension was inoculated on to new PDA plates and incubated

for 3 days at 30°C.nomic DNA was extracted from transformed Rhizopus oryzae

cultures as well as subculture and submitted for PCR analysis to find out if the vector

was present in the second generation compared with the first.

6.13 Primer design for amplifying LDHA gene along with cutinase intron:

Cutinase intron sequence was taken from P silent 1 vector and primers were designed that can

amplify LDHA gene along with cutinase intron. Two possible orientations for vector having

two copies of LDHA gene was LDHA- CUT and CUT-LDHA. Cutinase intron gene sequence

was as shown below:

Page 48: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

37

gtgagccttt cttcttgcct ctctttgttt tttttttgtt ctttttgccg aatagtgtac ccactggaga tttgttggcc

atgcaaataa atggaaggga ctgacaagat tgtgaaattg ttcaaaacac acag

Two primer sets designed for amplifying part of LDHA gene along with cutinase intron were

mentioned in table below:

Table 6.13.1: FP = Forward Primer, RP = Reverse Primer, 3=set3, 4 = Set4.

Label Sequence

FP3 5' TATTACACTCAAAGGTCGCCA 3'

RP3 5'TCACAATCTTGTCAGTCCCT 3'

FP4 5'AGCCTTTCTTCTTGCCTCTC 3'

RP4 5'GAGAGGGTCTTTGCAATGTG 3'

Both these primers can amplify DNA of length around 530 bp.

Using the set of primers in table 6.13.1the genomic DNA from transformed cultures and sub

culture were amplified with PCR and the presence of the integrated vector was investigated

by DNA electrophoresis.

Page 49: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

38

7. Results and Discussion

7.1 LDHA gene amplification through PCR:

PCR amplification of selected portion of LDHA gene was performed using two sets of

primers having varying flanks at their respective 5’ ends. One set of primers have the flanks

containing restriction sites for KpnI and BglII. Second set of primers contain flanks with

recognition sites for XhoI and HindIII endonucleases. These flanks help in the insertion of

LDHA gene in to open Psilent-1 plasmid in a fashion that it can transcribe an hpRNA

structure after transforming in to host organism.

Figure 7.1.1: 1% Agarose gel electrophoresis showing bands of PCR product of R.Oryzae

genome with LDHA primers.

Page 50: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

39

From the gel picture shown above (figure 7.1.1), it can be confirmed that the region amplified

was of correct length (420bp) as expected.

7.2 Psilent 1 with one LDHA gene inserted in it:

The two sets of LDHA gene were inserted in to empty vector in a sequential order. First set of

LDHA gene was inserted by opening the plasmid using restriction enzymes XhoI and HindIII.

The size of the empty vector was around 6.9 kb and after inserting one copy of LDHA gene, it

was around 7.32 kb. This is shown in the picture below (figure 7.2.1). The difference between

the empty vector and vector with one copy of LDHA gene is small (420 bp). So there is only a

little difference observed in the gel picture.

Figure 7.2.1: 1.7% agarose gel electrophoresis showing bands of linear Psilent1 with LDHA

(left), Psilent 1 (middle) and 1kb+ ladder (right). Psilent 1 was 6.9 kb in length and LDHA

gene was 0.42 kp in length.

During the insertion of LDHA gene in to the empty vector, the plasmid was cut open using

double digestion. This reaction cuts the plasmid twice which will not help in self ligation of

the vector. All the transformants with ampicillin resistance were considered to be having the

plasmid with LDHA gene inserted in it. But the average transformed colonies obtained per µg

of DNA were very less. Each time of transformation, around 2 µg of DNA was used.

Page 51: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

40

After double digestion of the plasmid and the PCR product using different restriction

enzymes, XhoI and HindIII were deactivated by heating at 80°C for 10 minutes.

But when coming to the inactivation of second set of restriction enzymes, KpnI and BglII,

they are not sensitive to heat. So they were separated using common Phenol chloroform

extraction. Huge amount of DNA was lost during this step and to get the considerable amount

of DNA after purification, large amount of DNA has to be used. During this experiment,

restriction enzymes from different manufacturers were employed for double digestion and

there was no compatibility issues in the buffers used. All the enzymes worked well in Fast

digest buffer from Fermentas®.

Higher concentrations of plasmid were obtained by transforming it in to E.coli, amplifying it

and extracting plasmid DNA.

7.3 Complete vector after inserting second LDHA gene in to the vector:

After inserting the second copy of LDHA gene, the difference between the empty vector and

vector with two copies of LDHA gene was around 840 bp. This is clearly shown in the picture

below (figure 7.3.1). The vector was cut open using HindIII and loaded in to the gel for

analysis all the time.

Figure 7.3.1: 1.7% agarose gel electrophoresis picture showing bands of completely

constructed vector (right), vector with one copy of LDHA gene (in lane 2), empty vector (left)

and 1kb+ DNA ladder.

The final vector after inserting two copies of LDHA gene was around 7.74 Kb in length.

Page 52: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

41

7.4 Protoplast formation from R. oryzae:

The efficiency of protoplast formation depends on various factors such as the age of the

culture used, enzyme used for breaking cell wall and incubation time. The regeneration

capability was also affected by varying above mentioned factors. Lysozyme and Yatalase

were used for generating the protoplast. Among these enzymes, Lysozyme did not give better

yield when compared to Yatalase. The regeneration of protoplast was better and protoplast

formation took long time if the three days old culture was used. If two days old culture was

used, regeneration efficiency decreased and protoplast formation was faster when compared

to the previous case. This might be due to well developed cell wall formation in the organism

with age. The average regeneration time was around 2 days and spore formation was after 4

days.

Figure 7.6.1: Microscopic pictures showing the formation of protoplast from hyphae of

R.oryzae.

The number of protoplasts formed increases with time of incubation in Yatalase lysis solution.

The protoplasts were observed under the microscope after every hour.

The protoplasts prepared were subjected to transformation by PEG/CaCl2 method using the

new vector DNA cut opened by SpeI (linearized). Linearization of vector prior to

transformation is very important as that enhances the chance of getting the transformed vector

to integrate in to host genome through homologous recombination. This was clearly

demonstrated by previous researchers in various fungal species with regards to integration of

linear plasmids in to chromosomal DNA (Griffiths, 1995).

7.5 Hygromycin resistance for Rhizopus oryzae:

To find out if the strain used in this study has antibiotic resistance, spores of wild type were

inoculated in to semi defined media along with varying concentrations of Hygromycin. To

know the maximum level of tolerance, the range was set from 200µg/ml to 1000µg/ml.

Page 53: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

42

Figure 7.5.1: Flask fermenters showing growth of R.Oryzae wild type at various

concentrations (200 to 1000 µg/ml) of hygromycin.

It was seen that wild type Rhizopus oryzae had hygromycin resistance and it can tolerate the

concentrations as high as 1000 µg /ml which was common among fungal species. The growth

pattern was almost similar and there is no decrease in hyphal mass with increase in antibiotic

concentration. This demonstrates that Hygromycin does not have any lethal effect on the

mycelium.

Because of this a great problem came up when selecting the transformants having the vector

from wild type. Hygromycin couldnot be used as a selection marker in this organism

transformed with Psilent 1 vector.

7.6 Confirmation of transformed R.oryzae and demonstration of the stability in the

progeny:

As hygromycin resistance gene in the vector could not be used as a marker for knowing the

transformants the problem had to be solved in another way. DNA was extracted from wild

type and transformed R. oryzae, both first and second generation. PCR was performed using

the primers (see 6.13) that can amplify Cutinase gene along with LDHA sequence. The

different primers with varying annealing temperatures were tested to have the specific binding

to amplify the target sequence. The primers that were mentioned in the section 6.12 gave

bands that can be helpful to show the variation between transformants and wild type. The

picture below (figure 7.7.1) shows the bands of PCR products of genomic DNA from

transformants first generation, tranformants second generation, and wild type.

Page 54: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

43

Figure 7.7.1: Gel picture showing PCR products of DNA extracted from wild type (-ve control),

transformed, subculture of R.oryzae and P silent 1 (+ve control).

The expected length of the amplicon is 530 bp with the designed primers. From picture 7.7.1,

it can be confirmed that, transformed R.oryzae first generation and second generation of

transformed R.oryzae has the vector present in them. This clearly shows that the vector was

transferred to its progeny, and that a stable transformant of R. oryzae with an ingral vector for

hpRNA expression.

8. Conclusion

Many vectors are in use for gene silencing in plants and animal cell lines till date but,

considerably less when coming to fungal species. The present work shows the vector

construction (figure 8.1) and transformation of a Rhizopus oryzae species. It also shows that

this vector is stable in the progeny and thereby creates a permanent change to the genome of

the host cell mostly through homologous recombination. Finally, it is shown that there is a

Hygromycin resistance in the R. oryzae strain CCUG 28959 that in the literature never has

been reported before. In the future the constructed vector (figure 8.1) can be used as a model

for silencing various genes of interest in this species and thereby achieving a stable silencing

effect.

Page 55: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

44

Figure 8.1: Diagrammatic representation of vector construction, transformation and final synthesis of

hpRNA.

8.1 Draw backs of present work:

This vector cannot be selected directly in this strain as it processes Hygromycin resistance.

Hygromycin resistance was the only marker present in this vector for selection in fungi. The

only way of knowing the transformation is PCR analysis of genomic DNA using specific

primers that can amplify particular region in the transformed plasmid. This is very laborious

process when compared to direct selection.

9. Further research

9.1 Optimization and analysis of Fermentation with transformed Rhizopus oryzae:

The next step of interest would be to do fermentation using the transformed Rhizopus oryzae

with constructed vector and to optimize the process to increase ethanol production on the

behalf of lactate silencing in a R.oryzae strain with high lactate production.

9.2 Changing the inserted LDHA gene sequence in the vector:

The LDHA gene sequence that was used in the present study was 430 bp, only a part of the

total gene (ca 900 bp) and it was chosen according to the siRNA used in the previous

Page 56: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

45

research. Other part of the gene can be tried for constructing the vector followed by tests of

the silencing efficiency.

9.3 Improving the silencing vector by inserting eGFP gene:

This vector can be further improved by inserting eGFP gene which will have the property of

giving fluorescence in the transformers and thereby will help in differentiating the

transformants from wild type. This will solve the purpose of direct selection instead of PCR

analysis as done in the present work and thereby saves time in analyzing the transformants for

the presence of transformed vector.

Page 57: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

46

10. References

ABEDINIFAR, S., KARIMI, K., KHANAHMADI, M. & TAHERZADEH, M. J. 2009.

Ethanol production by Mucor indicus and Rhizopus oryzae from rice straw by separate

hydrolysis and fermentation. Biomass and Bioenergy, 33, 828-833.

ADACHI, E., TORIGOE, M., SUGIYAMA, M., NIKAWA, J.-I. & SHIMIZU, K. 1998.

Modification of metabolic pathways of Saccharomyces cerevisiae by the expression of

lactate dehydrogenase and deletion of pyruvate decarboxylase genes for the lactic acid

fermentation at low pH value. Journal of Fermentation and Bioengineering, 86, 284-

289.

AKASHI, H., MATSUMOTO, S. & TAIRA, K. 2005. Gene discovery by ribozyme and

siRNA libraries. Nature Reviews Molecular Cell Biology, 6, 413-422.

AMADIOHA, A. C. 1993. Production of Cellulolytic Enzymes by Rhizopus oryzae in Culture

and Rhizopus-Infected Tissues of Potato Tubers. Mycologia, 85, 574.

AMORE, A. & FARACO, V. 2012. Potential of fungi as category I Consolidated

BioProcessing organisms for cellulosic ethanol production. Renewable and

Sustainable Energy Reviews, 16, 3286-3301.

ARCHER, D. B. & WOOD, D. A. 1994. Fungal Exoenzymes The Growing Fungus. In:

GOW, N. & GADD, G. (eds.) The Growing Fungus. Springer Netherlands.

ARGANOZA, M. T., MIN, J., HU, Z. & AKINS, R. A. 1994. Distribution of seven homology

groups of mitochondrial plasmids in Neurospora: evidence for widespread mobility

between species in nature. Current Genetics, 26, 62-73.

BALASUBRAMANIAN, N., JULIET, A., SRIKALAIVANI, P. & LALITHAKUMARI, D.

2003. Release and regeneration of protoplasts from the fungus Trichothecium roseum.

Canadian Journal of Microbiology, 49, 263-268.

BARNES, S., ALCOCER, M. & ARCHER, D. 2008. siRNA as a molecular tool for use in

&lt;i&gt;Aspergillus niger&lt;/i&gt. Biotechnology Letters, 30, 885-890.

BAUTISTA, L. F., ALEKSENKO, A., HENTZER, M., SANTERRE-HENRIKSEN, A. &

NIELSEN, J. 2000. Antisense silencing of the creA gene in Aspergillus nidulans.

Applied and environmental microbiology, 66, 4579-4581.

BERTRAND, H., CHAN, B. S. & GRIFFITHS, A. J. 1985. Insertion of a foreign nucleotide

sequence into mitochondrial DNA causes senescence in Neurospora intermedia. Cell,

41, 877-884.

BIOPROTOCOLS.INFO. LB Medium and Agar Plate Recipe [Online]. Available:

http://www.bioprotocols.info/model_organisms/bacteria/lb_medium_and_plates.php

[Accessed 07-17 2012].

BIOPROTOCOLS.INFO. TBE Buffer Recipe (Tris-Borate-EDTA) [Online]. Available:

http://www.bioprotocols.info/reagent_and_buffer_recipes/10X-TBE.php [Accessed

07-26 2012].

BLAKESLEE, A. F., CARTEDGE, J. L., WELCH, D. S. & BERGNER, A. D. 1927. Sexual

dimorphism in Mucorales I. Intraspecific Reaction. Botanical Gazette, 84, 51-57.

BROACH, J. R., GUARASCIO, V. R. & JAYARAM, M. 1982. Recombination within the

yeast plasmid 2mu circle is site-specific. Cell, 29, 227-234.

BROADINSTITUTE. 2012. Rhizopus oryzae Project information [Online]. Available:

http://www.broadinstitute.org/annotation/genome/rhizopus_oryzae/Info.html

[Accessed 2012-07-03].

Page 58: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

47

BÜYÜKKILECI, A. O., HAMAMCI, H. & YUCEL, M. 2006. Lactate and ethanol

productions by Rhizopus oryzae ATCC 9363 and activities of related pyruvate branch

point enzymes. Journal of Bioscience and Bioengineering, 102, 464-466.

CAI, Y., LAI, C., LI, S., LIANG, Z., ZHU, M., LIANG, S. & WANG, J. 2011. Disruption of

lactate dehydrogenase through homologous recombination to improve bioethanol

production in Thermoanaerobacterium aotearoense. Enzyme and Microbial

Technology, 48, 155-161.

CANAN. T, KAMER ÖZKANÖ, SELALE ONCUÖ & TUBA AVCı 2011. The relationship

of pellet morphology tp Polygalactutonase production of Rhizopus oryzae in various

media compositions. GIDA, 36, 25-31.

CASE, M., SCHWEIZER, M., KUSHNER, S. & GILES, N. 1979. Efficient transformation of

Neurospora crassa by utilizing hybrid plasmid DNA. Proceedings of the National

Academy of Sciences, 76, 5259-5263.

CENTER, F. G. S. Available: http://www.fgsc.net/ [Accessed 08-03 2012].

COURT, D. A., GRIFFITHS, A. J., KRAUS, S. R., RUSSELL, P. J. & BERTRAND, H.

1991. A new senescence-inducing mitochondrial linear plasmid in field-isolated

Neurospora crassa strains from India. Current Genetics, 19, 129-137.

DEBETS, F., YANG, X. & GRIFFITHS, A. J. 1995. The dynamics of mitochondrial plasmids

in a Hawaiian population of Neurospora intermedia. Current Genetics, 29, 44-49.

EAMENS, A., WANG, M.-B., SMITH, N. & WATERHOUSE, P. 2008. RNA silencing in

plants: yesterday, today, and tomorrow. Plant physiology, 147, 456-468.

EDEBO, L. 2000. Cultivation of Zygomycetes from spent sulfite liquor World Patent

WO0063344

ELKINS, J., RAMAN, B. & KELLER, M. 2010. Engineered microbial systems for enhanced

conversion of lignocellulosic biomass. Current Opinion in Biotechnology, 21, 657-

662.

FERREIRA. A.J, PATRIK R. LENNARTSSON, CLAES NIKLASSON, MAGNUS

LUNDIN, LARS EDEBO & TAHERZADEH, M. J. 2012. "Production of Rhizopus

sp. from SSL" BioResources 7(1), 173-188. 173.

FINCHAM, J. R. S. 1989. Transformation in fungi. Microbiological Reviews, 53, 148-170.

FIRE, A., XU, S., MONTGOMERY, M., KOSTAS, S., DRIVER, S. & MELLO, C. 1998.

Potent and specific genetic interference by double-stranded RNA in Caenorhabditis

elegans. Nature, 391, 806-811.

FISCHER, T. S. 2011. FastDigest® HindIII [Online]. Available:

http://www.fermentas.com/en/products/all/fastdigest-restriction-enzymes/fd050-

hindiii [Accessed 2012-07-12].

FISHER, T. S. 2010. Fermentas, Dream TaqTM Green Polymerase [Online]. Available:

http://www.fermentas.de/product_info.php?info=p601 [Accessed 2012-07-11 2012].

FISHER, T. S. 2011a. FastDigest® XhoI [Online]. Available:

http://www.fermentas.com/en/products/all/fastdigest-restriction-enzymes/fd069-xhoi

[Accessed 2012-07012].

FISHER, T. S. 2011b. T4 DNA ligase [Online]. Available:

http://www.fermentas.com/en/products/all/molecular-cloning/enzymes/el001-t4-dna-

ligase [Accessed 2012-07-12 2012].

FITZGERALD, A., VAN KAN, J. & PLUMMER, K. 2004. Simultaneous silencing of

multiple genes in the apple scab fungus, Venturia inaequalis, by expression of RNA

with chimeric inverted repeats. Fungal genetics and biology : FG & B, 41, 963-971.

Page 59: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

48

GARCIA, B. 1968. Cell Wall Chemistry, Morphogenesis, and Taxonomy of Fungi. Annual

Review of Microbiology, 22, 87-108.

GHEINANI, A. H., JAHROMI, N. H., FEUK-LAGERSTEDT, E. & TAHERZADEH, M.

2011. RNA silencing of lactate dehydrogenase gene in Rhizopus oryzae. Journal of

RNAi and gene silencing : an international journal of RNA and gene targeting

research, 7, 443-448.

GRIFFITHS, A. F., KRAUS, S., BARTON, R., COURT, D., MYERS, C. & BERTRAND, H.

1990. Heterokaryotic transmission of senescence plasmid DNA in Neurospora.

Current Genetics, 17, 139-145.

GRIFFITHS, A. J. 1995. Natural plasmids of filamentous fungi. Microbiological Reviews, 59,

673-685.

GRYGANSKYI, A., LEE, S., LITVINTSEVA, A., SMITH, M., BONITO, G., PORTER, T.,

ANISHCHENKO, I., HEITMAN, J. & VILGALYS, R. 2010. Structure, Function, and

Phylogeny of the Mating Locus in the Rhizopus oryzae Complex. PLoS ONE, 5,

e15273.

HAMMOND, S. M., BOETTCHER, S., CAUDY, A. A., KOBAYASHI, R. & HANNON, G.

J. 2001. Argonaute2, a Link Between Genetic and Biochemical Analyses of RNAi.

Science, 293, 1146-1150.

HASUNUMA, T. & KONDO, A. 2011. Development of yeast cell factories for consolidated

bioprocessing of lignocellulose to bioethanol through cell surface engineering.

Biotechnology Advances.

HAWKSWORTH, D. 2011. A new dawn for the naming of fungi: impacts of decisions made

in Melbourne in July 2011 on the future publication and regulation of fungal names. 1,

7-20.

HAYES, D. J. 2009. An examination of biorefining processes, catalysts and challenges.

Catalysis Today, 145, 138-151.

HÉBRAUD, M. & FÈVRE, M. 1988. Protoplast production and regeneration from

mycorrhizal fungi and their use for isolation of mutants. Canadian Journal of

Microbiology, 34, 157-161.

HINNEN, A., HICKS, J. B. & FINK, G. R. 1978. Transformation of yeast. Proceedings of the

National Academy of Sciences, 75, 1929-1933.

HOSKINS. L, M., HAHN LAB 1998. Quick Yeast DNA Prep: Isolation of Total DNA

(genomic and plasmid) [Online]. Available:

http://labs.fhcrc.org/hahn/Methods/mol_bio_meth/yeast_quick_dna.html [Accessed

08-03 2012].

HUAN. S. A, HONG-YE LI & LIU, X.-H. 2004. Isolation and regeneration of protoplasts

from Penicillium digitatum. Chinese Journal of Agricultural Biotechnology, 1.

IBRAHIM, A., GEBREMARIAM, T., LIN, L., LUO, G., HUSSEINY, M., SKORY, C., FU,

Y., FRENCH, S., EDWARDS, J. & SPELLBERG, B. 2010. The high affinity iron

permease is a key virulence factor required for Rhizopus oryzae pathogenesis.

Molecular Microbiology, 77, 587-604.

ISHIDA, N., SUZUKI, T., TOKUHIRO, K., NAGAMORI, E., ONISHI, T., SAITOH, S.,

KITAMOTO, K. & TAKAHASHI, H. 2006. d-Lactic acid production by

metabolically engineered Saccharomyces cerevisiae. Journal of Bioscience and

Bioengineering, 101, 172-177.

JAHROMI, N. & GHEINANI, A. 2011 modified. RNA Silencing of Lactate Dehydrogenase

Gene in Rhizopus oryzae [Online]. Borås: University of Borås. Available:

http://hdl.handle.net/2320/7991 [Accessed Master].

Page 60: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

49

JAMES D. WATSON, T. A. B., STEPHEN P. BELL, ALEXANDER GANN, MICHAEL

LEVINE, RICHARD LOSICK 2008. Molecular Biology Of The Gene, CSHL Press.

KADOTANI, N., NAKAYASHIKI, H., TOSA, Y. & MAYAMA, S. 2003. RNA silencing in

the phytopathogenic fungus Magnaporthe oryzae. Molecular plant-microbe

interactions : MPMI, 16, 769-776.

KARIMI, K., EMTIAZI, G. & TAHERZADEH, M. 2006. Ethanol production from dilute-

acid pretreated rice straw by simultaneous saccharification and fermentation with

Mucor indicus, Rhizopus oryzae, and Saccharomyces cerevisiae. Papers from the 1st

International Conference on Environmental, Industrial and Applied Microbiology

(BioMicroWorld-2005), 40, 138-144.

KESINGTON. 2009. Asexual reproduction of Fungi. Fungi infection [Online]. Available

from: http://fungi.health-tips-diseases.com/2009/09/asexual-reproduction-of-

fungi.html [Accessed 08-21 2012].

KHATRI, M. & RAJAM, M. V. 2007. Targeting polyamines of Aspergillus nidulans by

siRNA specific to fungal ornithine decarboxylase gene. Medical mycology, 45, 211-

220.

KITAMOTO, N., YOSHINO, S., OHMIYA, K. & TSUKAGOSHI, N. 1999. Sequence

analysis, overexpression, and antisense inhibition of a β- xylosidase gene, xylA, from

Aspergillus oryzae KBN616. Applied and environmental microbiology, 65, 20-24.

KITPREECHAVANICH, V., MANEEBOON, T., KAYANO, Y. & SAKAI, K. 2008.

Comparative Characterization of l-Lactic Acid-Producing Thermotolerant Rhizopus

Fungi. Journal of Bioscience and Bioengineering, 106, 541-546.

KRAJAEJUN, T., GAUTHIER, G. M., RAPPLEYE, C. A., SULLIVAN, T. D. & KLEIN, B.

S. 2007. Development and application of a green fluorescent protein sentinel system

for identification of RNA interference in Blastomyces dermatitidis illuminates the role

of septin in morphogenesis and sporulation. Eukaryotic Cell, 6, 1299-1309.

KUWANO, T., SHIRATAKI, C. & ITOH, Y. 2008. Comparison between polyethylene

glycol- and polyethylenimine-mediated transformation of Aspergillus nidulans.

Current Genetics, 54, 95-103.

LA GRANGE, D. C., DEN HAAN, R. & VAN ZYL, W. H. 2010. Engineering cellulolytic

ability into bioprocessing organisms. Applied Microbiology & Biotechnology, 87,

1195-1208.

LENNARTSSON, P. 2012. Zygomycetes and cellulose residuals: hydrolysis, cultivation and

applications.PhD Thesis, Chalmers University of Technology, Göteborg.

LEWIS, M. 2001. Agarose gel electrophoresis (basic method) modified [Online]. Department

of Pathology, University of Liverpool. Available:

http://www.methodbook.net/dna/agarogel.html [Accessed 07-26 2012].

LINHUI. L, QINGYU YIN, XIAOHONG LIU & YANG, H. 2010. An efficient protoplast

isolation and regeneration system in Coprinus comatus. African Journal of

Biotechnology.

LIU, H., COTTRELL, T., PIERINI, L., GOLDMAN, W. & DOERING, T. 2002. RNA

interference in the pathogenic fungus Cryptococcus neoformans. Genetics, 160, 463-

470.

LIU, Z. & FRIESEN, T. 2012. Polyethylene Glycol (PEG)-Mediated Transformation in

Filamentous Fungal Pathogens Plant Fungal Pathogens. In: BOLTON, M. &

THOMMA, B. (eds.). Humana Press.

LOMBRAÑA, M., MORALEJO, F. J., PINTO, R. & MARTÍN, J. F. 2004. Modulation of

Aspergillus awamori thaumatin secretion by modification of bipA gene expression.

Applied and environmental microbiology, 70, 5145-5152.

Page 61: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

50

LYND, L., ZYL, W., MCBRIDE, J. & LASER, M. 2005. Consolidated bioprocessing of

cellulosic biomass: an update. Current Opinion in Biotechnology, 16, 577-583.

MARSHALL, O. 2011. PerlPrimer open source PCR primer design [Online]. Available:

http://perlprimer.sourceforge.net/download.html [Accessed 2012-07-10 2012].

MATHES, M. 2001. Biology 205: General Botany, Spring 2001 [Online]. College of William

and Mary. Available: http://www.resnet.wm.edu/~mcmath/bio205/index.html

[Accessed 08-21 2012].

MAY, G. & TAYLOR, J. W. 1989. Independent transfer of mitochondrial plasmids in

Neurospora crassa. Nature, 339, 320-322.

MCDONALD, T., BROWN, D., KELLER, N. & HAMMOND, T. 2005. RNA silencing of

mycotoxin production in Aspergillus and Fusarium species. Molecular plant-microbe

interactions : MPMI, 18, 539-545.

MEUSSEN, B., DE GRAAFF, L., SANDERS, J. & WEUSTHUIS, R. Metabolic engineering

of &lt;i&gt;Rhizopus oryzae&lt;/i&gt; for the production of platform chemicals.

Applied Microbiology and Biotechnology, 1-12.

MEUSSEN, B., GRAAFF, L., SANDERS, J. & WEUSTHUIS, R. 2012. Metabolic

engineering of Rhizopus oryzae for the production of platform chemicals. Applied

Microbiology and Biotechnology, 94, 875-886.

MEYER, V. 2008. Genetic engineering of filamentous fungi — Progress, obstacles and future

trends. Biotechnology Advances, 26, 177-185.

MEYER, V., MUELLER, D., STROWIG, T. & STAHL, U. 2003. Comparison of different

transformation methods for &lt;i&gt;Aspergillus giganteus&lt;/i&gt. Current

Genetics, 43, 371-377.

MICHIELSE, C. B., HOOYKAAS, P. J. J., VAN DEN HONDEL, C. A. M. J. J. & RAM, A.

F. J. 2005. Agrobacterium-mediated transformation as a tool for functional genomics

in fungi. Current Genetics, 48, 1-17.

MICHIELSE, C. B., SALIM, K., RAGAS, P., RAM, A. F., KUDLA, B., JARRY, B., PUNT,

P. J. & VAN DEN HONDEL, C. A. 2004. Development of a system for integrative

and stable transformation of the zygomycete Rhizopus oryzae by Agrobacterium-

mediated DNA transfer. Molecular genetics and genomics : MGG, 271, 499-510.

MICROBIOLOCIGALCONCEPTS. Reproduction: Fungi [Online]. Available:

http://bugs.bio.usyd.edu.au/learning/resources/CAL/Microconcepts/Reproduction/fung

iRepro.html [Accessed].

MILLATI, R., EDEBO, L. & TAHERZADEH, M. J. 2005. Performance of Rhizopus,

Rhizomucor, and Mucor in ethanol production from glucose, xylose, and wood

hydrolyzates. Enzyme and Microbial Technology, 36, 294-300.

MOLNAR, A., SCHWACH, F., STUDHOLME, D., THUENEMANN, E. & BAULCOMBE,

D. 2007. miRNAs control gene expression in the single-cell alga Chlamydomonas

reinhardtii. Nature, 447, 1126-1129.

MORALEJO, F. J., CARDOZA, R. E., GUTIERREZ, S., LOMBRAÑA, M., FIERRO, F. &

MARTÍNI, J. F. 2002. Silencing of the aspergillopepsin B (pepB) gene of Aspergillus

awamori by antisense RNA expression or protease removal by gene disruption results

in a large increase in thaumatin production. Applied and environmental microbiology,

68, 3550-3559.

MOUYNA, I., HENRY, C., DOERING, T. & LATGÉ, J.-P. 2004. Gene silencing with RNA

interference in the human pathogenic fungus Aspergillus fumigatus. FEMS

Microbiology Letters, 237, 317-324.

MÜLLER, D., TESCHE, M., EICHLER, H., ENGELMANN, R., ALTHAUSEN, D.,

ANSMANN, A., CHENG, Y. F., ZHANG, Y. H. & HU, M. 2006. Strong particle

Page 62: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

51

light absorption over the Pearl River Delta (south China) and Beijing (north China)

determined from combined Raman lidar and Sun photometer observations.

Geophysical Research Letters, 33.

NAKAYASHIKI, H., HANADA, S., QUOC, N. B., KADOTANI, N., TOSA, Y. &

MAYAMA, S. 2005. RNA silencing as a tool for exploring gene function in

ascomycete fungi. Fungal Genetics and Biology, 42, 275-283.

NAKAYASHIKI, H. & NGUYEN, Q. B. 2008. RNA interference: roles in fungal biology.

Current Opinion in Microbiology, 11, 494-502.

NGUYEN, Q. B., KADOTANI, N., KASAHARA, S., TOSA, Y., MAYAMA, S. &

NAKAYASHIKI, H. 2008. Systematic functional analysis of calcium-signalling

proteins in the genome of the rice-blast fungus, Magnaporthe oryzae, using a high-

throughput RNA-silencing system. Molecular Microbiology, 68, 1348-1365.

NOBELPRIZE.ORG. 2006. "Press Release: The 2006 Nobel Prize in Physiology or

Medicine" [Online]. Nobelprize.org. Available:

http://www.nobelprize.org/nobel_prizes/medicine/laureates/2006/press.html

[Accessed 18 Jan 2012 2012].

OHMASA, M., KIKUCHI, S. & KAWAKAMI, K. 1999. Preparation and regeneration of

protoplasts of three fungi of Boletaceae. Mycoscience, 40, 433-436.

PAROO, Z., LIU, Q. & WANG, X. 2007. Biochemical mechanisms of the RNA-induced

silencing complex. Cell Research, 17, 187-194.

PRITCHARD 1971. An NAD+-independent l-lactate dehydrogenase from Rhizopus oryzae.

Biochimica et Biophysica Acta (BBA) - Enzymology, 250, 25-34.

PUNT, P. J., OLIVER, R. P., DINGEMANSE, M. A., POUWELS, P. H. & VAN DEN

HONDEL, C. A. M. J. J. 1987. Transformation of Aspergillus based on the

hygromycin B resistance marker from Escherichia coli. Gene, 56, 117-124.

ROMANO, N. & MACINO, G. 1992. Quelling: transient inactivation of gene expression in

Neurospora crassa by transformation with homologous sequences. Molecular

Microbiology, 6, 3343-3353.

RUIZ-DÍEZ, B. 2002. A Review: Strategies for the transformation of filamentous fungi.

Journal of Applied Microbiology, 92, 189-195.

RUTZ, S. & SCHEFFOLD, A. 2004. Towards in vivo application of RNA interference - new

toys, old problems. Arthritis Res Ther, 6, 78-85.

SAKAGUCHI, K. 1990. Invertrons, a class of structurally and functionally related genetic

elements that includes linear DNA plasmids, transposable elements, and genomes of

adeno-type viruses. Microbiological Reviews, 54, 66-74.

SALAS, M. 1988. Initiation of DNA replication by primer proteins: bacteriophage phi 29 and

its relatives. Current topics in microbiology and immunology, 136, 71-88.

SAMBROOK & RUSSELL 2001 modified. Molecular cloning : A laboratory manual. 3rd.

SCHUMANN, U., AYLIFFE, M., KAZAN, K. & WANG, M.-B. 2010. RNA silencing in

fungi. Frontiers in Biology, 5, 478-494.

SCHUSTER, A. & SCHMOLL, M. 2010. Biology and biotechnology of

&lt;i&gt;Trichoderma&lt;/i&gt. Applied Microbiology and Biotechnology, 87, 787-

799.

SERGEEVA, Y., GALANINA, L. A., ANDRIANOVA, D. A. & FEOFILOVA, E. P. 2008.

Lipids of filamentous fungi as a material for producing biodiesel fuel. Applied

Biochemistry and Microbiology, 44, 523-527.

SHAFRAN, H., MIYARA, I., ESHED, R., PRUSKY, D. & SHERMAN, A. 2008.

Development of new tools for studying gene function in fungi based on the Gateway

system. Fungal Genetics and Biology, 45, 1147-1154.

Page 63: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

52

SINGH, A., KUMAR, P. & SCHÜGERL, K. 1992. Bioconversion of cellulosic materials to

ethanol by filamentous fungi

Enzymes and Products from Bacteria Fungi and Plant Cells. Springer Berlin / Heidelberg.

SKORY, C. 2004a. SILENCING OF LACTATE DEHYDROGENASE GENE OF

RHIZOPUS ORYZAE [abstract]. American Society for Microbiology. p. 79.

SKORY, C. D. 2000. Isolation and expression of lactate dehydrogenase genes from Rhizopus

oryzae. Applied and environmental microbiology, 66, 2343-2348.

SKORY, C. D. 2004b. Lactic acid production by &lt;i&gt;Rhizopus oryzae&lt;/i&gt;

transformants with modified lactate dehydrogenase activity. Applied Microbiology

and Biotechnology, 64, 237-242.

STEVENSON, D. M. & WEIMER, P. J. 2002. Isolation and characterization of a

Trichoderma strain capable of fermenting cellulose to ethanol. Applied Microbiology

and Biotechnology, 59, 721-726.

SUZZANE, H. & FLAVIN, C. 2007. Biofuels for Transport: Global Potential and

Implications for Sustainable ... - Worldwatch Institute - Google Books, Earthscan.

TAHERZADEH, M. & KARIMI, K. 2008. Pretreatment of lignocellulosic wastes to improve

ethanol and biogas production: a review. International journal of molecular sciences,

9, 1621-1651.

TAHERZADEH, M. J., FOX, M., HJORTH, H. & EDEBO, L. 2003. Production of mycelium

biomass and ethanol from paper pulp sulfite liquor by Rhizopus oryzae. Bioresource

Technology, 88, 167-177.

TAKARA. Yatalase [Online]. Available:

http://www.clontech.com/takara/US/Products/Molecular_Biology/Modifying_Enzyme

s/Nucleases/Yatalase [Accessed 2012-07018 2012].

TANGUAY, P., BOZZA, S. & BREUIL, C. 2006. Assessing RNAi frequency and efficiency

in Ophiostoma floccosum and O. piceae. Fungal Genetics and Biology, 43, 804-812.

TAYLOR, J., JACOBSON, D., KROKEN, S., KASUGA, T., GEISER, D., HIBBETT, D. &

FISHER, M. 2000. Phylogenetic Species Recognition and Species Concepts in Fungi.

Fungal Genetics and Biology, 31, 21-32.

TAYLOR, M., ELEY, K., MARTIN, S., TUFFIN, M., BURTON, S. & COWAN, D. 2009.

Thermophilic ethanologenesis: future prospects for second-generation bioethanol

production. Trends in Biotechnology, 27, 398-405.

TRESKATIS, S. K., ORGELDINGER, V., WOLF, H. & GILLES, E. D. 1997.

Morphological characterization of filamentous microorganisms in submerged cultures

by on-line digital image analysis and pattern recognition. Biotechnol. Bioeng., 53,

191-201.

TUNGA, R., BANERJEE, R. & BHATTACHARYA, B. C. 1999. Some studies on

optimization of extraction process for protease production in SSF. Bioprocess and

Biosystems Engineering, 20, 485-489.

TYE, Y., LEE, K., WAN ABDULLAH, W. & LEH, C. 2011. Second-generation bioethanol

as a sustainable energy source in Malaysia transportation sector: Status, potential and

future prospects. Renewable and Sustainable Energy Reviews, 15, 4521-4536.

UENG, P. P. & GONG, C.-S. 1982. Ethanol production from pentoses and sugar-cane

bagasse hemicellulose hydrolysate by Mucor and Fusarium species. Enzyme and

Microbial Technology, 4, 169-171.

UREGINA. Phenol/Chloroform Extraction and Ethanol Precipitation [Online]. Available:

http://uregina.ca/~ngdann/Bioc422/proj1.htm [Accessed 08-03 2012].

Page 64: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

53

WELD, R., PLUMMER, K., CARPENTER, M. & RIDGWAY, H. 2006. Approaches to

functional genomics in filamentous fungi. Cell Research, 16, 31-44.

WESLEY, S. V., HELLIWELL, C. A., SMITH, N. A., WANG, M. B., ROUSE, D. T., LIU,

Q., GOODING, P. S., SINGH, S. P., ABBOTT, D., STOUTJESDIJK, P. A.,

ROBINSON, S. P., GLEAVE, A. P., GREEN, A. G. & WATERHOUSE, P. M. 2001.

Construct design for efficient, effective and high-throughput gene silencing in plants.

The Plant journal : for cell and molecular biology, 27, 581-590.

WINSTANLEY, C. & RAPLEY, R. 2000. The Nucleic Acid Protocols Handbook: Extraction

and Purification of Plasmid DNA. Humana Press Inc.,Totowa, NJ.

WU, J. F., LASTICK, S. M. & UPDEGRAFF, D. M. 1986. Ethanol production from sugars

derived from plant biomass by a novel fungus. Nature, 321, 887-888.

WU, T., ZIVANOVIC, S., DRAUGHON, A., CONWAY, W. & SAMS, C. 2005.

Physicochemical properties and bioactivity of fungal chitin and chitosan. Journal of

agricultural and food chemistry, 53, 3888-3894.

YAMADA, O., IKEDA, R., OHKITA, Y., HAYASHI, R., SAKAMOTO, K. & AKITA, O.

2007. Gene silencing by RNA interference in the koji mold Aspergillus oryzae.

Bioscience, biotechnology, and biochemistry, 71, 138-144.

YANG, X. & GRIFFITHS, A. J. 1993. Plasmid diversity in senescent and nonsenescent

strains of Neurospora. Molecular & general genetics : MGG, 237, 177-186.

ZAMANI, A. 2010. Superabsorbent Polymers from the Cell Wall of Zygomycetes Fungi. PhD

Tesis, Chalmers University of Technology, Göteborg.

ZAMORE, P. D., TUSCHL, T., SHARP, P. A. & BARTEL, D. P. 2000. RNAi: Double-

Stranded RNA Directs the ATP-Dependent Cleavage of mRNA at 21 to 23 Nucleotide

Intervals. Cell, 101, 25-33.

ZHANG, M., EDDY, C., DEANDA, K., FINKELSTEIN, M. & PICATAGGIO, S. 1995.

Metabolic Engineering of a Pentose Metabolism Pathway in Ethanologenic

Zymomonas mobilis. Science, 267, 240-243.

ZHENG, X. F., KOBAYASHI. Y & M, T. 1998. Construction of a low-serine-type-

carboxypeptidase-producing mutant of Aspergillus oryzae by the expression of

antisense RNA and its use as a host for heterologous protein secretion. Applied

Microbiology and Biotechnology, 49, 39-44.

ZHONG, S., LENG, Y. & BOLTON, M. 2012. Construction of hairpin RNA-expressing

vectors for RNA-mediated gene silencing in fungi. Methods in molecular biology

(Clifton, N.J.), 835, 623-633.

ZHOU. X , YAMIN WEI, HUIFANG ZHU, ZINAN WANG, JUAN LIN, LU LIU & TANG,

K. 2008. Protoplast formation, regeneration and transformation from the taxol-

producing fungus Ozonium sp. African Journal of Biotechnology, 7, 2017-2024.

Page 65: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

Appendix:

A1: Preparations

Preparation 1: LB (Luria-Bertani) medium (100 ml) (bioprotocols.info)

Materials required:

Tryptone, Yeast extract, NaCL, Distilled water, autoclave

Procedure:

1 gram Tryptone, 0.5 gram Yeast extract and 0.5 gram NaCL were weighed and added to 100

ml distilled water in conical flask or bottle. The medium is then autoclaved at 121°C for 20

minutes and can be used for cultivation of organism or can be stored for further use without

opening the container.

Preparation 2: LB agar plates with ampicillin (bioprotocols.info)

Material required:

Tryptone, Yeast extract, NaCL, Agar, Distilled water, ampicillin stock solution, autoclave

Procedure:

LB medium (from preparation 1) was prepared and 15g/l agar was added.

Finally the volume was adjusted to 500 ml using distilled water and autoclaved at

121°C for 20 minutes.

The medium was cooled to 55°C and added with required amount ampicillin stock

solution and poured in to Petri dishes near the flame.

These petri dishes were undisturbed until the agar solidifies and sealed with parafilm

for preventing contamination. These were stored in fridge for 2-3 days for use.

Preparation 3: Salt solutions (100 ml each)

Materials Required:

(NH4)2SO4, KH2PO4, CaCl22H2O, MgSO47H2O, distilled water, Volumetric flasks, magnetic

stirrer, weighing balance, autoclave.

Procedure:

(NH4)2SO4: 37.5 g of (NH4)2SO4 dissolved in 50 ml distilled water using magnetic

stirrer and then made up to final volume of 100 ml using volumetric flask.

KH2PO4: 17.5 g of KH2PO4 dissolved in 50 ml distilled water made up to 100 ml as

before.

CaCl22H2O: 20 g of CaCl22H2O dissolved in distilled water and made up to 100 ml.

Page 66: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

MgSO47H2O: 15 g of MgSO47H2O dissolved in distilled water and made up to 100

ml.

All these salt solutions are prepared separate bottles and autoclaved at 121°C for 20 min and

stored at room temperature.

Preparation 4: Vitamin solution (Jahromi and Gheinani, 2011 modified)

Materials required:

D-Biotin, 0.1M NaOH, 0.1M HCL, p-amino benzoic acid (PABA), Nicotinic acid, Ca-

Panthothenate, Pyroxidine HCL, Thiamine HCL, 2 M NaOH, m-Inositol, Distilled water, 1.5

ml eppendorf tubes, 0.45 µm filters, syringes

Procedure:

25 mg of D-Biotin dissolved in 10 ml of 0.1M NaOH and then the dissolved biotin

solution was diluted using 300 ml distilled water.

pH was adjusted to 6.5 using 0.1M HCL and the remaining vitamins were added as

given below.

Table A1.4.1: Vitamin solution composition

PABA 100 mg

Nicotinic acid 500 mg

Ca-Panthothenate 500 mg

Pyroxidine HCL 500 mg

Thiamine HCL 500 mg

pH was adjusted to 6.5 after adding all the vitamins using 2M NaOH and 12.5 g of m

Inositol was added. pH was adjusted to 6.5 using 2M NaOH.

Finally the volume was made up to 500 ml using distilled water.

The solution was filled in to sterile 1.5 ml eppendorf tubes using 0.45 µm filters and

store at -20°C.

Preparation 5: Trace metal solution (Jahromi and Gheinani, 2011 modified)

Procedure:

The following components as given in table below were dissolved in 600 ml distilled water

and the pH was adjusted to 4 using NaOH. Finally volume was made up to 1 liter.

Page 67: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

Table: A1.5.1: Trace metal solution composition

Component Amount (in grams)

EDTA 3

CaCl2.2H2O 0.9

ZnSO4.7H2O 0.9

FeSO4.7H2O 0.6

H3BO3 0.2

MnCl2.4H2O 0.19

Na2MoO4.2H2O 0.08

CoCl2.2H2O 0.06

CuSO4.5H2O 0.06

KI 0.02

The solution prepared was autoclaved at 121°C for 20 minutes and store at 4°C for further

use.

Preparation 6: Semi synthetic medium for cultivation of Zygomycetes Fungi (1000 ml)

Materials Required:

Glucose, Yeast extract, Salt solutions (from preparation 3), Vitamin solution (from

preparation 4), Trace metal solution (from preparation 5), 2M NaOH

Procedure:

Glucose solution was prepared by dissolving 30 g glucose, 5 g yeast extract in 500 ml

distilled water in a Erlenmeyer flask, sealed with a cotton plug, aluminum foil and

autoclaved at 121°C for 20 minutes.

Salt solutions were prepared and autoclaved in separate blue capped bottles as

described in preparation 2 and each of them was added in to the flask containing

autoclaved glucose solution in the order shown below near the flame after cooling.

Page 68: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

Table A1.6.1: Salt solutions composition

Salt solution Amount (ml)

(NH4)2SO4 20

KH2PO4 20

CaCl22H2O 5

MgSO47H2O 5

Distilled water 439

10 ml trace metal solution and 1 ml vitamin solution was added to the above mixture

in sterile conditions.

Care should be taken to prevent contamination while adding the components and this

medium can be used for inoculation with spores or mycelium of filamentous fungi.

Preparation 7: Potato Dextrose Agar plates

Material required:

Potato extract, Glucose, Agar, distilled water

Procedure:

2 g potato extract, 10 g glucose and 7.5 g agar was added to 500 ml distilled water and

autoclaved for 20 minutes at 121°C.

After cooling down to 55°C, media was poured in to petri plates near the flame and

undisturbed until solidified. The plates were sealed with parafilm and stored at room

temperature or fridge for longer storage.

Preparation 8: Yatalase Digestion Buffer (Takara)

Materials required:

0.2 M Maleate Buffer [5.8 g Maleic acid+1NaOH(50 ml)+250 ml distilled water]

MgSO4, Yatalase Enzyme from (Takara), distilled water.

Procedure:

Yatalase enzyme was in the form of powder and1 g was dissolved in 10 ml distilled

water in a 15 ml sterile plastic tube using vortex machine to get uniform solution.

Page 69: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

3.611 g of MgSO4 was dissolved in12.5 ml of 0.2M Maleate Buffer and 5 ml distilled

water.

Both the solutions were mixed and the pH was adjusted to 5.5 using 0.2N NaOH.

Final volume was adjusted to 50 ml using distilled water and can be used immediately

for effectiveness or can be stored at 4°C for 2-3 days.

Note: It was difficult to filter sterilize the solution as the solution was very thick and it can be

centrifuged and upper solution was used for protoplast preparation.

Preparation 9: STC Buffer

Material required:

Sorbitol, CaCl2, 1M Tris HCL (pH 8.0), distilled water

Procedure:

91.09 g Sorbitol and 2.774 g CaCl2 were dissolved in distilled water separately and

mixed.

25ml of 1m Tris HCL was added to the mixture and final volume was adjusted to 500

ml using distilled water.

The solution was autoclaved for 20 minutes at 121°C and stored at room temperature.

Preparation 10: PEG solution (25ml)

Materials required:

PEG 4000, CaCl2, 1M Tris HCl (pH 8), distilled water

Procedure:

15 g of PEG 4000 and 130 mg of CaCl2 were dissolved in 15 ml of 50mM Tris HCL

in 25ml volumetric flask

Finally volume was adjusted to 25 ml using distilled water and autoclaved at 121°C

for 20 minutes and stored at room temperature.

Preparation 11: 10X TBE Buffer (bioprotocols.info)

Materials Required:

Tris base, 0.5M EDTA, Boric acid, distilled water.

Procedure:

The following compounds were mixed in a500 ml blue capped bottle using 200 ml distilled

water.

Tris base – 54 gram

Page 70: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

Boric acid – 27.5 gram

0.5 M EDTA – 20 ml

Finally the volume of the buffer was made up to 500 ml using a measuring cylinder and was

stored at room temperatures for further use.

Preparation 12: Spore solution (Jahromi and Gheinani, 2011 modified)

Materials Required:

Distilled water, PDA agar plate (from preparation 7), Spreaders, disposable pipettes

Procedure:

The PDA plates were inoculated with Rhizopus Oryzae and incubated for 3 days at

30°C for formation of spores.

The procedure was performed near flame to avoid contamination by wearing gloves.

20ml of distilled water was added on the surface of agar plate containing the spores.

The disposable sterile microbiological spreader was used to scrub the surface of agar plate to

get spores and hyphae suspended in water. (as shown in figure below)

The disposable sterile pipette (5ml) was used for transferring the spore suspension as

inoculum. The spore suspension was used immediately.

Viability of frozen spore suspension was not yet accessed

Figure A1.12.1: Spores being suspended using a sterile spreader

Page 71: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

A2: List of Material used:

10X Dream TaqTM

Green buffer –

Fermentas

2 propanol - Scharlau

Agar bacterialogical - Scharlau

Agarose - Sigma life sciences

Ammonium sulphate - Sigma Aldrich

Ampicillin - Scharlau

BglII – Fermentas

Boric acid - Sigma life sciences

Calcium chloride - J: T Baker chemical

co..phillipsburg

D- Sorbitol - Sigma life sciences

D-Glucose (anhydrous reagent grade) -

Scharlau

Dream TaqTM

Polymerase (5u/µl) –

Fermentas

EDTA - Sigma Aldrich

Ethanol 96%, v/v (analytical grade,ACS,

reagent , USP - Scharlau

Glass beads, acid washed -Sigma life

sciences

HindIII – Invitrogen

KpnI – Invitrogen

Lysozyme - Fisher scientific

Magnesium sulphate -Merck

PCR purification kit – Wizard SV Gel and

PCR clean up system, Prime

Phenol/chloroform /isomyl alchol(25:24:1)

ratio - Fisher scientific

Polyethylene glycol l(PEG 4000) - Fluka

biochemika

Potassium iodide - Scharlau

Potassium phosphate - Scharlau

Potato extract - Fluka

Primers - Invitrogen

SDS (Sodium dodecyl sulphate) - Merck

Sodium chloride - Scharlau

Sodium hydroxide - Scharlau

Sodium phosphate - Sigma life sciences

T4 DNA ligase – Fermentas

Trizma base - Sigma life sciences

Tryptone - Fluka analytical

XhoI – Fermentas

Yatalase (Takara) – Fisher Scientific

Yeast extract – Scharlau

Page 72: Construction of hpRNA expression vector for silencing a ...bada.hb.se/bitstream/2320/11676/1/Penmatsa Balu.pdf · ii Construction of hpRNA expression vector for silencing LDHA gene

A3: List of Equipments used:

Autoclave – SHP Steri Hechnik AB

Centrifuge – mini Scan speed (2010)

Electrophoresis equipment – Easy cast electrophoresis system, model #B2, Owl Scientific Inc.

Glass beads (5mm) – Scientific Industries, inc

Heavy Phase Lock gelTM

tubes – 5 prime

Mega fuge 1.0 R – Heraeus sepatech

Microfuge tubes (10ml, 50 ml) – TPP (ISO certified 9001), Switzerland

Microscope – Zeiss, Axio cam MRC

Petri plates – NUNCTM

, made in Denmark

Pipette (sterile) - TPP

Shaking incubator – New Brunswick Scientific Excella E24 Incubator Shaker Series

Spectrophotometer – Nano drop 2000, Thermo scientific

Thermal cycler – Biometra Trio ThermoblockTM

, Biometra Trio Heated lid

UV lamp – UVP Cambridge, U. K

Voltmeter – Concort E443, Microcomputer electrophoresis power supply

Vortex machine – SI vortex-2 gene®