The RNA interference mechanism in molecular...

144
DIPLOMARBEIT The RNA interference mechanism in molecular research in general, detailed in the model organism Caenorhabditis elegans as well as in terms of nutritional research. Julia Katharina Liberda angestrebter akademischer Grad Magistra der Naturwissenschaften (Mag.rer.nat.) Wien, 2012 Studienkennzahl lt. Studienblatt: A 474 Studienrichtung lt. Studienblatt: Diplomstudium Ernährungswissenschaften Betreuer: Univ.-Prof. Dr. Jürgen König

Transcript of The RNA interference mechanism in molecular...

Page 1: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

DIPLOMARBEIT

The RNA interference mechanism in molecular research in general, detailed in the model

organism Caenorhabditis elegans as well as in terms of nutritional research.

Julia Katharina Liberda

angestrebter akademischer Grad

Magistra der Naturwissenschaften (Mag.rer.nat.)

Wien, 2012

Studienkennzahl lt. Studienblatt: A 474

Studienrichtung lt. Studienblatt: Diplomstudium Ernährungswissenschaften

Betreuer: Univ.-Prof. Dr. Jürgen König

Page 2: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 3: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 4: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 5: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

I

I. ACKNOWLEDGEMENTS

For my parents, Christine and Eugen, who encouraged and supported me at

every step of my life. Thank you for all your patience, endless love, and your

belief in me.

I would like to say thank you to all, who helped and supported me, that I finally

could successfully finish this paper.

Especially, I would like to thank Univ.-Prof. Dr. Jürgen König for all his time,

guidance, and advises, as well as Mag. Dr. Elisabeth Rudolph-König for her

time and assistance with my last steps.

Special thanks to my girls, who made my time at the university special and

unforgettable.

I would like to thank my brother, for opening his house and giving me a warm

and lovely home for half a year, while I tried to write all those following pages.

Thanks to my sister Alexandra for spending some nights, reading and correcting

all those pages.

Special thanks to my sister Michaela. Thanks you for all our great adventures,

your brilliant or at least funny advices and all together, for an awesome time

together.

Last but not least, special thanks to my love Jonas, for standing always next to

my side, giving me strength, support, and love at every moment, since we met.

Page 6: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 7: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

III

II. CONTENTS

I.  ACKNOWLEDGEMENTS ............................................................................. I 

II.  CONTENTS ................................................................................................ III 

III.  LIST OF FIGURES ................................................................................ VII 

IV.  LIST OF ABBREVIATIONS .................................................................... IX 

1.  Introduction .............................................................................................. 1 

2.  The different terms ................................................................................... 3 

2.1  Homology-dependent gene silencing ................................................ 3 

2.2  Transcriptional gene silencing ........................................................... 3 

2.3  Post-transcriptional gene silencing .................................................... 4 

2.4  Virus-induced gene silencing ............................................................ 4 

2.5  Co-suppression ................................................................................. 4 

2.6  RNA interference ............................................................................... 5 

3.  RNA interference - mechanism ................................................................ 7 

3.1  Discovery of RNA interference .......................................................... 9 

3.2  Important milestones in the discovery of RNA interference ............. 12 

3.3  The natural role of RNAi .................................................................. 14 

3.3.1  Ancient defense mechanism ...................................................... 14 

3.3.2  Transposon silencing and genome stabilization ......................... 16 

3.3.3  Immune function ........................................................................ 17 

3.3.4  Regulation of developmental timing ........................................... 17 

3.3.5  Involvement in heterochromatin modification and methylation ... 18 

3.4  The four big players ........................................................................ 18 

3.4.1  Double-stranded RNA ................................................................ 19 

3.4.2  Dicer........................................................................................... 21 

3.4.3  Small non-coding RNAs ............................................................. 22 

Page 8: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

IV

3.4.3.1  Small interfering RNAs ......................................................... 23 

3.4.3.1.1  Newly discovered small RNAs ....................................... 25 

3.4.3.2  MicroRNAs ........................................................................... 26 

3.4.3.2.1  Biogenesis of miRNAs ................................................... 29 

3.4.3.2.2  Small temporal RNAs: lin-4 and let-7 ............................. 32 

3.4.3.3  Difference between siRNAs and miRNAs ............................ 33 

3.4.3.4  Small nuclear RNA ............................................................... 34 

3.4.3.5  Tiny non-coding RNAs ......................................................... 35 

3.4.3.6  Small modulatory RNAs ....................................................... 35 

3.4.3.7  PIWI-interacting RNAs ......................................................... 35 

3.4.4  RNA induced silencing complex and microRNA ribonucleoprotein

complex .................................................................................................. 36 

3.5  Regulation of RNAi ......................................................................... 39 

3.6  Difficulties for RNA interference in mammals .................................. 39 

3.7  Structural design and chemical modifications ................................. 42 

3.7.1  Structural information for the design of double-stranded RNAs . 42 

3.7.2  Parameters for the design of small interfering RNAs ................. 43 

3.8  Special delivery methods ................................................................ 47 

3.8.1  RNA interference through chemically synthesized siRNAs ........ 47 

3.8.2  Short-hairpin RNA-mediated gene silencing .............................. 50 

3.8.3  Transfection agents ................................................................... 53 

3.9  Advantages and limitations of the RNA interference mechanism .... 56 

3.9.1  Off-target effects ........................................................................ 60 

3.10  Outlook ........................................................................................... 62 

3.10.1  The use of RNAi in the development of novel drugs ............... 63 

4.  RNA interference in the model organism Caenorhabditis elegans ......... 67 

Page 9: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

V

4.1  Introduction of Caenorhabditis elegans as model organism ............ 67 

4.2  RNA interference in Caenorhabditis elegans .................................. 70 

4.3  Specialties of the RNA interference mechanism in Caenorhabditis

elegans ...................................................................................................... 73 

4.3.1  Systemic RNAi ........................................................................... 73 

4.3.2  Inheritance ................................................................................. 75 

4.3.3  Amplification / RNA-dependent RNA polymerase ...................... 76 

4.3.4  Activation of transitive RNA interference under the generation of

secondary small interfering RNAs .......................................................... 79 

4.3.5  Genes required for RNAi in Caenorhabditis elegans ................. 81 

4.4  Practical aspects and methods of RNAi in Caenorhabditis elegans 85 

4.4.1  Microinjection ............................................................................. 87 

4.4.2  Feeding of dsRNA expressing bacteria ...................................... 89 

4.4.3  Soaking of Caenorhabditis elegans in dsRNA solution .............. 91 

4.4.4  Multiple gene interactions .......................................................... 93 

4.5  Knockout of the reduced folate carrier ............................................ 93 

5.  Conclusion ............................................................................................. 95 

V.  SUMMARY ............................................................................................. XVII 

VI.  ZUSAMMENFASSUNG ....................................................................... XIX 

VII.  REFERENCES ..................................................................................... XXI 

VIII.  CURRICULUM VITAE .......................................................................... XLI 

Page 10: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 11: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

VII

III. LIST OF FIGURES

Fig. 1 RNAi-mediated gene silencing. ................................................................. 8 

Fig. 2 Transgenic petunia flowers ....................................................................... 9 

Fig. 3 Two-step model ...................................................................................... 19 

Fig. 4 Cleavage of dsRNA by dicer ................................................................... 21 

Fig. 5 Typical structure of small interfering RNA ............................................... 23 

Fig. 6 A model for miRNA biogenesis and the translational inhibition of the

target RNA ........................................................................................................ 31 

Fig. 7 Predicted structure of lin-4 and let-7 ....................................................... 32 

Fig. 8 aǀ siRNA pathway bǀ microRNA pathway ................................................ 34 

Fig. 9 Target recognition and target cleavage by the RISC complex ................ 38 

Fig. 10 Activation of the PKR response and the 2‘-5‘ oligoadenylate synthase

pathway ............................................................................................................ 40 

Fig. 11 The design of an effective siRNA .......................................................... 46 

Fig. 12 Strategies to deliver the silencing signal into the organism ................... 49 

Fig. 13 Strategies for siRNA production in vivo and in vitro .............................. 55 

Fig. 14 Life cycle of C. elegans at 22°C ............................................................ 68 

Fig. 15 Generation of an inherited agent through RDE-1 and RDE-4 and

persistence of the silencing signal trough RDE-2 and MUT-7 ........................... 75 

Fig. 16 Amplification mechanism in worms and plants ...................................... 78 

Fig. 17 The four possible methods for RNA transfection ................................... 86 

Fig. 18 Predicted structure of the folt-1 protein ................................................. 94 

Page 12: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 13: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

IX

IV. LIST OF ABBREVIATIONS

21U RNA RNA, 21 nucleotides in length, starting with Uridine

A Adenine / Adenosine

Arabidopsis t. Arabidopsis thaliana

ATP Adenosine 5’-triphosphate

bp Base pair

C Cytosine / Cytidine

C. elegans Caenorhabditis elegans

CGC Caenorhabditis Genetic Center

CNS Central nervous system

DExH/DEAH RNA helicase domain

dFMR Homologous to the human fragile X mental

retardation protein

Dicer RNase III enzyme

DIDS 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid

DNA Deoxyribonucleic Acid

Drosha RNase III enzyme

Drosophila m. Drosophila melanogaster

d-siRNA Diced siRNA

dsRNA Double-stranded RNA

E. coli Escherichia coli

Page 14: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

X

E. coli strain HT115 E. coli strain, which lacks in double-strand-specific

RNase III

elF2C1 Translation initiation factor 2 C1

elF2C2 Translation initiation factor 2 C2

elF2α Translation initiation factor 2 alpha

endo-siRNA Endogenous small interfering RNA

esiRNA Endonuclease-generated short interfering RNA

F1 First filial generation

G Guanine/Guanosine

H1 Human promoter region

HCV Hepatitis C

HDGS Homology-dependent gene silencing

HIV Human immunodeficiency virus

hRFC Human reduced folate carrier

isRNA Immunostimulatory RNA

kDA Kilodalton

L1-L4 Larval stages of C. elegans

Mg²+ Magnesium

miRNA MicroRNA

miRNA* Complementary strand of miRNA

miRNP MicroRNA Ribonucleoprotein Complex

mRNA Messenger RNA

Page 15: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XI

N Any Nucleotide

N2 C. elegans wild type Bristol N2

Na+ Natrium

Neurospora c. Neurospora crassa

nM Nano Mol

nt Nucleotide

nuclear factor-кB Nuclear factor kappa-light-chain-enhancer of

activated B cells

PAZ Domain conserved in PIWI, Argonaute, Zwille

PCR Polymerase chain reaction

pH Hydrogen ion concentration

piRNA PIWI-interacting RNA

PKR DsRNA-dependent protein kinase

Pol II RNA polymerase II

Pol III RNA polymerase III

PPD domain Containing a PAZ and a PIWI domain

Pre-miRNA MicroRNA precursor

Pri-miRNA Primary microRNA transcript

PTGS Post-transcriptional gene silencing

R2D2 Tandem dsRNA binding domain (R2)-Dicer (D2)

complex

rasi-RNA Repeat-associated small interfering RNA

RdDM RNA-directed DNA-methylation

Page 16: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XII

RdRP RNA-dependent RNA Polymerase

RFC Reduced folate carrier

RISC RNA Induced Silencing Complex

RISC* Activated RNA Induced Silencing Complex

RNA Ribonucleic acid

RNAi RNA Interference

SAS Systemic acquired silencing

scnRNA Small scan RNA

sdRNA Sno-derived RNA

shRNA Short hairpin RNA

siRNA Small-interfering RNA

SITS 4-acetamido-4’-isothiocyanostilbene-2,2’-disulfonic acid

smRNA Small modulatory RNA

snoRNA Small nuclear RNA

SNP Single nucleotide polymorphism

snRNA promoter Promoter element in human

stRNA Small temporal RNA

T Thymine/Thymidine

T7 promoter Recognition sequence for T7 RNA polymerase

tasiRNA Endogenous trans-acting RNA

Tc1 Transposon in C. elegans

TGS Transcriptional gene silencing

Page 17: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XIII

tnc-RNA Tiny-non-coding RNA

Tudor SN Tudor staphylococcal nuclease

U Uracil/Uridine

U6 promoter Gene/Promoter element in mice

UTR Untranslated Region

VIG Vasa intronic gene

VIGS Virus-induced-gene-silencing

VSV Vesicular Stomatitis Virus

Genes:

ago-1 Protein ArGOnaute (Arabidopsis t.)

ago-2 ArGOnaute 2 (Drosophila m.)

alg-1 Argonaute (plant)-Like Gene (C. elegans)

alg-2 Argonaute (plant)-Like Gene (C. elegans)

ban BANtam (Drosophila m.)

csr-1 Chromosome-Segregation and RNAi deficient (C.

elegans)

dcr-1 DiCeR-1 (Drosophila m.) / DiCer Related (C.

elegans)

ego-1 Enhancer of Glp-One (C. elegans)

eri-1 Enhanced RNAI (C. elegans)

folt-1 FOLate Transporter family (C. elegans)

folt-2 FOLate Transporter family (C. elegans)

Page 18: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XIV

Gemin3 alias DDX20 DeaD (Aps-Glu-Ala-Asp) boX polypeptide 20 (Homo

sapiens)

Gemin4 Gem (nuclear organelle) associated protein 4 (Homo

sapiens)

hbl-1 HunchBack Like (fly gap gene related) (C. elegans)

hda-1 Histone DeAcetylase (C. elegans)

him High Incidence of Males (C. elegans)

isw-1 yeast ISW (imitation SWI) homolog (C. elegans)

let-7 LEThal (Drosophila m.)

lin-4 abnormal cell LINeage (C. elegans)

lin-14 abnormal cell LINeage (C. elegans)

lin-15B abnormal cell LINeage (C. elegans)

lin-28 abnormal cell LINeage (C. elegans)

lin-35 abnormal cell LINeage (C. elegans)

lin-41 abnormal cell LINeage (C. elegans)

mrg-1 human MRG (Mortality factor-Related Gene) (C.

elegans)

mut-1 MUTator (C. elegans)

mut-2 alias rde-3 MUTator (C. elegans)

mut-6 MUTator (C. elegans)

mut-7 MUTator (C. elegans)

mut-9 MUTator (C. elegans)

nrde-3 Nuclear RNAi DEfective (C. elegans)

Page 19: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XV

qde-1 Quelling-DEfective 1 (Neurospora c.)

qde-2 Quelling-DEfective 2 (Neurospora c.)

rde-1 RNAi DEfective (C. elegans)

rde-2 alias mut-8 RNAi DEfective (C. elegans)

rde-4 RNAi DEfective (C. elegans)

rrf-1 RNA-dependent RNA polymerase Family (C.

elegans)

rrf-2 RNA-dependent RNA polymerase Family (C.

elegans)

rrf-3 RNA-dependent RNA polymerase Family (C.

elegans)

rsd-2 RNAi Spreading Defective (C. elegans)

rsd-3 RNAi Spreading Defective (C. elegans)

rsd-4 RNAi Spreading Defective (C. elegans)

rsd-6 RNAi Spreading Defective (C. elegans)

sde1 RNA-dependent RNA polymerase (Arabidopsis t.)

sgs2 PTGS deficient mutant (C. elegans)

sid-1 alias rsd-8 Systemic RNA Interference Deficient (C. elegans)

sting aubergine (Drosophila m.)

unc-22 UNCoridinated (C. elegans)

Page 20: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 21: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

1

1. Introduction

This paper provides a summary of the RNA interference mechanisms, from the

beginning till today. All the great discoveries, the smaller successes, as well as

the drawbacks of the past couple years are put together to build a whole picture

of the effect.

Already the fact, that the discovery of the RNA interference mechanism has

been less than two decades ago, and the new technology got adopted already

quickly in labs around the world and is used nowadays as a standard tool in the

genetic-tool-box, shows how important this mechanism is [HOWARD, 2003]

[SUGIMOTO, 2004]. RNA interference has the power to connect forward and

reverse genetics, which implicates the direct link of biological functions and

gene sequencing [CHANG et al., 2005].

C. elegans developed into the model organism of choice, when it comes down

to RNAi approaches in the first years. This is based on the perfectly timed

coincidence of sequencing the genome of C. elegans and the finding of the

Interference effect [SUGIMOTO, 2004]. Not only was the silencing effect

discovered first in the worm, also the first microRNAs, including their function in

regulation of developmental timing were discovered in the worm [FISCHER,

2010]. More and more details of the mechanism were identified, such as an

RNAse III enzyme called dicer, which cleaves long double-stranded RNA into

one of the main players of the silencing effect called small-interfering RNAs, the

knock-down mechanism linked with the microRNA pathway, and the discovery

and the identification of some parts of the effector complex referred to as RNA

induced silencing complex [COUZIN, 2002] [AGRAWAL et al., 2003]. Through

this expanding information and the identification of the machinery of RNAi,

every step of a whole physiological pathways and its function can be explored

[LEUNG & WHITTAKER, 2005].

With the knowledge of the potency and specificity, the easy handling, and the

possible application into mammalian cells, big hopes arose for developing new

therapeutic agents or even individualized therapy [SEMIZAROV et al., 2003]

Page 22: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

2

[PADDISON et al., 2002]. By a short review of knocking out the reduced folate

carrier of the model organism C. elegans, it will be shown that this tool can also

be maintained in nutritional research.

Till today, the complete mechanism with all players and co-factors is still not

unrevealed und further research will be necessary to be able to see the

complete picture [SHAN, 2010].

Page 23: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

3

2. The different terms

A whole group of different but closely related phenomena represent a

conserved ancestral process [MELLO & CONTE JR., 2004]. RNA interference

(RNAi) in animals, post-transcriptional gene silencing (PTGS), co-suppression,

and virus-induced-gene-silencing (VIGS) in plants, and quelling in fungi

describe similar effects. Today, all of them are referred to as RNA silencing

[PLASTERK, 2002]. All of them provide sequence-specific degradation of

homologous messenger RNA, are indicated to function in different regulation

pathways, share some homologous genes, like ego-1 in C. elegans, sde1 in

Arabidopsis t., and qde-1 in Neurospora crassa, but they appear phenotypically

different [CATALANOTTO et al., 2000] [AGRAWAL et al., 2003] [COGONI &

MACINO, 2000].

2.1 Homology-dependent gene silencing

Homology-dependent gene silencing (HDGS) describes a silencing

phenomenon of homologous nucleic acid sequences. It can occur either at the

transcriptional level through a higher number of promoter methylations, or at the

post-transcriptional level in the cytoplasm, through degradation of the

complementary sequence (target) to the trigger [KOOTER et al., 1999]. This

term HDGS, summarizes many homology-dependent silencing mechanisms

including RNAi, co-suppression, VIGS, and quelling [PADDISON et al., 2002].

2.2 Transcriptional gene silencing

In transcriptional gene silencing (TGS), like the name already explains, the

silencing effect occurs at the transcriptional level through methylations of the

promoter sequences. Compared to post-transcriptional gene silencing, TGS is a

stable change of the genome and as a result of this alterations, it is heritable

into the next generations [HANNON, 2002].

Page 24: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

4

2.3 Post-transcriptional gene silencing

Post-transcriptional gene silencing (PTGS) was first discovered in plants. It is a

natural silencing mechanism that is activated by viruses, mobile genetic

elements such as transposons or transgenes, and is implicated in gene

regulation in a variety of biological processes [SIJEN & KOOTER, 2000]

[COGONI & MACINO, 2000]. PTGS has the ability to spread through the

organism and can also be seen in the progeny [BASS, 2000].

2.4 Virus-induced gene silencing

Post-transcriptional gene silencing triggered by viruses is referred to as virus-

induced gene silencing (VIGS) [COGONI & MACINO, 2000]. Some of the

viruses developed strategies to inhibit the process, so that they can escape the

degradation. VIGS is not heritable, but has, like PTGS, the ability to spread

through different tissues [BERNSTEIN et al., 2001] [PLASTERK, 2002].

2.5 Co-suppression

Co-suppression is related with RNAi; both use some of the same gene

products, both act either post-transcriptionally or transcriptionally, both are

implicated in transposon silencing, but they are not identical [GRISHOK,

2005b]. For example in contrast to co-suppression, RNAi has the ability to

spread in C. elegans [KETTING et al., 2003]. Co-suppression represents the

possibility to reduce the expression of both, the introduced transgenes and their

homologous endogenous gene, and was first discovered in petunia [TUSCHL,

2001] [BERNSTEIN et al., 2001]. The co-suppression phenomenon in

Neurospora crassa is called quelling [COGONI & MACINO, 2000].

Page 25: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

5

2.6 RNA interference

RNA interference (RNAi) is a widespread phenomenon and is emerged in

organisms ranging from plants, fungi, insects to mammalian. The term is

normally used to describe the degradation of homologous messenger RNA

triggered by double-stranded RNA [KETTING et al., 2001].

Page 26: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 27: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

7

3. RNA interference - mechanism

RNA interference is an evolutionarily conserved gene silencing phenomenon

[SIJEN et al., 2001]. The RNAi effect is not a phenomenon restricted to one

organism, like the first assumption was. It was pointed out to work in a variety of

organisms, such as plants, nematodes, protozoa, and insects and later also in

vertebrates [CAPLEN et al., 2001]. First it was suggested, that RNAi works only

at the post-transcriptional level. But now, evidence shows, that RNAi works

post-transcriptionally (degradation of mRNA) as well as transcriptionally

(suppression of the transcription) [AGRAWAL et al., 2003].

After years of research, it is proved that RNA interference and other silencing

phenomena like PTGS, quelling, co-suppression are closely linked and related

to each other [ELBASHIR et al., 2001b]. All mechanisms require some related

proteins and use some parts of each other’s pathway [ZAMORE, 2002]. The

phenomenon is referred to RNAi because of the function. After injection of

double-stranded RNA (dsRNA) into the cell/tissue, a highly specific silencing of

the complementary sequence to the introduced dsRNA is displayed [ELBASHIR

et al., 2001b]. After the recognition of dsRNA, chemically synthesized small

interfering RNAs (siRNAs), endogenous expressed siRNAs, or microRNAs

(miRNAs), a complementary-dependent degradation of the target mRNA gets

started [LEE et al., 2006]. This degradation results in the absence of the

cytoplasmic transcript of the target or more exactly, only a very low level can be

measured, because the accumulation of the normal cytoplasmic concentration

was disturbed. Due to the fact that the nuclear gene expression was still

working, and not perturbed at all, the degradation step was placed into the

cytoplasm [COGONI & MACINO, 2000]. In some organisms it is possible that

the effect gets inherited to the next generation, but it is important to mention,

that there are no genetical changes of the genome involved KETTING et al.,

2003]. RNA interference is a mechanism which is dependent on the

concentration of the trigger molecule and is temporarily limited [TIMMONS et

al., 2003]. This ancient, but newly discovered mechanism made rapid gene-

Page 28: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

8

function analysis possible; first in worms and flies and afterwards also in

mammalian cells [ZAMORE, 2001].

Fig. 1 RNAi-mediated gene silencing.

Through processing of either long dsRNA, plasmid-based shRNA, or endogenous miRNA by an RNAseIII enzyme dicer, the RNA gets cleaved into smaller fragments (siRNA), with the characteristic 2-3nt 3’overhangs and the 5’ phosphate group. In contrast, the synthetic siRNA is converted in the active formby an endogenous kinase. Small interfering RNAs associate with RISC to guide the mRNA cleavage. Ingeneral, the strand with the lower stability at the 5’ end enters preferred RISC. An RNA with a perfectmatch to the target RNA act like a siRNA and results in mRNA degradation, and RNA with a partialmatch works as an miRNA and results in translational repression. [MITTAL, 2004]

Page 29: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

9

3.1 Discovery of RNA interference

RNA interference is one of the most important discoveries in the last century,

and was honored with the Nobel Price for physiology or medicine in the year

2006 [Nobelprize.org, 2011].

„RNAi itself is at least one billion years old. Biological mechanisms

are far more constant than the positions of continents on our planet”

[MELLO, 2007].

For long time, it seemed that RNAs act only

as messengers and possess catalytical,

structural, and information decoding function

in the biogenesis of proteins. But with the

discovery of RNAi and the growing

knowledge of the mechanism, the picture

changed, and it got more and more

complicated [NOVINA & SHARP, 2004].

Silencing of an endogenous gene was

discovered by accident. Introduction of

transgenes in purple petunia plants, with the

intention to make the flower darker purple,

resulted in white or patchy plants. There was

no expected over-expression of the color;

both, the transgene and the expression of

the endogenous gene were silenced. The

observed phenomenon was termed co-

suppression. Co-suppression is widespread

in the kingdom of plants, and can also be

found in fungi, termed quelling. The ability of

Fig. 2 Transgenic petunia flowers

Through supplementation of a chalcone synthase transgene, the purple pigmentation is rather suppressed than enhanced. modified from [NAPOLI et al., 1990]

Page 30: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

10

the silencing signal to travel long distances to other tissues of the plants was

seen [COGONI & MACINO, 2000].

The mechanism was thought to act as a “primitive immune system” of the

genome [PLASTERK, 2002]. The hypothesis that it must be an evolutionarily

conserved, natural defense response against “parasitic” sequences was born

[KOOTER et al., 1999].

RNAi arose to a powerful tool and one of the standard methods for silencing of

gene expression in diverse organisms. It is a simple method for knocking-down

the gene expression in diverse organisms. It started with expanding our

understanding and knowledge at the whole genome level in Caenorhabditis

elegans. The discovery of RNA interference in C. elegans and the completion of

its whole genome sequence was a perfect coincidence [SUGIMOTO, 2004].

Andrew Z. Fire and coworkers published their first observations that introduction

of either antisense or sense RNA into Caenorhabditis elegans were equally

effective in inhibiting specific gene expression [ZAMORE et al., 2000].

Introduction of long double-stranded RNA was at least tenfold more efficient

than the single-stranded sense or antisense RNA alone [HANNON, 2002]. The

surprising result that both single-stranded RNAs work with the same efficiency

could be figured out. They showed that neither purified sense nor antisense

RNA could introduce any silencing effect. The potent trigger for the silencing

effect resulted from double-stranded RNA. The solution of the puzzle was found

in contaminations of the single-stranded RNA by double-stranded RNA

[ZAMORE et al., 2000]. Craig C. Mello named the mechanisms after the

observed silencing processes, RNA interference [FIRE, 2007].

Introduction of smaller and smaller amounts indicated that already a few

molecules per cell, of double-stranded RNA, were efficient enough to initiate

RNA interference. Not only that a few molecules per cell can trigger silencing, it

was also seen that the silencing occurred in tissues in the entire body. The

effect lasted up to several days, and in some cases it was inherited into the next

generations [FIRE, 2007]. Because of those results, it was suggested that the

Page 31: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

11

mechanism involves a catalytical and/or amplificational step [SIJEN &

KOOTER, 2000].

Phenotypes of null-mutants were created after injection of dsRNA [COGONI &

MACINO, 2000]. Further research found an easier method to deliver dsRNA

into the worm. It was found that feeding C. elegans with bacteria expressing

dsRNA corresponding to unc-22 resulted in similar phenotypes like unc-22-

mutants [HUNTER, 2000]. Also soaking in a high concentrated dsRNA solution

was efficient enough to induce the silencing phenomenon. Those efforts made it

possible to perform large-scale genome-wide RNAi screens. From this point on,

it was possible to search for all genes that are essential in diverse biochemical

pathways like embryogenesis, longevity, apoptosis, and more [GOLDEN &

O'CONNELL, 2007].

The newly discovered phenomenon worked not only in plants and nematodes. It

was also present in Arabidopsis thaliana, Xenopus laevis, Drosophila

melanogaster, Trypanosoma brucei, Hydra magnipapillata, and zebrafish

[BARSTEAD, 2001]. To find the mechanism also in mammalian cell lines, more

time had to pass [ELBASHIR et al., 2001a]. Experiments suggested that the

“core” mechanism of the silencing process is preserved in all those named

species. So the evolutionarily ancient nature of RNA interference was

uncovered and proved [SUGIMOTO, 2004]. Interestingly, there is no evidence

for RNAi in archea and prokaryotes, so it seems that RNA silencing is an

innovation from the eukaryotic organisms [ZAMORE, 2002].

The possibility that different small RNA classes could be the key to the RNAi

process came from experiments in plant systems. A class of small RNAs,

ranging in size from 21 to 25 nucleotides, was found. And it looked like they

were closely related to the silencing mechanisms. Evidence showed that those

small RNAs, referred to as small interfering RNAs, derive from the long double-

stranded RNA. Dicer, an RNase III enzyme cleaves the long dsRNA into these

smaller fragments [FIRE, 2007]. Through cleavage, the important characteristic

structures of the 5’ and 3’ end of siRNAs are generated [CAPLEN et al., 2001].

It was further suggested, that a multiprotein complex, later referred to as RNA

Page 32: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

12

induced silencing complex (RISC) recognizes the complement mRNA and

initiates the degradation [HUTVÁGNER & ZAMORE, 2002].

A lot of research was done in those years, and a lot of discoveries were made.

It got possible to inject successfully chemically synthesized siRNAs into

mammalian cells, to induce the RNAi effect. The first successful approach was

with embryo cells of mice [ZAMORE, 2001]. Furthermore vectors, employed as

transfection vehicles, were invented, such as plasmid-based or viral-based

vectors. Nowadays, these vectors are well integrated into the tool box for RNAi

[SHI, 2003]. Also not only similarities were discovered, differences like

introduction of long double-stranded RNA into mammalian cells, resulted in

activation of non-specific dsRNA response such as apoptosis. Systemic RNA

interference was only observed in plants and C. elegans. And the proposed

amplification step, including the use of target mRNA as template, employment

of RNA-dependent RNA polymerases (RdRPs), and generation of secondary

siRNAs, was only seen in C. elegans [SUGIMOTO, 2004].

New drugs and therapeutic agents can be developed, since the connection of

genomics, proteomics, and functional genomics is provided through the

knowledge of genome sequences of diverse species and the attribute of RNAi

to investigate details of physiological processes and pathways [SCHEPERS,

2005].

3.2 Important milestones in the discovery of RNA interference

1974, Sydney Brenner introduced the new model organism C. elegans

[BRENNER, 1974].

1979, the Caenorhabditis Genetic Center (CGC) was founded [STIERNAGLE,

1999 reprinted 2005].

PTGS was first discovered through an experimental accident in the plant

petunia in the late 1980s [LAI, 2003].

Page 33: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

13

1993, the first non-coding RNA (later called microRNA), lin-4 was successfully

cloned [HE & HANNON, 2004].

In 1995, it was established that introduction of either antisense or sense RNA

resulted in RNA-mediated interference [SUGIMOTO, 2004].

1998, Caenorhabditis elegans was the first multicellular organisms with its

whole genome completely sequenced [KIM, 2001]. It was perfectly timed with

the discovery, that injection of double-stranded RNA into nematodes, results in

sequence-specific gene silencing, termed RNA interference by Craig C. Mello

[DYKXHOORN et al., 2003] [FIRE, 2007].

Post-transcriptional gene silencing was established in 1999 as antiviral

mechanisms in plants [PARRISH et al., 2000].

2000, the genome of Drosophila melanogaster was completely sequenced

[KIM, 2001]. RNA induced silencing complex was purified in Drosophila cells

[HAMMOND et al, 2000]. Let-7, a second microRNA was discovered in C.

elegans, and it was shown that it is conserved in diverse bilaterian species [HE

& HANNON, 2004]. RNA interference was successfully shown in mouse

embryos [WIANNY & ZERNICKA-GOETZ, 2000].

2001, the human genome was completely sequenced [KIM, 2001]. The enzyme

dicer was identified [COUZIN, 2002]. Small 21-23 nucleotide fragments, cleaved

from long double-stranded RNA, seem to mediate RNA interference

[ELBASHIR et al., 2001b]. During the year, the link between small interfering

RNA and microRNA biogenesis was proved [LAI, 2003]. It was demonstrated

that synthetic double-stranded siRNAs induce RNAi in different mammalian cell

cultures, including HeLa cells [ELBASHIR et al., 2001a].

2002, it got possible to transfect siRNAs into the targeted cell through plasmid-

or viral-vectors [SHI, 2003]. In the same year, it was discovered that

approximately 1% of the metazoan genome encodes microRNAs, and proved

their function in the regulation of gene expression [WADHWA et al., 2004].

Page 34: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

14

For their discovery of the gene silencing method RNAi, by dsRNA, the Nobel

Prize in physiology or medicine 2006 was awarded to Andrew. Z. Fire and Craig

C. Mello [Nobelprize.org, 2011].

3.3 The natural role of RNAi

RNA interference is not only a silencing mechanism, where corresponding

messenger RNA is degraded by an RNA trigger. The mechanism emerged most

likely as mechanism to protect the genome of invading elements, and works as

an ancient immune system [DYKXHOORN & LIEBERMAN, 2005].

RNAi or more specific the “core” machinery of RNAi is linked with a lot of other

natural functions, like antiviral response to invading viruses, protecting the

genome against transposable elements, maintaining the stability of the genome,

introduction of a non-specific interferon response, remodeling of chromatin

structure, and regulation of gene expression and developmental programs, as

well as cell differentiation and cell death [DYKXHOORN & LIEBERMAN, 2005]

[KIM et al., 2005]. The whole picture is still not complete and more research will

help to identify the missing parts [SHAN, 2010].

3.3.1 Ancient defense mechanism

It is necessary for every organism to protect its genetic code against invasions

by mobile genetic elements, such as transposons and viruses, and genetic

modifications [ELBASHIR et al., 2001b]. It seems that RNA interference is the

most ubiquitous and ancient antiviral system developed in plants as well as

animals [SHARP, 2001]. The first assumptions that RNA interference and post-

transcriptional gene silencing might represent an ancient cellular defense

mechanisms evolved already early. These assumptions are based on

observations such as; first, viral RNAs can be targets for PTGS; second, a

number of viruses evolved mechanisms to bypass the PTGS-reaction; third,

through the spreading effect, from cell to cell, a systemic response is possible;

Page 35: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

15

and fourth, plants without an effective PTGS response are more affected by

some virus-infections [FIRE, 1999].

Because double-stranded RNA is a key intermediate in the life-cycle of many

viruses, and their ability to produce aberrant RNA, it is suggested that by

generation of double-stranded RNA the defense mechanisms could be

activated [FIRE, 2007] [TUSCHL, 2001]. Viruses might act as targets and

inducers for the activation of the whole process, involving the generation of

short dsRNA fragments by dicer, the cleavage and the destruction of the target

RNA [TUSCHL, 2001] [MARTINEZ et al., 2002]. And indeed, confirmation was

found. So far, for C. elegans, no natural viral pathogens are identified. By

introduction of a mammalian pathogen called vesicular stomatitis virus (VSV)

into the nematode, reliable defense mechanisms were detected. RNAi-deficient

mutants display higher affinity for infections than the wild-type strain N2. Data

also confirmed that some components of the RNAi pathway are involved, like

RDE-1 and RDE-4. In addition, in RNAi-sensitive strains, rrf-3 and eri-1

mutants, the percentage of infected cells were significantly lower than in N2

[WILKINS et al., 2005]. Also observations, that some viruses are capable to

inhibit the spreading effect through the plasmodesmata of the plant, and the

discovery that some viruses encode proteins with the ability to suppress the

PTGS phenomenon, to bypass the cellular immunity, and allow further viral

replication, approved the early assumptions [BERNSTEIN et al., 2001]

[WILKINS, 2005].

In summary there is enough evidence of the ability of RNAi to function as

defense mechanism against viruses and/or other selfish elements in simple

organisms. But it might be possible that the antiviral effect of RNAi has been

lost in mammals [FIRE, 2007].

Page 36: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

16

3.3.2 Transposon silencing and genome stabilization

One key function for every organism is to provide the stability of the genomic

sequence. Evidence showed us, that RNAi is employed to maintain the genomic

stability [NYKÄNEN, 2001]. Transposons are endogenous selfish/parasitic

mobile genetic elements with the ability to jump around within the genome.

They either move as RNA sequences (retrotransposons) or as DNA fragments

and integrate into the genome via replicative mechanisms or via a “cut-and-

paste” mechanism. Up to 45% of the human genome and 12% of the C.

elegans genome is built by these mobile elements, and different numbers of

copies are present in the cells [VASTENHOUW & PLASTERK, 2004]. It is

important to regulate them strictly, because their activity can result in genome

instability by acting as potential mutagens. They can provide potential sites for

non-homologous crossover during DNA repair, as well as changed activity of

gene functions can occur [BERNSTEIN, 2001]. At this point, RNA interference

appears in the picture. It is possible that RNAi may play its part in the silencing

of transposons. It was shown, that alteration in four genes, mut-1, mut-7, rde-2,

and rde-3 lead to a decreased RNAi effect, but an increases mobilization of the

mobile elements [TABARA et al., 1999]. One way to control the activity is to

suppress the mobilization of transposons and the accumulation of repetitive

DNA sequences in germline [ZAMORE, 2001]. Suggestions implicate RNAi as

defense mechanisms against the accumulation of those mobile elements in the

germline [TABARA et al., 1999]. Double-stranded RNA might act as trigger, or a

second possibility, transposon transcripts (for example Tc1-transcripts) may

serve as template for RdRPs resulting in double-stranded RNA, to induce

transposon silencing. Afterwards a cleavage step by dicer might generate Tc1-

siRNAs, and the corresponding targets get destroyed [VASTENHOUW &

PLASTERK, 2004]. It seems that transposon silencing in germline works mainly

post-transcriptional [GRISHOK, 2005b].

It is an important fact to keep in mind, that transposon silencing is often counted

as an RNAi phenomenon, but actually those two mechanisms are not identical,

they only share some factors and their pathways converge [FISCHER, 2010].

Page 37: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

17

3.3.3 Immune function

It was early recognized that long double-stranded RNAs are very potent triggers

of non-specific responses, especially activation of an interferon response in

vertebrate systems [SHARP, 1999]. RNA silencing, especially triggered by

dsRNA, can be seen as one part of an innate immune system against RNA

viruses, transposable elements, and other double-stranded RNA invasions

[MEISTER & TUSCHL, 2004].

Long double-stranded RNA, at least 30 nucleotides in length, activates the non-

specific immune response [PADDISON et al., 2002]. As a result, an antiviral

state of the cell is activated, which leads to a general shutdown of the protein

synthesis of the affected cell [COGONI & MACINO, 2000]. The dsRNA-

dependent protein kinase (PKR) gets activated and further inactivates the

translation factor eIF2α by phosphorylation. This results in an inactivation of the

RNAi effect, translation, and protein synthesis and leads to the initiation of a

non-apoptotic and apoptotic suicide program of the cell [CAPLEN et al., 2001].

To bypass this immune response, viruses developed a variety of skills to inhibit

the activation of the PKR response [MONTGOMERY & FIRE, 1998].

3.3.4 Regulation of developmental timing

The influence of RNA interference to regulate time developmental pathways is

diverse. For example: dicer (dcr-1) mutants in C. elegans display various

developmental phenotypes, including abnormal oocytes, inability to fertilize

eggs, seam-cell production is altered, and a non-functional vulva which tends to

burst. Lin-4 and let-7, both genes, encode small non-coding RNA fragments,

which are involved in the developmental timing of C. elegans [BERNSTEIN et

al., 2001]. Another gene, ego-1, is required for germline development and

fertility in C. elegans [MAINE, 2001].

Page 38: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

18

3.3.5 Involvement in heterochromatin modification and methylation

The first evidence was discovered in plants; afterwards also in fungi and

animals, that co-suppression is linked with modifications in chromatin structures

[BERNSTEIN et al., 2001]. It was suggested that RNAi influences also the gene

expression at the chromatin level in various organisms [HANNON, 2002]. Years

later, the RNAi mechanism was actually correlated with the chromatin regulation

in fission yeast, and the important role of RNAi in heterochromatin silencing was

established in all three kingdoms (plants, fungi, and animals) [MELLO &

CONTE JR., 2004]. It was further discovered that modifications at the chromatin

level by RNAi plays a part for the inheritance of the silencing signal [MELLO,

2007]. Evidence was found, that via the RNAi mechanism, double-stranded

RNA or rather small interfering RNAs are involved in chromatin DNA

modifications and heterochromatic gene silencing [AGRAWAL et al., 2003]. It is

now established that RNAi plays also an important part in the regulation of gene

expression, chromosome behavior, and evolution [LIPPMAN &

MARTIENSSEN, 2004].

RNA-directed DNA-methylation (RdDM) is another mechanism to influence

chromatin alteration. It describes the process, predominantly in plants, where

nearly all sensitive cytosine residues at the homologous regions of the

chromatin to the trigger dsRNAs are methylated [AGRAWAL et al., 2003].

3.4 The four big players

After years of speculations and suggestions, the mechanism is now mostly

solved. Not all of the first assumptions proved to be wrong, like the thoughts

that the mechanism is a bit variable in different organisms, but uses the same,

evolutionarily conserved, core components and the same pathway [PARRISH et

al., 2000]. RNAi works as a two-step mechanism, the initiation phase and the

effector phase. Those two steps include the cleavage of double-stranded RNA

into small interfering RNAs by the RNase III enzyme called dicer (step one).

These newly generated siRNAs are incorporated into a multi-protein enzyme

Page 39: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

19

called RISC and guide the actual

degradation of the corresponding mRNA

(step two) [ZAMORE, 2001]. Through

the destruction of the mRNA, no further

translation into a protein is possible and

further results that the displayed

phenotype shows the function of the

silenced gene [TIJSTERMAN et al.,

2002]. The following parts, describe the

four key players, double-stranded RNA,

RNase III enzyme dicer, small

interfering RNA, and the RNA induced

silencing complex, of the RNAi

machinery and their function in the

whole mechanism.

3.4.1 Double-stranded RNA

Double-stranded RNA interference is established as a very powerful, simple,

and rapid tool to inhibit gene expression of the corresponding gene. Introduction

of dsRNA in species as different as C. elegans, Drosophila m., and

Trypanosoma brucei. results in cleavage of homologous endogenous

messenger RNA [FIRE, 1999]. The transcription of the affected gene is not

disturbed, only the accumulation of the transcripts is not possible, because of

the degradation [CATALANOTTO et al., 2000].

“Double-stranded RNA might be as old or nearly as old as life

on earth” [FIRE, 2007].

Fig. 3 Two-step model

[TIJSTERMAN, et al., 2002]

Page 40: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

20

Long time it was thought, that the key role for dsRNA was established in the

replication cycle of viruses, and that there was no other place for double-

stranded RNA in the normal cell information. Double-stranded DNA and single-

stranded RNA was thought to be employed only as short-term information

storage [FIRE, 2007]. Double-stranded RNAs were recognized the first time in

plants and afterwards observed in a variety of organisms. The conservation of

those fragments suggested an ancestral mechanism [SIJEN & KOOTER, 2000].

With the discovery of RNAi, it was recognized that dsRNAs can do a lot more

than actually assumed [FIRE, 2007]. And it was found that some viruses

developed mechanisms to partially or fully resist the degradation [FIRE, 1999].

Already a few molecules per cell are potent enough to trigger the silencing

effect in C. elegans and Drosophila. So the suggestions, there might exist a

catalytically and/or an amplification feature, were mentioned [MONTGOMERY &

FIRE, 1998]. Without the knowledge of siRNAs, dsRNA was thought to be the

key trigger of the silencing effect, regardless if the dsRNA origins from

exogenous or endogenous sources [PARRISH et al., 2000]. Researchers

figured out the effective length of dsRNA in diverse experiments. Double-

stranded RNA under 49bp was ineffective, slightly active was dsRNA with

149bp, and obvious robust effects were displayed at a length of 505bp and

997bp [TUSCHL et al., 1999]. Other published results indicated that dsRNA

must be longer than ~200bp to be effective [BASS, 2000]. Further results

pointed out that longer dsRNA are processed more efficiently than shorter ones.

That correlated with the observations that long double-stranded RNA induces

more effectively RNA interference [KETTING et al., 2001]. It was also figured

out, that only exon sequences have silencing activity and no intron or promoter

sequence is active [FIRE et al., 1998]. One exception appeared; an intron

sequence is then effective, when also an exon sequence is present [TABARA et

al., 1998]. Potent silencing was found only with 96% of identical sequence to

the target, 78% and 72% resulted in no, and 88% triggered only less

interference [PARRISH et al., 2000].

Page 41: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

21

3.4.2 Dicer

Dicer, an RNase III enzyme,

cleaves the long dsRNA into

smaller fragments, of around 21-

25 nucleotides in length. Those

fragments are the true

intermediates of the RNA

interference phenomenon and are

referred to as small interfering

RNAs [ZAMORE, 2001]. This

cleavage step explains why

dsRNA shorter than 30bp are

inefficient to induce RNAi, in contrast to longer ones [ELBASHIR et al., 2001b].

RNase III is the only known enzyme that can cleave dsRNA at the specifically

suggested site to form smaller fragments of around 21-25 nucleotides in length

and include the characteristic structure of the 5’ end phosphate-, 3’ end

hydroxyl-group, and two-nucleotides 3’ end overhang [BASS, 2000] [ELBASHIR

et al., 2001b]. Dicer is an evolutionarily conserved family of proteins among a lot

of organisms such as fungi, plants, and animals including C. elegans,

Drosophila m., and mammals [TIJSTERMAN et al., 2002].

RNase III enzymes are represented by three families. The first one, the

bacterial RNase III, is composed of a single catalytical domain and a dsRNA-

family binding domain [HANNON, 2002]. The second family is represented by

drosha (from Drosophila), consisting of the C-termini two RNase III domains,

one binding domain, and an N-termini with still unknown function [SHARP,

2001] [TUSCHL, 2001]. The enzyme drosha will be further discussed in chapter

3.4.3.2.1., biogenesis of miRNAs. The third class represents the dicer family.

Dicer, located in the cytoplasm, is a multidomain ribonuclease III protein

containing 6 different domains [KENT & MacMILLAN, 2004]. The N-terminal,

composed of an ATP dependent DExH/DEAH RNA helicase domain, PAZ

domain, and a DUF283 domain with unknown function; the C-terminal contains

Fig. 4 Cleavage of dsRNA by dicer

modified from [NYKÄNEN et al., 2001]

Page 42: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

22

two RNase III catalytic domains and a dsRNA-binding motif [PADDISON &

HANNON, 2002] [HE & HANNON, 2004].

Evidence for the requirement of dicer, for the RNA interference pathway, is

existent. Homology of dicer exists among many organisms such as DCR-1

between Drosophila and C. elegans [KETTING et al., 2001]. Some organisms

encode more than one dicer homologue. Suggestions indicate that each dicer

processes dsRNA from a specific source. Individuals with only one dicer

homologue may contain proteins to help dicer recognize the different sources of

dsRNA [MEISTER & TUSCHL, 2004]. Dicer-defective animals are sterile and

exhibit developmental abnormalities during larval growth. Dcr-1 mutants are

defective for RNAi in germline, but not in somatic cells [KETTING, et al., 2001].

For the cleavage of double-stranded RNA, ATP is needed to form the smaller

duplex fragments [NYKÄNEN et al., 2001]. It is not clear if ATP is the limiting

factor, because in presence of Mg2+ and absence of ATP the production of

siRNA was also visible [AGRAWAL et al., 2003].

Additional to the production of siRNAs, dicer is also employed in the maturation

of microRNAs. Precursors, deriving from the nucleus, are cleaved into

microRNAs, in a length of about 21-25 nucleotides [HE & HANNON, 2004]. It

was also demonstrated that dicer-defective organisms displayed defective

miRNA maturation [MELLO, 2007].

3.4.3 Small non-coding RNAs

A new discipline developed with the discovery of the small non-coding RNAs,

RNomics. It is the science of the structure, the enzymatic and regulatory

function of the non-coding fragments [APPASANI, 2005].

Small RNA molecules are key players of an ancient and conserved form of

silencing [ALMEIDA & ALLSHIRE, 2005]. But it is not their only function. It

seems that they are employed in many biochemical pathways but most of the

functions remain unknown [LAU et al., 2001]. RNA interference’s two main

members are miRNAs and siRNAs [COUZIN, 2002]. Next to those two, tiny-

Page 43: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

23

non-coding RNAs (tncRNAs), small nuclear RNAs (snoRNAs), and small

modulatory RNAs (smRNAs) are already discovered and certainly some more

will be found [KIM, 2005] [TAFT et al., 2009].

3.4.3.1 Small interfering RNAs

The function of small interfering RNAs was recognized the first time in plants

and afterwards in Drosophila embryo lysate [COGONI & MACINO, 2000] [LIM

et al., 2003]. It is now established that siRNAs are no byproducts of the double-

stranded cleavage reaction by dicer. Instead, they are the true intermediates of

the RNAi reaction [ZAMORE, 2001]. Small interfering RNAs appear to be

universal in all organisms. Only the length is a little bit variable and species-

dependent, but that might reflect the different dicer homologous [HUTVÁGNER

& ZAMORE, 2002]. SiRNAs are double-stranded RNAs about 21-25 nucleotides

in length. The characteristic structure of each side of each strand includes the

5’phosphate- and the 3’hydroxyl ends and overhangs of 2 basepairs on the 3’

end on every side [CAPLEN et al., 2001].

SiRNAs are incorporated into a multicomponent enzyme, termed RNA induced

silencing complex, and function as guide RNAs to recognize the perfect

complementary mRNA and initiate their degradation [TUSCHL, 2002]. The

already mentioned structural characteristics are important to enter the silencing

pathway. The phosphate group on the 5’ end is necessary for the RISC activity.

Introduction of synthetic siRNAs with free 5’ ends are immediately

phosphorylated [MARTINEZ et al., 2002]. In general, siRNAs with the lower

stability at the 5’ end enter preferred RISC and are more effective silencers

Fig. 5 Typical structure of small interfering RNA

[DYKXHOORN, et al., 2003]

Page 44: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

24

[HANNON & ROSSI, 2004]. Small RNAs with the characteristic double-

nucleotide 3’ overhang are more effective than similar ones with blunt-ends

[TUSCHL, 2001]. Modifications are tolerated on both ends of the sense strand

in contrast to the antisense strand. Every chemical change at the antisense

strand abolishes the function as guide of RNAi and no silencing effect is

displayed [MARTINEZ et al., 2002]. After the duplex strand splits, only the

antisense strand is detected. While the sense strand is degraded, the antisense

strand of the RNA duplex is incorporated into RISC to cleave the mRNA starting

from the center [AGRAWAL et al., 2003] [SHARP, 2001]. This explains why

modifications of the sense strand cause no obvious problems [TUSCHL, 2001].

The target is cleaved in regular intervals, around 21-23 nucleotides, in the

region of absolute complementarity between siRNAs and the mRNA, and get

degraded afterwards [TIJSTERMAN et al., 2002]. Already single basepair

mismatches between the guide strand and the target can abolish the silencing

effect [SHARP, 2001].

Long double-stranded RNA injected into mammalian cells trigger non-specific

effects. Those effects are discussed in more details in chapter 3.6., difficulties

for RNA interference in mammals. Experiments verified that chemical

synthesized siRNAs can bypass those effects and induce efficient mRNA

degradation, also in mammalian cells [CAPLEN et al., 2001]. Another

characteristic of siRNAs is, that they can function similar to miRNA and inhibit

translation of mRNA instead of the degradation from mRNA. The mechanism

depends on the degree of the complementarity between the siRNA and the

mRNA [BARTEL, 2004].

Many advantages pushed the adaption of siRNAs as research tool. Problems,

such as stability of the duplex and delivery into the organisms, were already

figured out early by trying to adapt the promising antisense oligonucleotides as

therapeutic agents. The method is easy to work with, the costs are high but

reasonable [PAROO & COREY, 2004]. SiRNAs are highly sequence-specific,

are working at low concentrations, and can be used for large-scale screens and

high-throughput experiments, regarding gene function and drug validation

Page 45: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

25

[BOESE et al., 2005]. The hope for using small interfering RNAs in the future as

therapeutic agents is very high [CAPLEN et al., 2001]. The high efficiency, the

specificity, and the endurance of the silencing made siRNAs one of the best

tools for target validation in biomedical research [TUSCHL & BORKHARDT,

2002]. Not only advantages are found for introduction of siRNAs. One limitation

includes the transitory of the silencing mechanisms. After the transfection of

siRNAs, the silencing effect persists only for around one week. To avoid this

drawback, diverse delivery methods were developed, such as siRNA

expressing vectors [PADDISON et al., 2002].

3.4.3.1.1 Newly discovered small RNAs

In the last years, many different subclasses of small interfering RNAs were

discovered. Here, I provide a short overview of those I came across during my

research.

Repeat-associated small interfering RNAs (rasiRNAs) are composed of

homologous regions of repetitive DNA such as transposons, retrotransposons,

centromeric repeats, satellite, and microsatellite DNA [DYKXHOORN &

LIEBERMAN, 2005]. Their role is established in heterochromatin silencing by

regulation of methyltransferases and histone-modifying enzymes [KIM, 2005].

Endogenous small interfering RNAs (endo-siRNAs), 22nt, are generated from

protein-coding sequences and are suggested to have perfect complementarity

to their targets in Caenorhabditis elegans [AMBROS et al., 2003]. The

biogenesis is so far not clear, but it might be possible that they involve dicer,

RNA-dependent RNA polymerases, and other mechanisms shared with the

RNAi pathway [FISCHER, 2010]. It seems that their role is established in

chromatin methylation and silencing [LAI, 2003].

Small scan RNAs (scnRNAs) are also involved in the regulation of histone

methylation. It is suggested that they are also generated by dicer, but their

whole biogenesis and their roles are still unknown and need further research

[KIM, 2005].

Page 46: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

26

The endogenous trans-acting RNAs (tasiRNAs) are, so far, discovered only in

plants and nematodes which exhibit RNA-dependent RNA polymerases activity.

It seems that they origin from introns of non-coding genes. Their functions

remain to be identified [KIM, 2005].

New studies emerged that some siRNA might be able to trigger an interferon I

response, the production of pro-inflammatory cytokines, and the activation of

nuclear factor-кB. All those siRNAs are referred to immunostimulatory RNAs

(isRNAs). It seems that small RNAs with 3’ blunt-ends and GU-rich sequences

have greater potential to induce those responses than others [SHAN, 2010].

Also other important characteristics like length, double/single-strand

configuration, and nucleoside modifications have influence on the

immunostimulatory power [SCHLEE et al., 2006].

3.4.3.2 MicroRNAs

The discovery of miRNAs was very surprising, nobody expected small RNAs to

encode, instead of a protein, an RNA fragment [MELLO, 2007]. The first

identified microRNA, in 1993, was lin-4 and 7 years later the second one, let-7,

was found [HE & HANNON, 2004]. Those two were referred to as small-

temporal RNAs (stRNAs), because of their function in regulation of the

developmental timing in C. elegans [LAU et al., 2001]. Lin-4 seemed to be

restricted to C. elegans and did not have any homologous in other organisms.

So in the beginning, it was thought that the phenomenon is limited to C.

elegans. After the detection of let-7 and its homology in other organisms the

picture changed, and researchers recognized that they discovered only the tip

of the iceberg [ZAMORE, 2001]. Till 2002, hundreds of miRNAs have been

identified in organisms, ranging from worms to flies to humans and plants

[AGRAWAL et al., 2003]. The approximated numbers of miRNAs in those

organisms range between 0.5% and 1% of all contained genes in the genome,

for example in C. elegans: 105 ± 15 and in humans: 230 ± 30. In single-cell

organisms no miRNAs were identified [LIM et al., 2003]. This whole group of

Page 47: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

27

non-coding tiny RNAs is referred to as microRNAs, including the subclass of

stRNAs [LAU et al., 2001]. Some of them are highly evolutionarily conserved

between flies, worms, and humans [MOSS, 2002]. Nearly one third of all known

C. elegans miRNAs are close in sequences to one or more insects, and/or

vertebrate miRNAs [AMBROS et al., 2003]. Others, like lin-4 exist only in one

organism [MOSS, 2002].

Functions of miRNAs are largely unknown, except for lin-4 and let-7 [AMBROS,

2001]. The fast rising number of identified miRNAs established the idea that not

all of the identified miRNAs work as regulation factor of developmental timing

[HE & HANNON, 2004]. This argument and the evolutionarily conservation

implied, that they must function in a variety of regulatory pathways, additionally

to their already discovered role as regulation factor in form of lin-4 and let-7

[LAU et al., 2001]. Suggestions are even that far; if miRNAs are as widespread

and diverse as they seem to be, they might be able to play their part as

regulation/control factors in multifaceted pathways [LEE & AMBROS, 2001]. It

would be possible that miRNAs could regulate any imaginable biological

process involving RNA/RNA or RNA/protein interactions [AMBROS, 2001]. And

the evidence is mounting, that animal miRNAs are more numerous and that

their regulation functions are more present than was suspected in the first place

[AMBROS, 2004]. Actually it might be that miRNAs, rather than siRNAs, are the

true intermediates of the small-mediated regulation of gene expression

[GROSSHANS & SLACK, 2002].

The first miRNA showing a function other than control of developmental timing

was found in Drosophila m., called bantam. Bantam is involved in the regulation

of apoptosis and cell proliferation in the developing fly [AMBROS et al., 2003]. A

lot of research was done and miRNAs are now successfully associated with

tissue specifications, cell death, fat metabolism, and cell proliferation in flies,

neuronal patterning in nematodes, modulation in hematopoietic lineage

differentiation in mammals, and control of leaf and flower development in plants

[LAGOS-QUINTANA et al., 2001] [BARTEL, 2004]. Evidence was found that

Page 48: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

28

deregulations of miRNAs are associated with human diseases such as cancer

[DYKXHOORN & LIEBERMAN, 2005].

MicroRNAs are produced from ~70 nucleotide endogenous hairpin stem-loop

precursor and are further processed by dicer to their single-stranded mature

form of ~22 nucleotides [DYKXHOORN, et al., 2003]. MiRNAs recognize their

targets by imperfect base-pairing (50-85%) to the 3’ UTR (untranslated region)

region of the target mRNA, causing repression of the translation and inhibition

of the protein synthesis [WADHWA et al., 2004]. As noted above, dicer does

also generate microRNAs. So it is not a surprise that they appear to have the

same length as siRNAs, are just a little tighter in their distribution, and 20-24

nucleotides in length. Also the characteristic structures of dicer cleavage

products, the 5’phosphate group and two-nucleotide 3’ overhangs were not

missing [LIM et al., 2003].

It is important to mention that there is one main difference between miRNAs in

plants and animals. In plants, miRNAs function as natural endogenous siRNAs.

Not like in animals, where miRNAs partially bind to the 3’ UTR of the target and

inhibit the mRNA translation, miRNAs in plants work like siRNAs and trigger

degradation of their targets. This results from the fact that plant miRNAs are

mostly perfectly complementary to their target [NOVINA & SHARP, 2004].

Only little is known about the exact mechanism of miRNAs, and this leaves a

broad range for speculations and suggestions [HE & HANNON, 2004]. It is still

unclear if every miRNA has one, few, or many targets, or if one target might be

regulated by multiple miRNAs [MOSS, 2002]. It was assumed, that miRNAs

targeting a single sequence, had the opportunity to undergo evolutionary

determined modifications more easily compared to conserved miRNAs, which

may act on more than one target sequence [LAGOS-QUINTANA et al., 2001].

In fact, it was discovered that single miRNA species can bind to different mRNA

targets, and multiple binding sites implicated the possibility that more than one

miRNA attacks the same target sequence to achieve translational inhibition

[DYKXHOORN & LIEBERMAN, 2005].

Page 49: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

29

Still one of the major hurdles is the prediction of miRNA targets in animals. It is

more difficult because of their imperfect base-pairing. It is necessary at this

point, to mention, that RNA cleavage by miRNA could be overrepresented, for

the reason, that their targets can be identified easier. MiRNA databases,

containing all up to date information of newly published miRNAs, are

established. But there is still a lot of research to do, to clarify all the unanswered

questions [HE & HANNON, 2004]. Nowadays, synthetic miRNAs can be

produced to mimic siRNAs or miRNAs and target special genes for silencing

[McMANUS et al., 2002].

3.4.3.2.1 Biogenesis of miRNAs

It is confirmed that miRNAs origin rather from intronic regions and from

sequences between protein-coding genes than, like predicted, from genes itself

[MOSS, 2002]. Many seem to be regulated by their own promoters, but some

use the promoter from the introns, which would be a very smart mechanism to

express miRNA and a protein at the same time. Some miRNAs occur in

clustered regions in the genome, which are sometimes, but not always, related

to each other [BARTEL, 2004]. Many miRNAs are continually expressed, and

others appear only at specific developmental stages or in certain tissues

[AMBROS, 2001]. The amount from a few hundreds to 50,000 molecules per

cell is not very predictable [BARTEL, 2004].

The generation of miRNAs requires two different steps in two different parts of

the cell. MiRNAs were transcribed by Pol II in the nucleus as long transcripts

called pri-miRNAs (primary microRNAs). The first cleavage step to generate the

mature miRNA starts afterwards, also in the nucleus [LEE et al., 2002]. Drosha,

an RNase III enzyme, containing proline-rich regions and an arginine-serine-rich

domain, two RNase III domains, dsRNA binding domain, and an amino-terminal

with unknown function, cleaves pri-miRNA into pre-miRNA hairpins

[MURCHISON & HANNON, 2004] [HE & HANNON, 2004]. Pre-miRNAs, also

known as miRNA precursors, are about 60 to 70 nucleotides in length including

Page 50: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

30

a stem loop structure and bulges. Drosha leaves a staggered cut and a typically

phosphate group on the 5’ terminal. This cut already characterizes one end of

the mature miRNA. The precursor is actively transported into the cytoplasm by

Ran-GTP and exportin-5, an export receptor. The second end is cleaved and

defined in the cytoplasm by RNase III enzyme dicer. After the cleavage of both

strands of the hairpin precursor a 5’phosphate group and two-nucleotides 3’

overhang are left. Dicer may have a preference for the 5’phosphate and the

dinucleotide overhang of the stem loop [BARTEL, 2004]. Dicer is important in

the maturation of miRNAs. In fact, in dicer-deficient mutants, accumulation of

pre-miRNA hairpins was shown, while mature miRNAs disappeared [LEE et al.,

2002]. The mature miRNA duplex is a fragment, similar in size like siRNA, only

without perfect complementary and single-stranded, because it derives from

one part of one arm of the former precursor loop. The double-stranded

miRNA:miRNA* composed by the mature miRNA and the complementary

strand miRNA*, does not survive very long in contrast to single-stranded

miRNA. The mature miRNA associates with a multiprotein complex called

miRNA ribonucleoprotein complex (miRNP), which might be a particular

subtype of RISC. The single-stranded miRNA gets incorporated into miRNP

while miRNA* gets degraded. Which strand gets loaded into RISC/miRNP

depends on the 5’ end. The one which is not that tightly bound, gets

incorporated. Very rarely, both get loaded into miRNP; this happens when the

duplexes have nearly the same stability at their 5’ ends [BARTEL, 2004]. After

incorporation, the miRNA guides the base-pair to the 3’ UTR region of the target

to inhibit the expression. It is still unclear, at which exact point of the protein

synthesis the translation occurs [FISCHER, 2010].

Also in the biogenesis, plants differ from animals. Actually, in plants,

researchers have not found any pre-miRNAs. This fact indicates that both

cleavage steps happen in the nucleus by drosha [BARTEL, 2004].

Page 51: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

31

Fig. 6 A model for miRNA biogenesis and the translational inhibition of the target RNA

modified from [HE & HANNON, 2004]

Page 52: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

32

3.4.3.2.2 Small temporal RNAs: lin-4 and let-7

Lin-4 and let-7 are both members of the small

temporal RNAs (stRNAs), which present a subclass

of miRNAs, but only the second mentioned one is

evolutionarily conserved. Their function lies in

controlling the developmental timing in

Caenorhabditis elegans [LEE & AMBROS, 2001]. Lin-

4 homologous were only found in C. elegans and

related species of C. elegans. That was the reason,

that, till the identification of the second stRNA, let-7, it

was believed to be a nematode-wide unique feature.

In contrast to lin-4, let-7 is conserved among various

organisms including mollusks, flies, zebrafish, mice,

and human [GROSSHANS & SLACK, 2002].

As quoted above, lin-4 and let-7 are miRNAs. That

means that those two also display all the

characteristic marks of miRNAs, such as, that they do

not encode for a protein, are transcribed as pri-

miRNA, are further processed to hairpin precursor

containing the typical bulges and stem-loops, maturate through dicer, and the

down-regulation of the target occurs without affecting the mRNA level by

degradation. Because of the surprising fact, that lin-4 encodes small RNAs

instead of proteins, lin-4 could remain so long undetected [MELLO, 2007]. It is

established that lin-4 encodes 22 nucleotide long, non-coding RNA fragments.

Lin-4 RNA is antisense complementary to multiple conserved 3’ UTRs of lin-14

and lin-28. Experiments discovered that lin-4 affects negatively the translation of

both, by basepairing to their 3’ UTRs. The down-regulation of lin-14 results in

developmental change from the first larval stage (L1) to the second stage (L2)

in the nematode C. elegans. Further, the down-regulation of lin-28 in the second

larval stage results in developmental progress into the third one (L3) [HE &

HANNON, 2004].

Fig. 7 Predicted structure oflin-4 and let-7

[BARTEL, 2004]

Page 53: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

33

Let-7 is phylogenetically conserved, encodes a 21 nucleotide long RNA, and

appears to operate in the same way as lin-4. Let-7 acts by binding to the 3’ UTR

of lin-41 and hbl-1. This results in their translational repression and further in the

transformation of the nematode from the fourth larval stage (L4) into an adult

worm [HE & HANNON, 2004].

3.4.3.3 Difference between siRNAs and miRNAs

MicroRNAs and small interfering RNAs share some evolutionary conserved

pathways, but differ in some others; they can replace each other under special

conditions, but are not identical [BARTEL, 2004].

MicroRNAs and small interfering RNAs are identical in their length (21-25

nucleotides) and display the typical 3’hydroxyl-, 5’phosphate end and the

dinucleotide overhang on 3’ termini. Both require dicer for their maturation and a

multiprotein complex (RISC / miRNP) to fulfill their destiny. MiRNAs are usually

evolutionarily conserved endogenous encoded sequences, and form precursor

hairpin transcripts with imperfect complementarity. Each hairpin molecule

generates one single-stranded miRNA processed from one arm [LIM et al.,

2003]. Typically, miRNA regulates negatively the target expression by binding

imperfectly to 3’ UTR regions at multiple sites of the mRNAs without cleavage

and degradation [LAU et al., 2001]. In contrast to miRNAs, siRNAs often

originate from double-stranded RNA molecules such as transposons and

viruses, or are rarely conserved endogenous siRNAs. Every double-stranded

RNA produces numerous double-stranded siRNAs, which are perfectly

complementary to their target sequence [LIM et al., 2003]. Noted already in one

of the previous chapters, small interfering RNAs basepair with perfect

complementarity to their target mRNA and initiate the degradation. MiRNAs and

siRNAs are interchangeable. It depends on the complementarity, if the complex

guides the degradation or the translational inhibition of the target mRNA. If

miRNA and the target are sufficiently complementary, the target is cleaved at

the same site as seen for siRNA-guided degradation. Otherwise, without

Page 54: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

34

enough complementarity, the miRNA directs a translational repression

[BARTEL, 2004].

3.4.3.4 Small nuclear RNA

This highly evolutionarily conserved group of small RNAs is classified into two

different classes, first C/D snoRNAs and second, H/ACA-box snoRNAs. The

first mentioned ones are between 70 and 120 nucleotides long and are

implicated to guide the methylation step of the target RNAs. H/ACA box

snoRNAs are longer, between 100 and 200 nucleotides in length, and are

employed to guide pseuduridulation. It seems that small nuclear RNAs are a

complex, made from two miRNA-like hairpins and further, snoRNAs themself

produce another small RNA class called sdRNA (sno-derived RNAs).

Experiments displayed that the biogenesis of sdRNA is controlled by

components of the RNAi pathway and further, that sdRNAs are implicated in the

regulation of gene expression and transcriptional silencing [TAFT et al., 2009].

Fig. 8 aǀ siRNA pathway bǀ microRNA pathway

[DYKXHOORN et al., 2003]

Page 55: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

35

3.4.3.5 Tiny non-coding RNAs

Tiny non-coding and endogenous siRNAs are similar in their biogenesis; the

difference is that tncRNAs are transcribed from non-coding genomic sequences

[LEE et al., 2006]. They occur to have the similar length like miRNAs, are alike

miRNAs in view of imperfect complementarity to their target, but lack in the

characteristic hairpin precursor sequences. For this reason they are not counted

as members of miRNAs. TncRNAs, in contrast to miRNAs, are rarely conserved

among related species [AMBROS et al., 2003]. The functions are still unknown,

but the discovery, that tncRNAs and siRNAs share some components of the

RNAi pathways like DCR-1 and RDE-4, suggests, they might be involved in the

gene silencing mechanism [LEE et al., 2006].

3.4.3.6 Small modulatory RNAs

Small modulatory RNAs are 20 nucleotides long and were identified in neural

stem cells of mice [NOVINA & SHARP, 2004]. They appear to have a role in

neural differentiation. The biogenesis and the pathway of their function remain

to be worked out [KIM, 2005].

3.4.3.7 PIWI-interacting RNAs

PIWI-interacting RNAs are referred to as piRNAs because of their interaction

with a subclass of the argonaute protein family, the PIWI-proteins [PETERS &

MEISTER, 2007]. PiRNAs are around 24-30 nucleotides long and are

generated dicer- and drosha-independently. So far only one function could be

investigated. PIWI-interacting RNAs are implicated in the transposon silencing

of flies to maintain the genome stability [HUTVÀGNER & SIMARD, 2008].

Another subclass of small RNAs was detected in C. elegans, called 21U-RNAs.

Those small RNAs contain a uridine 5’- monophosphate and a modified 3’ end.

Later it was figured out, that those 21U-RNAs count actually as piRNAs, in the

case of C. elegans. In other organisms those two are different subclasses of

small RNAs [FISCHER, 2010].

Page 56: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

36

3.4.4 RNA induced silencing complex and microRNA ribonucleoprotein complex

RNA induced silencing complex and microRNA ribonucleoprotein complex are

the effector enzymes of the RNA Interference phenomenon. Both are

multiprotein complexes with the difference that RISC incorporates siRNA and

miRNA enters miRNP. The complexes vary in size and composition, and have

several forms, which indicate already their different functions. Both have the

ability to cleave the target mRNA through one of the argonaute proteins, which

works as nuclease [MEISTER & TUSCHL, 2004].

Some parts of the RISC complex have been identified, but still, some parts

remain to be discovered. The first piece, described in Drosophila, was

argonaute 2 (AGO2) [BERNSTEIN et al., 2001]. AGO2 is a member of the big

argonaute family. It is highly conserved and a small (~130 kDA), basic protein,

homologous to RDE1 in C. elegans, QDE-2 in Neurospora c., and AGO1 in

Arabidopsis t., containing a PAZ and PIWI domain (called PPD domains)

[AGRAWAL et al., 2003]. All argonaute proteins possess the two characteristic

domains, a piwi-argonaute-zwille (PAZ) and a PIWI domain. Only recently a

third functional domain was found, the MID-domain. It seems that the MID-

domain, after the recognition of the 5’phosphate of small RNAs, binds them

tightly to the argonaute protein [PETERS & MEISTER, 2007]. Early studies

proved the suggestions, that PAZ might interact as domain between dicer and

AGO. PAZ identifies the terminal of the basepaired duplex and the

characteristic two-nucleotide 3’ overhangs [MEISTER & TUSCHL, 2004].

Recent experiments implicated argonaute proteins in diverse functions like

embryonic development, cell differentiation, regulation of protein synthesis,

maintaining of the genome stability, in translational repression, and are involved

as co-factor in the methylation for heterochromatic silencing [PETERS &

MEISTER, 2007] [HUTVÀGNER & SIMARD, 2008]. Argonaute proteins might

also play their part in the development of some certain diseases like cancer or

autoimmune diseases [PETERS & MEISTER, 2007]. Some organisms, which

have lost their entire family of argonaute genes, are not able to respond with an

RNAi effect at all [HUTVÀGNER & SIMARD, 2008]. Next to AGO2, additional

Page 57: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

37

proteins and factors are identified for RISC: two RNA-binding proteins, vasa

intronic gene (VIG) and dFMR (homologous to the human fragile X mental

retardation protein), and a minimum of one protein with nuclease function

(Tudor-SN) activity [AGRAWAL et al., 2003] [KENT & MacMILLAN, 2004].

Recently the long-expected slicer activity, responsible for cleaving the mRNA,

was discovered as an attribute of the human AGO2 [KIM, 2005]. In the human

model two proteins from the argonaute family eIF2C1 and eIF2C2, as well as

Gemin3 and Gemin4 were identified as parts of the complex [CERUTTI, 2003].

The phosphate group on the 5’ end of siRNA is required to access RISC.

Introduction of synthetic siRNAs without phosphorylated 5’ ends were

immediately phosphorylated by an endogenous kinase [MARTINEZ et al.,

2002]. Also other critics are implicated in the incorporation like relative and

absolute stability. Functional asymmetry means, that the strand with the lower

thermodynamics (is less tightly bound to its complementary) enters the RISC

and the other strand gets degraded [SCHWARZ et al., 2003]. In general, 5’

terminus, starting with an A-U basepair, is preferred. Also energy differences

can favor one strand [SCHEPERS, 2005]. Naturally, the antisense strand is the

one which gets incorporated into RISC. This knowledge is very handy for the

design of synthetic siRNA. It got predictable which strand will enter the RISC

complex and guides the degradation [SCHWARZ et al., 2003]. The degree of

the degradation depends on the stability of the target RNA. It is shown, that

stable RNAs are to a greater extent degraded than RNAs which are quickly

synthesized and degraded in the organism [FIRE, 1999].

RISC is generated as precursor molecule and gets activated by ATP to form

RISC* [HANNON, 2002]. The early suggestions proved to be right, which

placed an RNA helicase of the DEAH-box helicase super family as the

mechanism to unwind the duplex siRNA [SHARP, 2001]. If the ATP requirement

is necessary to change the RISCs conformation / composition or if the ATP-

requirement is needed for the destabilization of the double-stranded RNA was

long not known [MEISTER & TUSCHL, 2004]. Later it was figured out that the

cleavage step itself does not necessarily require ATP, but in presence

Page 58: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

38

of ATP the reaction is

more efficient, and Mg²+ is

required [MONOHARAN,

2004]. While the antisense

strand enters RISC to

guide the complex to the

target RNA and induce the

degradation step, the

sense strand is left behind

for degradation by

exonuclease activity

[DYKXHOORN et al.,

2003] [WADHWA et al.,

2004]. It depends on the

rate of complementarity if

the silencing is achieved through mRNA degradation or through translational

repression [LEUNG & WHITTAKER, 2005]. It was detected that cleavage of the

target mRNA appeared only in regions of complementarity with the introduced

dsRNA/siRNA after 21-23 nucleotide intervals. The cleavage site, relative to the

5’ end of the guide-strand, is located next to the center of the homologous

target, between nucleotide 11 and 12, downstream [ELBASHIR et al., 2001b].

Incorporated siRNA into RISC or miRNA into miRNP, those enzymes can direct

multiple rounds of target cleavage and give an explanation for the high

efficiency of RNAi. The stability might be the limitation of the reaction. Eri-1 is

suggested to hide the siRNAs from the RISC complex by cleaving off their 3’

overhang [MURCHISON & HANNON, 2004]. Some ideas place RdRPs after the

cleavage, to convert parts of the mRNA to duplex forms. These newly

generated duplexes might reenter the RNAi cycle from the beginning cleavage

step through dicer [AGRAWAL et al., 2003].

To explain the differences of the RNAi pathway regarding the heterogeneity of

RISC, it is important to mention, that many organisms have more than one AGO

protein; for example C. elegans possess 20, humans 8. And not to forget that

Fig. 9 Target recognition and target cleavage by the RISCcomplex

modified from [NYKÄNEN et al., 2001]

Page 59: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

39

the most organisms also have more than one different dicer protein [MEISTER

& TUSCHL, 2004].

3.5 Regulation of RNAi

Till now, only little is known about the regulation of the RNA interference

pathway. Recently, a small molecule, enoxacin, was identified. It is a non-toxic

molecule which was not seen to trigger non-specific effects, and further seems

that its enhancing characteristics work universal. Results indicate that enoxacin

improves the efficiency of the knockdown effect in vivo, by reduction of the

required dosage. The effect was not only displayed on siRNA-mediated RNAi,

also miRNAs seem to be effected. According to the experiment, levels of pri-

miRNAs and pre-miRNAs decrease while the level of the mature miRNAs

increase [SHAN et al., 2008]. The, in the previous chapter mentioned, gene eri-

1 might also play its part in the regulation of RNAi. It seems to stop the usage of

siRNAs for multiple rounds by cleaving off the two-nucleotide 3’ overhang. After

this cleavage step, siRNAs appear to be invisible for the RISC and cannot get

incorporated anymore [MURCHISON & HANNON, 2004]. Another part of the

negative regulation of RNAi might be the gene lin-35, which was found only

recently in C. elegans [FISCHER, 2010].

3.6 Difficulties for RNA interference in mammals

RNA interference, triggered by introduction of long dsRNA (~500bp), brought a

lot of information about gene functions in various organisms. It started with C.

elegans, further in Drosophila melanogaster. It seemed that it worked perfect in

all invertebrates, but in mammals the mechanism failed [LEUNG &

WHITTAKER, 2005]. These first problems, mammalian organisms were not

amenable for RNAi, indicated different, not identical pathways between

mammals and lower organisms [MILHAVET et al., 2003]. The major problem

was the activation of one arm of the innate immune pathway, including a non-

specific interferon response, which is triggered by introduction of long double-

Page 60: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

40

stranded RNA in mammalian somatic cells [HANNON, 2002] [HANNON &

ROSSI, 2004].

Interferons, signaling molecules, are secreted naturally in cells, after the cells

get infected by RNA-viruses or other overrepresented dsRNA [TUSCHL, 2001].

The dsRNA-dependent protein kinase gets activated by -interferon

Fig. 10 Activation of the PKR response and the 2‘-5‘ oligoadenylate synthase pathway

[DYKXHOORN & LIEBERMAN, 2005]

Page 61: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

41

[SCHEPERS, 2005]. As consequence, PKR inactivates the translation factor

eIF2α by phosphorylation. This pathway results in a general shutdown of protein

synthesis, and in an apoptotic and non-apoptotic cell death. A second pathway

gets activated too [CAPLEN et al., 2001]. Double-stranded RNAs, longer than

30 nucleotides, induces, next to the activation of the PKR pathway, the

production of 2’-5’ oligoadenylate synthase. Which further results in the

activation of a sequence-nonspecific RNase (RNase L) [DYKXHOORN et al.,

2003]. RNase L cleaves single-stranded RNA in a non-specifically way, that

leads to a general degradation of the mRNA and to general shutdown of the

protein synthesis of the infected cell [WADHWA et al., 2004].

The stimulation of the interferon response by dsRNA is length-dependent.

Double-stranded RNAs longer than 11 basepairs can already interact with PKR,

but double-stranded RNAs less than 30 nucleotides cannot trigger the full PKR

response [SCHLEE et al., 2006]. For the full PKR activation, at least dsRNAs

longer than 80 nucleotides are required [CAPLEN et al., 2001]. In 2001, Tuschl

and co-workers found in Drosophila embryo lysates, that chemical synthesized

siRNAs can bypass this reaction. This method was further processed, so that

an interference response in cultured mammalian cells was successfully

displayed [ELBASHIR et al, 2001a]. Those synthesized small interfering RNAs

mimic the natural dicer product, including all the siRNA characteristic structures,

5’phosphate and 3’hydroxyl group, and the two-nucleotide 3’ overhangs. With a

length of 21 nucleotides they are too short to trigger the non-specific response,

but are long enough to activate the full RNAi mechanism [HANNON, 2002]. This

newly discovered possibility leads to a widespread use to study gene functions

in general and in particular genes, which are known to be involved in inheritable

diseases [DYKXHOORN et al., 2003].

There are still drawbacks to deal with. First is to mention, the transient

characteristics of the silencing effect. The ability of amplification and spreading,

like in plants and C. elegans, is not emerged in mammals, because mammals

lack the RdRP enzymes [HANNON, 2002]. This means that the RNAi effect is

restricted to four to eight cell divides and abolishes afterwards, because of the

Page 62: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

42

dilution of the siRNAs through cell division. Afterwards, the cells recover and

cannot display any RNAi effect anymore [WADHWA et al., 2004]. It is possible

to achieve a silencing effect up to weeks, in terminally differentiated and non-

dividing cells such as neurons [DYKXHOORN & LIEBERMAN, 2005]. Second, it

is important to keep in mind, that chemically synthesized siRNAs are not able to

perform a complete “knock-out” of the target. Instead it is always a “knock-

down” technique [SHI, 2003]. Third, a big challenge is still the transfection of the

synthesized siRNAs. Further information about synthetic siRNAs is given in

chapter 3.8.1., RNA interference through chemically synthesized siRNAs.

Fourth, and possibly the biggest problem is, as recent experiments displayed,

that also synthetic generated siRNAs as well as DNA vectors expressing

shRNAs are not short enough to bypass the immunorecognition. Both are able

to induce the expression of parts of the interferon pathway in some cell types

[HANNON & ROSSI, 2004]. It was shown that the levels of, for example, type I

interferon, interleukin 8, tumor necrosis factor-alpha, were increased. So, the

results have to be interpreted very carefully [AULNER & JAGLA, 2008].

3.7 Structural design and chemical modifications

RNAi is a phenomenon based on the homology between the trigger and the

target. For that reason, the sequence of the trigger must be selected carefully to

avoid cross-interferences and off-target effects. The critical cut-off point for

cross interference is about 80% of similarity [BOSHER & LABOUESSE, 2000].

3.7.1 Structural information for the design of double-stranded RNAs

It was already mentioned that neither the sense, nor the antisense strand alone

had silencing effects, and a duplex structure is required [ZAMORE et al., 2000].

Important is, that the dsRNA includes a region of at least 88% of

complementarity between the target and the trigger to induce the silencing

effect [TIJSTERMAN et al., 2002]. The length is one of the key factors. Double-

stranded RNAs smaller than 49bp are ineffective, slightly active about 149bp,

Page 63: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

43

and efficient with a length of 505bp and 997bp [TUSCHL et al., 1999]. Usually,

dsRNA greater than 500bp are used [BOSHER & LABOUESSE, 2000]. Longer

dsRNAs are more efficiently cleaved by dicer. This results in a larger amount of

siRNAs, then shorter dsRNAs. Another fact to consider are modifications, either

at the backbone or on the bases of the RNA. In general, modifications of the

antisense strand of the duplex decrease the speed of RNAi more efficient than

modifications of the sense strand [ZAMORE, 2001]. Also, promoter sequences

and intronic sequences should be avoided, because they cannot trigger an

effective silencing response [HANNON, 2002].

3.7.2 Parameters for the design of small interfering RNAs

It is important to say, that the two main critics for the design of siRNAs are, that

the functionality and the specificity gets maximized [BOESE et al., 2005]. There

are some key reasons, why the design of siRNAs and transporter molecules,

emerged as such an important field of research. First, the improvement of the

stability and the thermodynamic characteristics of the siRNAs are important for

therapeutic applications. Small interfering RNA duplexes are quite stable and do

not need chemical modifications to extend the half-life in cell-cultures. For

applications in vivo, the picture changed. It is necessary to increase the half-life

of siRNA duplexes and their thermal stability to escape the degradation of

nuclease digestion [PAROO & COREY, 2004]. For example, through backbone

modifications of the siRNA, they were able to stand against the serum

nucleases and the half-life of the siRNAs got increased [HANNON & ROSSI,

2004].

Second, the biodistribution and the pharmacokinetic qualities have to be

enhanced. After injection, siRNAs are detectable up to 72 hours and primarily

accumulation appears in the liver and the kidneys [PAROO & COREY, 2004].

During cell-divisions, the originally injected siRNAs get diluted and the RNAi

effect abolishes quickly [LEUNG & WHITTAKER, 2005]. But through chemically

modification it was possible to induce the silencing effect from around 5 days up

Page 64: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

44

to several weeks [DYKXHOORN & LIEBERMAN, 2005]. The route of delivery

has to be optimized and the potential to use siRNAs as therapeutic agents has

to be maximized [PAROO & COREY, 2004].

Third, the affinity to the target must be increased with the help of chemical

modifications to avoid off-target effects [PAROO & COREY, 2004].

Fourth, in the future, for the introduction of siRNA as a novel and functional

therapeutic molecule, chemical modifications will be required. Ideally,

modifications improve the resistance against cellular destruction and increase

the cellular uptake [HADWIGER & VORNLOCHER, 2005].

Some limitations should be considered. For example, cells like neurons and

muscle cells are more difficult to transfect in contrast to stem cells, fibroblasts,

and tumor cells [MILHAVET et al., 2003]. Another disadvantage, it is a time

consuming process and the costs are astronomically [BOESE et al., 2005]. One

of the main challenges is to provide an enhanced ability of the silencing effect

regarding higher efficiency, specificity, and potency at which less chemical

modifications stands for less cell toxicity [PAROO & COREY, 2004] [JACKSON

& LINSLEY, 2004]. And a reduce amount of siRNA stands for a decrease

chance for off-target effects [HANNON & ROSSI, 2004]. Also the transportation

into the target tissue is still a huge challenge as well [PAROO & COREY, 2004].

But in the best case scenario, the ideal siRNA is absolutely specific and down-

regulates only the target gene of interest without any side effects, works in low

dose, and is very effective [JACKSON & LINSLEY, 2004].

The natural characteristics and required structure for siRNAs includes the two-

nucleotide 3’ end overhangs on both sides, the hydroxyl group on the 3’ end,

and the phosphorylated 5’ end. In fact, RNA duplexes with two-nucleotides

overhang are more efficient in mRNA degradation than the similar ones with

blunt-ends, or with a four nucleotide overhang [ELBASHIR et al., 2001b]. In

general, the 5’phosphorylated antisense RNA, about 22 to 40 nucleotides in

length, activates the gene silencing phenomenon. For that reason, modifications

on the 5’ end of the antisense strand are not tolerated at all, several mutations

Page 65: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

45

of the antisense strand resulted in a non-RNAi response, in contrast to

modifications on both sides of the sense strand and the 3’ end of the antisense

siRNA [MARTINEZ et al., 2002].

So, it is already obvious, that for a siRNA approach some important facts have

to be considered, including the selected siRNA sequence of the target gene,

usage of either chemically synthesized siRNAs or construction of plasmids

transporting DNA sequences, optimized transfection methods for the used

approach, cell culture conditions, type and amount of transfection agent, quality

and amount of siRNA, length of the exposition time of siRNA on to the cells, and

monitoring/controlling of the silencing efficiency with both, a positive and a

negative control [AGRAWAL et al., 2003] [MILHAVET et al., 2003]. Parameters,

especially for the design of the siRNA, are the length, the secondary structure,

the sugar backbone, and the sequence specificity. Some basic rules emerged,

like, in general the G/C concentration should be kept between 30 and 70%, the

sequence of one strand should be AA(N19)TT, the 2nucleotide 3’ overhang of

uridine should not be missing, the length of the siRNA should be 21 nucleotides,

and the two characteristic groups, the 5’phospate, and the 3’hydroxyl, must be

present [AGRAWAL et al., 2003]. A lower thermodynamic stability between the

last bases counted from the 5’ end, of the antisense strand, as well as a lower

thermostability in the middle of the siRNA (9-14 basepairs) are recommended,

because this enhances the incorporation into RISC [DYKXHOORN &

LIEBERMAN, 2005]. A/U basepairs have less binding energy, compared to G/C

bindings, which leads to the recommendation of the two uridine overhang at the

5’ end. For some positions, favorable basepairs emerged, like for position 1:

C/G, 3: G/C, 10: A/U, 19: A/U, at position 11: A, G, or C increase the chance for

a functional effective siRNA. Mismatches should not occur, but in general,

mismatches at the end of the siRNA have more influence on the RNAi activity

than mismatches located in the center [JAGLA & AULNER, 2008]. The

importance of the sequence position can be seen in the ratio of sense/antisense

strands, and which of those two actually enters the RISC complex [MEISTER &

TUSCHL, 2004].

Page 66: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

46

One important point is to identify the mechanisms of immunorecognition of RNA

regarding to avoid the design of immunostimulatory RNAs; or for the future, to

design potent, useful immunostimulatory RNAs with both functions included,

silencing and immunostimulation. The novel drug might be effective for disease

such as viral infections and cancer [SCHLEE et al., 2006]. Characteristics for

isRNAs are 3’ blunt-ends and rich sequences of GU [SHAN, 2010]. And it is

never a mistake to check, if there are already tested siRNA for the intended

target gene [JAGLA & AULNER, 2008].

Fig. 11 The design of an effective siRNA

[LEUNG & WHITTAKER, 2005]

Page 67: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

47

3.8 Special delivery methods

Two different possibilities exist to induce the intended silencing effect, either

through synthetic siRNAs or through short hairpin RNA (shRNA) vectors. The

first possibility is used for short-term knockdown, and normally applied directly

into the cytoplasm. The second possibility provides the option to generate

stable cell lines, and it is able to extend the knockdown duration. ShRNAs are

introduced into the cell via a vehicle, either a plasmid or a viral vector. The

chosen vector starts to express shRNAs which are further processed into

siRNAs through the enzyme dicer [SHAN, 2010].

In the end, it is to say, that there never will be the one and only method. All

methods have their advantages and limitations, and it always depends on the

approach which method is the best-suited one. Also the shRNA-mediated gene

silencing and the siRNA-mediated gene silencing are not competing. They

should be seen as a complement to each other [PADDISON et al., 2002]. But to

summarize all the advantages and disadvantages of vector expressed siRNAs,

synthetic siRNAs still represent the golden standard reagent with the broadest

applicability [CLAYTON, 2004].

3.8.1 RNA interference through chemically synthesized siRNAs

With the introduction of synthetic siRNAs, the whole application system of

siRNAs into the RNAi pathway got revolutionized. The synthetic siRNAs,

between 21 and 22 nucleotides in length, with the typical characteristics of the

natural siRNAs, are able to initiate the degradation step without any need of the

typical cleavage step through the enzyme dicer [ELBASHIR et al., 2001b]. The

small interfering RNAs are able to incorporate directly into the RISC complex by

imitating natural siRNAs and guide the target degradation step [ZAMORE,

2001]. A positive effect was the discovery that synthetic siRNAs function as well

in mammalian cells, by bypassing the dsRNA-dependent immunity response

[ELBASHIR et al., 2001b]. But some drawbacks still remain. First, the later

mentioned, transient attribute after the transfection into the cell does not allow

Page 68: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

48

long-term approaches; second, the requirement for chemical or enzymatic

synthesis of siRNAs before the application into the cells; third, the

astronomically high cost of the chemical production of the synthetic siRNAs;

and fourth, difficulties in the transfection of siRNAs into certain cell types

[TUSCHL, 2002] [AGRAWAL et al., 2003] [KIM, 2005]. But nevertheless,

synthetic siRNAs emerged to the method of choice for investigating gene

function in cultured mammalian cells [PADDISON et al., 2002]. The huge

possibility of efficient homologous degradation of the target gene, guided by

those synthetic siRNAs, leads to high throughout approaches in mammals for

genome wide analysis of gene functions in general, and also regarding

modulation of gene expression in human diseases [CHI et al., 2003].

Like already mentioned, the chemical synthesis of siRNAs is a very expensive

effort with the disadvantage, that there is no guarantee that the first produced

siRNA will work at all, or with the expected efficiency and specificity [SOHAIL &

DORAN, 2005]. Normally instead of only one, several oligonucleotides need to

be investigated first, to check which one works with the highest efficiency and

specificity, before the target gene is successfully silenced [MYERS & FERREL,

2005]. Naked siRNA can be applied to some tissues and organs such as eye,

lung, and CNS through injection, instillation, or infusion and get integrated into

the tissue by an unknown mechanism. But other cell types are not able to

incorporate the naked siRNAs, so that other transfection methods had to be

figured out [SHAN, 2010]. One of the most used and very effective transfer

methods for short-term silencing is the incorporation of siRNAs into liposomes

[MILHAVET et al., 2003]. Liposomes are coated by a bilayer of phospholipids,

which fuse with the cell membrane and deliver the integrated molecules into the

cytoplasm [SHAN, 2010]. Other methods like electroporation, microinjection,

and hydrodynamic shock, also covalent binding to hydrophobic carrier

molecules or covalent coupling to cell penetrating peptides, and bombardment

with nucleic acid coated gold particles have evolved [MILHAVET et al., 2003]

[WADHWA et al., 2004] [LU & WOODLE, 2005].

Page 69: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

49

To lower the production costs and to overcome the transient problematic, many

research centers developed alternative strategies to extend the period of

efficient gene silencing and to establish the production of stable loss-of-function

RNAi [WADHWA et al., 2004]. Possibilities to achieve the longer and stable

gene silencing, is by designing DNA templates to express siRNAs

intracellularly. As delivery agent, plasmids and viral vectors, like retroviral

vectors and lentiviral vectors are often used, to carry short hairpin RNA

(shRNA) into the cell [KIM, 2005]. Libraries for siRNA and shRNA-containing

vectors, which cover entire genomes, are already developed [EIFERT et al.,

2008]. Also diverse chemically synthesized siRNAs are already available from

several companies [LATHAM et al., 2005].

For in vitro experiments, bacterial or viral enzymes are used [SOHAIL &

DORAN, 2005]. For example, purified recombinant dicer enzymes or bacterial

RNase III enzymes can be employed to cleave large amounts of in vitro

transcribed dsRNA. The cleavage results in a huge pool of so-called diced

siRNAs (d-siRNAs) or endonuclease-generated short interfering RNAs

Fig. 12 Strategies to deliver the silencing signal into the organism

[LEUNG & WHITTAKER, 2005]

Page 70: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

50

(esiRNAs) [RAMOS-NINO & MOSSMAN, 2005]. One disadvantage of using

those pools of RNAs is that the chance for off-target effects might be increased,

because the chance that a mixture of a variety of siRNAs matches to more off-

targets is higher than for copies of a single siRNA [MYERS & FERREL, 2005].

In the beginning, it was assumed, based on facts, that synthetic siRNAs can

easily bypass the immunity response, that the gene silencing effects are highly

gene-specific, and no side-effects, secondary effects were actually detected

[CHI et al., 2003]. Unfortunately the first assumption proved to be wrong.

Chemical synthesized siRNAs are able to induce parts of the interferon pathway

[MITTAL, 2004].

3.8.2 Short-hairpin RNA-mediated gene silencing

One possibility of gene silencing has already been discussed. The applications

of synthetic siRNAs, which act like products of dicer, enter directly the RISC

complex to guide the degradation step of the target RNA. One important

disadvantage is the transient attribute of the synthetic siRNA induced RNAi. To

bypass that effect, an alternative strategy was developed [PADDISON &

HANNON, 2002]. Short-hairpin RNAs are composed to down-regulate in a

sequence-specific way the expression of the target gene. Two possible ways

exist to induce shRNAs silencing, either by transfection of exogenously

synthesized shRNAs or directly by in vivo transcription from RNA polymerase II

or III promoters. Through the availability of endogenous expression of shRNAs,

the door is now wide open for production of cell lines, in which RNA interference

is used to stably suppress the gene expression [PADDISON et al., 2002]. A

continuous and heritable silencing effect can be achieved, without fading away

after a few cell divisions [MITTAL, 2004]. So the big advantage compared to

chemical synthesized siRNAs is the continuous and stable transcription in cells,

cultures, or in vivo [MILHAVET et al., 2003].

Short-hairpin RNAs adapted as effective method to trigger gene silencing in

diverse organisms as plants, C. elegans, Drosophila m., and mammalian cell

Page 71: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

51

lines such as HeLa cells, monkey epithelial cells, and murine fibroblasts. It

seems so far, that shRNAs can also escape the immunity response of

mammalian cells. That leads to the possibility to use shRNAs as tool for stable

gene suppression in mammalian cells [PADDISON et al., 2002]. There are two

different classes of shRNAs. Class number I represents the hairpin RNAs

designed to work like siRNAs. The second class includes the members of

shRNAs, which are modeled after miRNAs [McMANUS et al., 2002].

MicroRNAs are encoded by hairpin precursors and are further processed by

dicer into their mature forms of ~21 nucleotides in length. There, typical natural

characters are the imperfect base pairing, the hairpin structures including

several bulges, and the fold-back stem-loop of around 30 nucleotides.

Interestingly, shRNAs, which are designed to contain a simple hairpin structure

with perfect homology to the target, are the most potent inhibitors [PADDISON

et al., 2002].

In general, a few recommendations for the design of the shRNAs construct

emerged. The duplex should contain between 19 and 30 nucleotides for the

hairpin construct, because longer ones are often more effective than shorter

ones. It is important to keep an eye on the orientation of the guide and the

passenger strand [JAGLA & AULNER, 2008]. Constructs designed with Pol III

promoter are usually designed to express first the sense strand of the siRNA,

and then a short spacer, followed by the antisense strand of the siRNAs, and

then terminated by a 5-6 thymidines sequence as transcription stop. The

opposite arrangement of sense and antisense strand does not interfere with the

potency of the transcribed shRNAs. The long use of Pol III promoters, such as

U6 and H1, results from the facts that Pol III promoters contain a transcription

start point and a termination point. Additional is the fact that the construct

encode already the required uridine overhang, which is perfect for the newly

synthesized siRNAs and the further process. But nowadays, Pol II promoters

are recommended. Pol II promoters produce a longer and stable shRNA

production in the cells [LATHAM et al., 2005]. With the change to Pol II

promoters, the potency of the silencing effects was increased sufficiently

[AULNER & JAGLA, 2008]. Especially in cells with a weak or even not visible

Page 72: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

52

answer, Pol II promoters are the right choice to display phenotypes

[DYKXHOORN et al., 2003]. The length of the loop (tested 4 to 23 nucleotides)

does not interfere with the degree of the suppression. Experiments displayed

that the silencing effect depends on the correct and stable integration of shRNA

regarding the orientation into the genome [PADDISON et al., 2002]. The

transfection of the construct can be achieved in different ways. All standard

delivery methods of conventional transfection and several viral vectors, ranging

from retroviruses to adenoviruses, can be used. The expression of the shRNA

in vivo is either driven by constitutive or inducible promoter system [HANNON &

ROSSI, 2004].

Like for every method, some advantages and limitations occur. ShRNA vectors

are a good choice for cells which are hard to transfect or cells which cannot

survive chemical modification [SHAN, 2010]. For very stable proteins or for

approaches which require a continuously knock-down of the expression, this

method is the recommended one [QUERFELLI, 2008]. In contrast to

synthesized siRNAs, by using shRNAs the ability to control the shRNA

expression is given [AULNER & JAGLA, 2008]. On the other hand, the level of

gene silencing mediated by shRNAs is variable and less reproducible, and does

not allow any control of the achieved dose for the silencing effect inside the cell,

compared to siRNAs, where the dose to achieve an effective knockdown can be

controlled [CLAYTON, 2004] [QUERFELLI, 2008]. Limitations are also in

studies of genes essential for cell survival, cell-cycle regulation, and

development. The stable down-regulation over extended periods can result in

non-physiological, compensatory responses, which can mask the true biological

consequence [MITTAL, 2004]. The previous work, before the actual experiment

can be started, is a lot greater than using siRNAs. First, the DNA fragment of

the target has to be sequenced and afterwards cloned, to produce the

construct. Second, the design of the shRNA must be ruled out carefully to result

in an effective and sensitive construct [HANNON & ROSSI, 2004]. But with the

development of variety libraries for siRNAs and shRNAs, this part of the

experimental approaches got a lot easier [AULNER & JAGLA, 2008].

Page 73: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

53

3.8.3 Transfection agents

Since 2002, it is possible to transport siRNAs into various cells via DNA vectors,

either with plasmid vectors or viral vectors [SHI, 2003]. Plasmid-based delivery

agents express shRNAs directed by RNA polymerase III, such as U6, H1,

snRNA, or tRNA promoters, or RNA polymerase II promoters [WADHWA et al.,

2004]. The perfectly homologous stem loop of the shRNA to the target mRNA is

further processed by dicer into siRNA. Those siRNAs are then integrated into

RISC to guide the degradation of mRNAs. The cost, compared to chemically

synthesized siRNAs, is lower for plasmid-based RNAi [MITTAL, 2004]. As

mentioned earlier, the transient effect in mammalian cells results from the

missing amplification system. One plus of DNA-vectors is that those transient

effects can be bypassed. As a result, a stable siRNA expression in the target

tissue is given, and this leads further to a longer and more stable gene silencing

effect [SHI, 2003].

Because of some limitations of the usage of the plasmid vehicles, such as less

potency compared with transfected siRNAs, and the difficult part of the

transfection in general, viral vectors were developed [AGRAWAL et al., 2003].

Using the viral transfection agents, an increased efficiency, regarding the

transfection of the cells, was achieved. Also a longer and even more stable

gene silencing in vivo of the targeted gene was gained with the employment of

viral vectors [MILHAVET et al., 2003]. Different kinds of viral vectors are

developed; retroviral, lentiviral, and adenoviral vectors were used to deliver

siRNAs into cells and tissues, which are difficult to transfect with other methods

[WADHWA et al., 2004]. One attribute of the adenovirus is that they do not

integrate into the host genome. This actually, makes it to a more suitable

transport system for RNAi-based gene therapy in humans [MITTAL, 2004].

They are non-enveloped DNA viruses and contain a large and complex genome

structure. They can be successfully transfected into cells, which are hardly to

transfect with standard methods. Characteristics of the retroviral-vector are the

small and simple genome structure. So it is quite easy to clone the gene of

interest into the vector. After that production step, it is fast to amplify the vector.

Retroviruses can target only dividing cells and can be integrated into the

Page 74: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

54

genome. Their characteristic, to be an enveloped virus, is valuable. It is easy to

change the target, only the envelope has to change. Retroviral-delivery agents

are best suited to build libraries [MAO et al., 2005]. Lentivirus, a subclass of the

retroviral viruses, possesses the ability to get efficiently integrated into the host

genome of non-dividing cells, like stem cells or terminally differentiated cells

[MITTAL, 2004].

A limitation turned out to be that viral-agents have to be injected locally and

cannot target individual tissues. Another option would be the sampling of some

cells and after ex vivo treatment the cells get replaced [CLAYTON, 2004].

Recently published studies indicate that it is now possible to inject the virus-

vector straightforward into the target tissue or organ [SHAN, 2010]. All the

positive effects of the viral-agents are clouded though the concerns, that the

integration of the viral structure into certain genomic locations might lead to

pathogenic gene expression or that the expression of viral proteins leads to

negative biological effects for the host organism. Altogether, non-viral vectors

are the safer choice for therapeutic treatment [LU & WOODLE, 2005].

Page 75: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

55

Fig. 13 Strategies for siRNA production in vivo and in vitro

SiRNAs can be synthesized in vitro and afterwards transfected into the cell (A,B or F), produced in vivo from DNA templates (C,D, or E), or derive from hairpin precursor (C, E). [MYERS & FERREL, 2005]

Page 76: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

56

3.9 Advantages and limitations of the RNA interference mechanism

Already in the early days of the newly discovered method, a very high potency

was expected for further research in a wide variety of organisms and different

fields [HUNTER, 1999]. Theoretically every expression of every gene can be

inhibited; the better suited term would be down-regulated [NIRENBERG, 2005]

[MAINE, 2001]. This newly discovered method may open the doors to a new

class of drugs to treat a variety of human diseases. Because of the astonishing

sequence specificity, the possibility to identify and validate new drug targets,

and design sequence-specific drugs, RNAi arose to the method of choice for

drug targeting and pathway validation [SEMIZAROV et al., 2003]. RNA

interference developed into the method of choice, to investigate gene functions,

to identify cloned genes, determine loss-of-function phenotypes of genes

without known mutations, and creation of null mutant phenotypes of specific

genes, and in vivo target validation in C. elegans and later in Drosophila

melanogaster [HUNTER, 1999] [NISHIKURA, 2001]. With the discovery that the

introduction of siRNA into mammalian cells leads to an efficient and robust

silencing effect, hardly any boundaries exist for the mechanism [PADDISON et

al., 2002]. Nowadays it is already possible to buy RNA production kits

[SCHEPERS, 2005].

RNAi provide a simpler way to study gene functions, especially regarding genes

essential for viability or genes expressed in maternal germline in C. elegans, but

RNAi does not supersede classical genetics in the nematode [BARSTEAD,

2001]. In species, which are not amenable to genetic analysis, RNAi proved

especially useful, because with RNAi the possibility emerged to knock-down the

gene expression of diverse genes [BOSHER & LABOUESSE, 2000]. One

advantage compared to classical forward genetics is, that lethal null-mutant

phenotypes, which are missing in classical forward genetics, can be identified in

RNAi screens [POULIN et al., 2004]. A very handy characteristic is that the

maternal and zygotic gene function is simultaneously inhibited [HUNTER 1999].

RNAi is a very practical tool to investigate gene function in a wide variety of

organisms. But it is important to keep in mind, that both methods, the classical

forward genetic and RNAi are not competing, they complete each other and

Page 77: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

57

none of them can supersede the other one [FRASER et al., 2000]. The ability of

RNAi, to be used for large-scale and whole-genome screens should not be

unmentioned. It can be used, for example, to investigate all genes necessary for

a certain physiological pathway. In general, a whole-genome screen can be

carried out for every biological process [LEUNG & WHITTAKER, 2005]. Large-

scale screens of loss-of-function experiments provide important information

about pathways involved in development of diseases. This information can be

further used for research and development of therapeutics, especially for those

classified targets [APPASANI, 2005].

RNA interference is an easy to use method, especially for organisms with a

complete sequenced genome. And it is an inexpensive and rapid method to

analyze gene functions in a variety of organisms [TUSCHL, 2001]

[DYKXHOORN et al., 2003]. Approaches in liquid cultures can be carried out

easily by one person [POULIN et al., 2004]. It would take months to years to

clone all the genes by classical forward genetics, responsible for all the mutant

phenotypes, while RNAi can deliver the results in about a month [SUGIMOTO,

2004]. For example, to generate a knock-down mouse, it is required to insert a

single transgene. First, a part of the gene on both chromosomes has to be

removed. Afterwards, the silenced type can be generated in just a few months

of work. In contrast to produce knockout mice, this would take a few years

[DYKXHOORN & LIEBERMAN, 2005].

One of the biggest pluses is the possibility to connect already known gene

sequences to their phenotypes/functions and the unbelievable speed of the

possibility to investigate all those new phenotypes [BARSTEAD, 2001]. There

are two different approaches to work with. First is to silence one gene after the

next, so the identification of molecular parts of specific pathways and processes

is possible [FRASER & AHRINGER, 2003]. The second one is to collect all data

of every phenotype in a systematic way [SUGIMOTO, 2004].

Page 78: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

58

RNAi is not a method without limitations. With an RNAi approach, never a

complete knock-down can be achieved. It is rather a gene knock-down than a

gene knock-out phenomenon [DYKXHOORN & LIEBERMAN, 2005]. It is a fact,

that RNAi-based screens miss some related genes, because of the drawback;

RNA interference does not inhibit all gene expressions [FRASER et al., 2000].

The diminution of the mRNA levels is required at a critical threshold to display

the resulting phenotypes. This rate depends on factors, such as the balance

between dsRNA mediated mRNA destruction and de novo mRNA synthesis, the

natural stability of the mRNA, or the dsRNA uptake and further processing, and

the availability of the RNAi machinery [TIMMONS et al., 2001]. This results in

that; RNA interference does not necessarily display always visible phenotypes

or express phenotypes with the same intensity [KUMABARA & COULSON,

2000]. And this might lead to false-negative results [MITTAL, 2004]. Also the

only way to respond to RNAi is with a loss-of-function phenotype. Compared

with other methods, like usage of mutagens, they are able to display gain-of-

function and partial loss-of-function phenotypes too [JOHNSON et al., 2009].

Another difficult part is that not all tissues answer with the same efficient

response, for example the nervous system is mainly refractory to the

phenomenon and sperms occur to be not very sensitive [KUMABARA &

COULSON, 2000]. This implicates, that genes with essential functions in

neuronal tissue are most likely missed in RNAi screens [SUGIMOTO, 2004].

One of the biggest challenges is to overcome the cross-interference between

closely related sequences or in gene families, and to avoid off-target effects in

general [KUMABARA & COULSON, 2000]. Sequences which are partly equal,

with an only trigger-target identity of 80%, can be inhibited [MAINE, 2001]. This

affects especially sequences of related genes or gene family. This results in

that, the interference effect can be triggered also in the related, not-targeted

genes [FIRE et al., 1998]. Most of the off-targets are concentration dependent,

so researchers try to achieve efficient effect with the lowest possible dose

[VANHECKE & JANITZ, 2005]. So far, the only recommendations are to work

with the lowest possible dose and to select sequences which share as little

homology with other mRNAs as possible. If off-target effects are expected,

Page 79: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

59

alternative trigger sequences of the target should be chosen [JOHNSON et al.,

2009]. Another side effect of the RNAi response in mammalian cells is the

activation of an interferon pathway. This problem has already been discussed in

chapter 3.6, difficulties for RNA interference in mammals. Also the transient

RNAi, including the generation of secondary siRNAs should not be missed at

this place. The secondary siRNAs, containing a bit different sequence

configuration, may lead to phenotypes not specific for the actual silenced gene

[CHI et al., 2003].

There are also some problems with the reliability of the results. RNAi results

from the same experiment design executed from different research groups, but

also experiments executed in the same laboratory, showed a variability of 10%

to 30%. This is explained by an extreme highly false negative rate in every

RNAi screen. And it does not matter, if the same laboratory or the same

experimental designs, including the same conditions, are used [SIMMER et al.,

2003]. Some conditions, which might influence the results, are slight differences

in the timing regarding the developmental stages of the nematode, length of

exposure to the trigger RNA, used concentrations of dsRNA, and fluctuation of

temperature during the experiment [SUGIMOTO, 2004].

An important thought is that it is unknown, how the introduced siRNAs, either

the synthetic ones or the siRNA expressing vectors, will interfere with the RNAi

machinery. RNAi emerged as a natural mechanism, and is required in so many

pathways, that it is not known what will happen if the mechanism gets

kidnapped. Till now, there is no evidence that there are any negative impacts on

the processing, regulation of the natural process regarding endogenous siRNAs

and of course miRNAs, beside the mentioned side effects [CHI et al., 2003].

Page 80: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

60

3.9.1 Off-target effects

Actually, the first suggestions for cross-silencing were reported already in the

first year of the discovery of RNAi in C. elegans [TABARA et al., 1998]. The

idea emerged that dsRNA corresponding to a gene in an “operon” may be able

to silence other co-transcribed genes too [MAINE, 2001]. But for long time,

there was no consistent off-target effect displayed by siRNA-mediated RNAi.

The synthesized siRNAs occurred to be highly sequence specific, and seemed

to target only the mRNA of interest [CHI et al., 2003]. But evidence showed that

the fear of RNAi not being perfectly specific, came true. For long time,

researchers thought that the generated siRNAs are undeniable specific. There

was no proof, that for example 17 nucleotides out of 21 from a siRNA can

trigger off-target effects [COUZIN, 2004]. But evidence displayed already off-

target effects with siRNAs containing 11 consecutive nucleotides homologous to

another mRNA [AULNER & JAGLA, 2008]. The picture changed with the ability

to induce RNAi in mammalian cells with synthesized siRNAs [COUZIN, 2004].

From this moment on, it was confirmed that siRNAs are able to guide gene

silencing without a complete corresponding sequence [QIU et al., 2005].

Introduction of siRNAs resulted not only in the expected down-regulation of the

targeted gene; instead a down-regulation of multiple genes was displayed. The

first evidence for the ability of siRNAs to “cross-react” was demonstrated.

Endogenous miRNAs, are similar in size, but are not perfect complementary to

their targets and match only partly. It was figured out, that hundreds of miRNAs

play an important role in diverse pathways of the mammalian organisms by

using some parts of the RNAi machinery. The introduced siRNAs are mistaken,

and thought to be endogenous miRNAs, which results in the unwanted off-

target effects. Off-target effects are visible under microarrays analysis, with the

huge disadvantage that microarrays display only the gene expression, but do

not say anything about the protein levels [COUZIN, 2004]. The main problem of

negative results is, that the protein levels could be, after the gene silencing, still

low, but high enough to trigger the natural function without any negative

consequences for the organism and without any phenotype displayed [FRASER

& AHRINGER, 2003]. Possibilities for negative and positive controls are at the

Page 81: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

61

mRNA levels, microarrays, northern plot, and real-time PCR and to check on

protein levels, western plot can be executed [SHAN, 2010].

Off-targets effects can be classified into sequence-specific and non-specific

effects. Knock-down or influence of the expression from a sequence-related

gene or from a gene family member is counted into sequence-specific off-target

effects [SEMIZAROV et al., 2003]. Non-specific effects are, for example, the

linking of siRNAs to cellular proteins, activation of apoptosis and stress-

response genes, activation of an interferon response, or translational silencing

through a miRNA effect [SEMIZAROV et al., 2003] [JACKSON & LINSLEY,

2004]. To explain the side effects on the level of translation, it seems that

siRNAs are wrongly recognized from the cell/organism as endogenous miRNA

[JACKSON & LINSLEY, 2004]. It also might be possible that introduced siRNAs

have such a widespread effect like changes in gene expression [SEMIZAROV

et al., 2003].

The idea arose, that the off-target effects might be concentration dependent. To

determine the critical threshold, researchers experimented with different siRNA

concentration. With concentrations of 100nM, every tested siRNA triggered a 4

to 8-fold increased activation of gene expression, including activation of

apoptosis related genes. In contrast to the experiments with concentrations of

5nM or 20nM, silencing was triggered with an equal potency compared to

100nM, but no statistically significant side effects were measured. Because of

that result it is suggested, that with the proper design of siRNA and a low-

concentration introduction-dose, no off-target effects should be triggered

[SEMIZAROV et al., 2003]. Some studies also explain the high doses as toxic

for the organism, and this toxic overdose results in the displayed off-target

effects [JACKSON & LINSLEY, 2004]. A second factor is the length of the

trigger dsRNA. It was demonstrated that siRNAs of 21 nucleotides in length are

the optimal size. The probability for side-effects, increase with the length of the

dsRNA. In general, the chance for side-effects is significantly lower for 21nt-

long siRNAs, than for shorter or longer (100-400nt) ones. Another fact to

consider regarding off-targets is the size of the genome. The length of the

Page 82: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

62

original introduced dsRNA has more severe influence in larger, more complex

genomes, than in more basic ones [QIU et al., 2005].

In the end, the unlimited use of siRNA is not unlimited at all, like hoped in the

first place [SEMIZAROV et al., 2003]. It is still not clear, how widespread those

off-target effects are, and researchers are struggling with the perfect solution to

avoid those effects [COUZIN, 2004]. The design of the synthesized siRNAs has

to be executed very carefully. The golden goal is to maximize the knock-down

effect by using the lowest possible siRNA concentration, and to minimize/avoid

all cross-reactions, and off-target effects [LEUNG & WHITTAKER, 2005]. For

interpretation of the results, it must be considered, that the displayed phenotype

could be evoked by the side effects [SEMIZAROV et al., 2003]. But it is not

necessary, that off-targets disturb the phenotype [COUZIN, 2004]. In fact, the

high potential of this pretty young discovered tool is unmatched for identification

of gene functions, that the possibilities of off-target effects must be tolerated

[JACKSON & LINSLEY, 2004].

3.10 Outlook

In 2003, the expectations for clinical trials were that they might not start until

2008, or even more likely 2013 [HOWARD, 2003]. But actually, the way for

application of siRNAs as novel drugs and genetic therapy proved to be much

longer and harder than first expected [MILHAVET et al., 2003]. With an

optimized transfection method, perfectly designed and effective

siRNAs/shRNAs, and the right amount of transfection agent, the dream of the

individual gene therapy might come true soon [AULNER & JAGLA, 2008].

In general, the siRNA-based treatments proved to be very potent in the fight

against diseases like cancer, infectious diseases, cardiovascular,

cerebrovascular, and neurodegenerative diseases, but a lot of challenges have

to be solved first [MILHAVET et al., 2003]. The delivery of siRNA into the patient

and his/her proper tissue still remains a problem. Chemical modifications and

proper siRNA design are figured out so far to avoid immune-stimulation or more

Page 83: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

63

exact, to get only an acceptable level of the immune response. Other main

causes for modifications are to safe siRNAs from the fast degradation through

nucleases and to avoid cross-effects. Another concern is that through the

hijacking of the machinery through exogenous introduced siRNAs, the natural

endogenous function might be unattended, which might result in still unknown

factors. The required dose of siRNAs still needs to get reduced, and at the

same time the effective period needs to get increased [SHAN, 2010].

At last it remains to say, that there is still much to discover and a lot of

unanswered question remain [SHAN, 2010].

3.10.1 The use of RNAi in the development of novel drugs

Already early arose the idea of using siRNA-mediated silencing as therapeutic

agent for gene therapy, and developing novel drugs, specific for individual

weaknesses. But the way of achieving this goal is long and rich of drawbacks

[TUSCHL & BORKHARDT, 2002]. The variety of treatments is nearly unlimited,

the trigger is quite easy to synthesize, and the cost for the production of the

therapeutics are amenable. With this background and the characteristics of

siRNAs themselves, they developed into a very eye-catching class of small-

molecule drugs [DYKXHOORN & LIEBERMAN, 2005]. For the future, it seems

most likely to use RNAi-based drugs to treat diverse human diseases. We just

do not know now, how long we have to wait for it [HANNON & ROSSI, 2004].

Before this will happen, some challenges have to be overcome first. As

mentioned, the method would be ideal, if it would not display side effects like

the introduction of an immune response and the off-target effects [COUZIN,

2004]. But it seems most likely, that with diverse modifications like increasing

the stability, and bioavailability as well as adjustments, this problem can be

solved. More information about the design of siRNAs is provided in the chapter

3.7.2., parameters for the design of small interfering RNAs [HANNON & ROSSI,

2004]. Another two problems are the delivery in general and the delivery of an

effective therapeutically concentration. It depends largely on the development of

Page 84: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

64

an efficient in vivo transport system for siRNAs, if the novel-drug will be

successful in the future or not [LU & WOODLE, 2005]. As already mentioned, in

high concentrations, siRNAs trigger an interferon response. So the lowest

effective concentration should be applied [DYKXHOORN & LIEBERMAN,

2005]. Some concerns include the use of viral vectors and the possible toxicity

potential of the method [HANNON & ROSSI, 2004]. Through the high sequence

specificity of siRNAs, the concerns arose, that through mutations of the

sequence, the organisms might become resistant to the treatment. This fact can

only be bypassed with the introduction of a new designed siRNA, which targets

a different site of the same target mRNA [DYKXHOORN & LIEBERMAN, 2005].

The advantage of RNAi compared to methods like antisense, ribozymes, or

aptamers is that RNAi is a natural process, developed, established, and brought

to perfection over thousands of years of evolution. It is not a method developed

in the lab, where an unknown chemically synthesized intruder enters the cell.

The opposite is the truth, nearly every cell/tissue of the organism is able to

perform this process, is familiar to all components of the pathway, and

theoretically all genes can get silenced [HOWARD, 2003].

Some of the first preclinical studies, performed with RNAi as investigation tool,

were diseases like cancer, infectious, and viral diseases like human

immunodeficiency virus (HIV) and hepatitis C (HCV) as well as cardiovascular

and cerebrovascular diseases, and neurodegenerative disorders. Other

important targets for RNAi would be autoimmune disorders, and nutrition

associated diseases like obesity and diabetes, or in general diverse oncogenes,

growth factors, and single nucleotide polymorphisms (SNP) [MILHAVET et al.,

2003] [HANNON & ROSSI, 2004]. More and more published papers summarize

their results of in vivo RNAi-mediated silencing in cancer, HIV, HCV, or acute

respiratory syndrome [LEUNG & WHITTAKER, 2005]. Actually, HIV was one of

the first viral diseases, targeted by RNAi. The inhibition of HIV-encoded RNAs

was achieved in cell cultures. But for therapeutic application there are some

challenges which have to be solved first, like the delivery, the high mutation rate

of the virus itself, and the same appearance of the toxic side effects like using

Page 85: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

65

anti-retroviral drugs as treatment. Also in HCV, a significant knock-down of 99%

of the main antigene could be achieved in liver hepatocytes. But also here, the

delivery strategies, siRNAs or shRNA vectors, remain a big challenge

[HANNON & ROSSI, 2004]. Recently, clinical trials started for treatments of

macular degenerations, diabetic retinopathy, and hepatitis C. [SHAN, 2010].

Page 86: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 87: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

67

4. RNA interference in the model organism Caenorhabditis elegans

4.1 Introduction of Caenorhabditis elegans as model organism

In nature, C. elegans, named after its elegant simplicity, lives in soil, nearly in all

different temperate parts of the world [MELLO, 2007] [HOPE, 1999 reprinted

2005]. This organism is frugal; as food source serve microorganisms like

bacteria. Next to the food, humid environment, ambient temperature, and

atmospheric oxygen are needed for development and fertility [HOPE, 1999

reprinted 2005]. The nematode C. elegans developed into a very popular model

organism after S. Brenner introduced the worm as new model in 1974. The

worm is divided into two sexual forms, males and hermaphrodites. The

hermaphrodites produce both eggs and sperms, which results in the ability of

self-fertilization. The other possibility is to mate with a male, which produce only

sperm, to produce offspring [BRENNER, 1974]. As a result after self-

fertilization, around 300 worms are produced, compared with mating with a

male nematode, which results in a more than 3-fold higher number of offspring

[BÜRGLIN et al., 1998]. It also generates males and hermaphrodites in equal

numbers. In contrast to self-fertilization of hermaphrodites where all of the

offspring exhibit the XX gender. One attractive characteristic for using C.

elegans is the automatic generation of homozygous individuals through the self-

fertilization from hermaphrodites [HOPE, 1999 reprinted 2005]. In the

configuration of the genome only one big difference appears in the gender-

chromosome. Both, male and hermaphrodite possess five autosomes and one

sex-chromosome. Males contain only one X (for males X0) and hermaphrodites

contain two X (for hermaphrodites XX) [BRENNER, 1974]. There are only small

differences between the two genders; adult males are a bit shorter and thinner

than the adult hermaphrodite [HOPE, 1999 reprinted 2005]. The anatomy of the

multicellular organism is well characterized and the numbers of cells are fixed.

The analyses are already that far, that the cell lineage is known from every

developed cell [MAINE, 2001]. An adult hermaphrodite contains 959 somatic

Page 88: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

68

cells and an adult male has 1,031 somatic cells [BÜRGLIN et al., 1998]. The

nervous system, quite simple, is composed of 302 neurons in both genders.

Another amazing thing is the transparency of the worm through all

developmental stages, which allows an uninterrupted observation of every

second of the organogenesis, and all other developmental steps of the worm

[BARGMANN, 1993]. Another nice negligibility is that there is no appreciable

smell [HOPE, 1999 reprinted 2005].

The development of the nematode is regulated through environmental

conditions. Under special environmental conditions like food shortage, extreme

temperature or high density, an alternative pathway gets started, referred to as

dauer larva. If, for example, after food shortage, enough food is available again,

the worm starts to recover and grow into the adult animal [BARGMANN, 1993].

In contrast to the standard lifespan of an adult worm of 15 to 17 days, animals

Fig. 14 Life cycle of C. elegans at 22°C

[HALL & ALTUN, 2008]

Page 89: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

69

changed into the dauer larvae stage, and can survive in this condition several

months [BÜRGLIN et al., 1998]. The big advantages of this organism of being

an amazing model organism are numerous. The life cycle takes about three and

a half days, at 20°C, three days at about 25°C, or around 6 days at 15°C to

pass all 4 larvae stages, L1-L4, and grow into an adult worm [BRENNER, 1974]

[BARGMANN, 1993] [HOPE, 1999 reprinted 2005]. After 14 hours, at 25°C the

embryogenesis is completed [BARGMANN, 1993]. To pass all 4 larvae stages,

L1-L4, and grow into an adult worm, 12h for the L1, 8h for L2, 8h for stage 3, 10

hours for L4 and another 8 hours to develop into a young adult must be

calculated [HALL & ALTUN, 2008]. The short generation time allows genetic

approaches with follow-ups over multiple generations [KUWABARA & O'NEIL,

2001].

The easy handling of C. elegans helped to integrate the nematode as model

organism too. C. elegans can be cultured either on agar plates, in liquid culture,

or on axenic media as alternative [KUWABARA & O'NEIL, 2001]. These are

very cheap and easy methods to culture the worm in the lab. Contaminations

with other bacteria, mold, or yeast, do not hurt the nematodes, but bother the

experiments especially to score the phenotypes. Actually, it seems that the

worms like to swing on the roots of the mold. To get rid of the contaminations, it

can be cleared by using bleach. Eggs survive the bleaching treatment

[STIERNAGLE, 1999 reprinted 2005]. To the easy handling counts also the

food source, E. coli OP50 bacteria. Also the small size about 1mm length is a

huge advantage regarding the space problem. Because of their small size,

thousands of worms can be cultured and stored easily. The discovery from J.

Sulston, that the worm can be frozen for longer time in liquid nitrogen (-196°C),

was also very helpful to culture the worm in the laboratory. Especially worms in

early larval stage (L1 & L2) survive freezing and defrosting very well, in contrast

to eggs, which do not survive [BRENNER, 1974]. Another possibility next to

freezing in liquid nitrogen is storing worms at -70°C [BÜRGLIN et al., 1998]. The

recovering rate of 35 to 45% of worms stored in liquid nitrogen is a lot better

compared to the second method with a success rate of around 10%. Also to

mention is, that not all strains are able to freeze, some mutants do not survive

Page 90: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

70

the freezing at all. Nowadays, it is possible to order either wild-type strains or

the required mutant strains of the diverse choice of mutant strains from the

Caenorhabditis Genetics Center [STIERNAGLE, 1999 reprinted 2005]. More

information about how to order strains is given on their homepage [University Of

Minnesota, 2011]. Also the databases wormbook as well as wormbase provide

an up-to date information source [WormBook, 2010] [POULIN et al., 2004].

Another good reason for the use of the nematode as model is the small size

and simplicity of the genome [BÜRGLIN et al., 1998]. Also a plus is that the

nematode C. elegans was the first metazoan with a fully sequenced genome,

with first predicted 18,972 genes, and actually 19.762 genes [KIM, 2001]

[SCHWARZ, 2005]. Only because of the now known genetic sequence of the

worm, a door with so many different possibilities got opened, like the

investigation of functions of genes in general, and whole-genome screenings

via RNAi [FRASER & AHRINGER, 2003]. Early assumptions indicated that that

about 74% of the human genome have homologous matches in the nematode

[KUWABARA & O'NEIL, 2001]. So far, homologies for around 50% of all genes

in C. elegans were found in humans [KAMATH et al., 2003]. This fact made C.

elegans a perfect model to investigate developing errors, malfunction of the

metabolism, and various diseases [KUWABARA & O'NEIL, 2001].

All those characteristics made the worm a perfect model organism for genetic

studies, molecular assays, and biological approaches [KUWABARA & O'NEIL,

2001].

4.2 RNA interference in Caenorhabditis elegans

C. elegans is the first animal where RNAi was discovered. And this was only a

few years ago, and RNAi emerged rapid into a frequently used tool

[DYKXHOORN et al., 2003]. Nematodes proved as a very useful model

organism for the first steps of the investigations. Another big advantage for C.

elegans, to be employed as a very potent model organism is, not only the

already mentioned advantages, also a huge collection of diverse mutations of

Page 91: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

71

the genome exists [SUGIMOTO, 2004]. RNAi is simple and most effective in the

nematode. A highly specific mechanism, targeting only genes homologous to

the origin, introduced RNA sequence [MONTGOMERY et al., 1998]. It does not

affect the gene expression at all, and no gene damage or genetic alterations are

found [TUSCHL et al., 1999]. RNAi was used in C. elegans to verify

homologous genes in different species, to investigate unknown gene functions

in vitro and in vivo, and to complement the classical forward genetic method

[MAINE, 2001]. Through the development of large genome libraries of RNAi

clones, it got possible for large-scale approaches to screen the entire genome

for the genes of interest [JOHNSON et al., 2009]. And RNAi sensitive strains

like rrf-3 mutants made it possible to screen in tissues, which display normally

no phenotypes in the wild-type [STEPHENS et al., 2005]. An interesting fact is

that around 50% of all genes in C. elegans were found to have a human

homologue [KAMATH et al., 2003]. And for about 75% of the genes, which are

known to be employed in the development of disease in humans, were found to

have homologues in the worm [STEPHENS et al., 2005].

Like mentioned, the misbelieve that single-stranded RNA can induce a fully

RNAi response was soon overcome [FIRE et al., 1998]. Double-stranded RNA

produces a strong and effective RNAi response in C. elegans and does not

trigger any activation of the PKR-pathway or other immune response at all

[FISCHER, 2010]. In general, RNA sequences of about 200 to 1,000

nucleotides, or even longer ones, are able to trigger an RNAi response

[TABARA et al., 1998]. The effect requires the homology of at least 80%

between the exogenous trigger dsRNA and the endogenous mRNA [KAMATH

& AHRINGER, 2003]. RNAi starts after the introduction of long dsRNA. Long

dsRNA gets cleaved by the ATP-dependent RNase III enzyme dicer into 21-

25nt long RNA-fragments, siRNAs [HUTVÁGNER & ZAMORE, 2002]. Through

the cleavage the characteristic structures of the 5’- and 3’ end of siRNAs are

generated [CAPLEN et al., 2001]. After the incorporation of the single-stranded

siRNA into RISC, siRNA guides the whole complex to the homologous mRNA,

recognizes the complement, and initiates the degradation [HUTVÁGNER &

Page 92: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

72

ZAMORE, 2002]. Also the silencing effect at the translational level occurs in C.

elegans [VASTENHOUW & PLASTERK, 2004].

In the first studies, performed at C. elegans, dsRNA was injected into the

gonads or the body cave. Later on, it was discovered that feeding dsRNA

expressing bacteria or even soaking by a high-concentrated dsRNA solution

induces RNAi [GRISHOK, 2005b]. The resulting phenotype can be very strong

and specific and look alike null-mutations of the gene. But also phenotypes can

appear weaker [TABARA et al., 1998]. It has to be mentioned, that RNAi-

phenotypes can never be counted as null-mutant phenotypes, because the

mRNA is only for limited time degraded and never fully eliminated [MAINE,

2001]. Sometimes an affected gene displays even more than just one

phenotype. This happens, if the gene is involved in more than just one function.

Typically, highly conserved genes are more affected to induce an RNAi

response than non-conserved genes [FRASER et al., 2000]. It should be

mentioned, that the time between the treatment and the appearance of the

phenotype is variable. It depends quite a bit on the stability of the protein,

because every single one has to be degraded first, before the phenotype gets

visible [MAINE, 2001]. Not all genes and tissues react with the same sensitivity.

It seems that the neuronal tissue is nearly total refractory to the RNAi effect.

The phenotype displayed can differ from animal to animal and appears not

always at the same intensity. The mentioned cross-effects occur also in C.

elegans, if a certain amount of sequence homology is given [KAMATH &

AHRINGER, 2003].

Today, an enormous amount of different C. elegans strains are available at the

Caenorhabditis Genetics Center. For regular approaches, the wild-type strain

Bristol N2, introduced from S. Brenner is used. But as mentioned, sometimes a

more sensitive strain is required [SCHEPERS, 2005].

Page 93: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

73

4.3 Specialties of the RNA interference mechanism in Caenorhabditis elegans

The RNAi effect provides in C. elegans some specialties, which are not

developed in every other organism, such as systemic RNAi, inheritance of the

RNAi effect, amplification of the signal, and the generation of secondary

siRNAs, while the core components are conserved through a variety of

organisms [SUGIMOTO, 2005].

4.3.1 Systemic RNAi

It was already seen early, that dsRNA-mediated gene silencing possesses the

ability to travel across the cellular boundaries [FIRE et al., 1998]. In fact,

systemic RNAi gets implicated as an antiviral-defense mechanism and against

environmental pathogens, contaminations of growth media [SCHEPERS, 2005].

The gene silencing effects stays highly sequence-specific through the spreading

process. So far, the spreading mechanism is well established in plants,

planarias, and C. elegans, but not at all in flies or mammals [AGRAWAL et al.,

2003] [TIJSTERMAN et al., 2004]. The phenomenon observed in plants was

named systemic acquired silencing (SAS) [SIJEN & KOOTER, 2000]. In plants,

the signal spreads via plasmodesmata to the neighbor cells, and to other parts,

further away, through the phloem [AGRAWAL et al., 2003].

Spreading in nematodes was observed the first time, after the mistaken

injection of dsRNA into the body cavity instead of the germline [GRISHOK,

2005b]. This indicated that one distribution pathway in higher animals of dsRNA

works via the ceolomic fluid [SCHEPERS, 2005]. Nowadays, there is enough

prove, that exposure of the nematode to dsRNA in anyway, either by soaking,

feeding, or injection, leads to the expected systemic effect [TIMMONS et al.,

2001]. The effect is not limited to neighbor cells at all. The signal can spread to

another differentiated tissue far away from the part of the original injection

[TIJSTERMAN et al., 2002]. The signal has also the possibility to trigger the

silencing effect in the next several generations [AGRAWAL et al., 2003].

Important to consider is, that the inheritance further than to F1, occurs only if

Page 94: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

74

genes in the germline are targeted. In general, the silencing effect in the F1

generation is more efficient than in the parents [GRISHOK, 2005b]. Also it is

displayed that the spreading effect is stronger by feeding or soaking then by

injection of dsRNA [TIMMONS et al., 2001]. And injection into the intestine is,

by far, the most effective method [GRISHOK, 2005b].

Sid-mutants and rsd-mutants are animals which lack the phenomenon of

spreading [TIJSTERMAN et al., 2002] [TIJSTERMAN et al., 2004]. Sid-1

mutants display no other phenotype than the defect to spread through the

animals. And as a result they are not able to inherit the silencing signal to their

progeny [AGRAWAL et al., 2003]. SID-1 is proved to be required for systemic

RNAi. It encodes a transmembrane protein, working by an active transport via a

sort of receptor or channel [TIMMONS et al., 2003] [AGRAWAL et al., 2003].

The exact transportation of the signal remains unknown [BERNSTEIN et al.,

2001]. The explanation for the lack of systemic RNAi in mammals might be the

fact that only closely related homologous of rsd-3 was found [TIJSTERMAN et

al., 2004]. There are no homologous of sid-1 in Drosophila, which explains the

lack of the mechanism [AGRAWAL et al., 2003]. There exists no systemic

response in neuronal tissues, because of the missing expression of sid-1

[SCHEPERS, 2005]. It seems that the whole systemic silencing pathway is

highly complex and might be tissue-specific [TIMMONS et al., 2003].

Suggestions indicate that the whole system contains more than one transport

mechanism for cellular uptake [SCHEPERS, 2005].

A recently published study implicated next to sid-1 also rsd-2, rsd-3, and rsd-6

as required genes for systemic gene silencing from the soma to the germline

[FISCHER, 2010].

Page 95: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

75

4.3.2 Inheritance

Another specialty of C. elegans, which is not given in every other organism, is

the ability of the RNAi effect to get passed on to the next generation [FIRE et

al., 1998]. And not only the next generation, is able to show the RNAi effect.

The effect is also stable inherited into further progeny [COGONI & MACINO,

2000]. The inheritable effect causes no alterations of the genomic structure of

the target sequence. Especially sensitive, for the entire RNAi phenomenon, are

the genes encoded in the germline. That results in phenotypes which are

displayed for several generations. The RNAi effect, which is passed on to the

F1 generation only, can be explained by the persistence of the signal [TUSCHL,

2001]. While long-lasting RNAi is adopted as heritable and requires the

formation of an inherited agent first, this is based on the RNAi genes rde-1 and

rde-4 [TIJSTERMAN et al., 2002]. After the formation, the agent is passed on

extra-chromosomally, through sperm or egg. In the second step, the transferred

agent must initiate an RNAi response in the second generation [KETTING et al.,

2003]. Now, rde-2, rde-3, and mut-7, which are not required to inherit the RNAi,

are required for the persistence of the RNAi effect in general and rde-1 and rde-

4 are no longer needed [ZAMORE et al., 2000]. Recent studies also found more

factors for inheritance to depend on several chromatin remodeling factors such

as isw-1, mrg-1, and hda-1 [FISCHER, 2010].

Fig. 15 Generation of an inherited agent through RDE-1 and RDE-4 and persistence of the silencingsignal trough RDE-2 and MUT-7

[TIJSTERMAN et al., 2002]

Page 96: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

76

4.3.3 Amplification / RNA-dependent RNA polymerase

The fact, that injected dsRNA into the nematode does not act stoichiometrically

and only a few molecules can trigger an effective RNAi response, arose the

idea of an amplification pathway and/or catalytic step [MONTGOMERY et al.,

1998]. It is most likely that both effects play their part in RNAi, but not

necessarily in all organisms [SHARP, 1999].

It should be noted, that more than one factor is involved to amplify the signal.

Some degree of amplification is achieved through the cleavage of the 500 to

1,000 basepair long dsRNA into a larger amount of 21-23 nucleotide long

siRNAs. Results showed, after the cleavage, a 20 to 40-fold increased molar

level of the trigger RNA compared to the target [SIJEN et al., 2001]. But this

mechanism alone cannot explain the extreme high potency and persistence of

RNA interference, and the ability for systemic RNAi [AHLQUIST, 2002]. Another

explanation would be a very efficient and highly catalytic process by the RNA-

degradation step [SHARP, 1999]. And in fact the activated RISC complex is a

classic multiple-turnover enzyme and uses siRNAs more than once

[HUTVÁGNER & ZAMORE, 2002]. And the third part is, that siRNAs may act as

primers on mRNA to produce, so called, “secondary siRNAs” [PLASTERK,

2002]. The amplification step would provide an answer to the two following

questions: How is it possible that only a small amount of dsRNA can suppress a

much larger amount of mRNA? How comes, that the silencing effect can last for

days, survive a lot of cell division rounds, and can be inherited to the next

generation(s) [ZAMORE et al., 2000]?

Suggestions implicated RNA-dependent RNA polymerases as perfect

candidates for the amplification mechanism [BASS, 2000]. They are suggested

to cause the development of a silencing agent and/or the amplification process

[ZAMORE et al., 2000]. Naturally, RNA-viruses require those enzymes to

replicate themselves by using RNA as template [SCHEPERS, 2005]. This

implicates that they carry their own viral RNA polymerase with them. That

explains the fact that in VIGS no RdRPs are necessary [MELLO & CONTE JR.,

2004]. In C. elegans, aberrant transgenic and viral RNAs are recognized by

Page 97: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

77

RdRP enzymes, used as templates to replicate new siRNAs, and afterwards to

guide the sequence-specific degradation [AGRAWAL et al., 2003]. Four

different genes, homologous to RdRPs, were found in C. elegans: ego-1, rrf-1,

rrf-2, and rrf-3 [SIJEN et al., 2001]. First suggestions implicated an equal

function for ego-1 and rrf-1. The only difference would have been the

employment in different tissues [TIJSTERMAN et al., 2002]. Now, a RRF-1

complex is proved to be the main player for RdRP activity and therefore

responsible for the synthesis of secondary siRNAs [AOKI et al., 2007].

Homologous genes, which encode RdRP proteins, are also discovered in

Arabidopsis t. (SDE-1/SGS2) and in Neurospora crassa (QDE) [COGONI &

MACINO, 2000]. It appears that different strategies for the synthesis of

secondary siRNAs are used in the different tissues and also in different

organisms [AOKI et al., 2007]. Nowadays, evidence displayed the requirement

of RdRPs in the RNAi phenomenon in Neurospora c., Arabidopsis t., and C.

elegans. But no closely related genes are found to encode RdRPs in humans or

fruit flies [HUTVÁGNER & ZAMORE, 2002]. Those results go hand in hand with

the fact that the RNAi effect in human cells is transitory. After a single injection,

the effect abolishes after 4 to 6 days, because of the lack of an amplification

step [ZAMORE, 2002].

Several possible hypotheses are brought up to explain how RdRPs might

amplify the signal. First, the guide-strand of siRNA might act as a primer to

guide RdRPs to the single-stranded complementary target RNA [COGONI &

MACINO, 2000]. Second, RdRPs could recognize products after the

degradation step and use those to generate new silencing triggers. Third, it

could also be possible that dsRNAs or siRNAs by themselves get replicated,

using the same mechanism like viruses [BERNSTEIN et al., 2001]. It is now

figured out, that single-stranded siRNAs are used as primers to synthesize new

dsRNA by using target mRNA as templates for the RdRP replication step.

Effectively, target mRNAs are recycled to new replicated dsRNAs [NISHIKURA,

2001]. Inactivation of RdRPs in RdRP-mutants like ego-1 mutant leads to a

decreased amount of siRNAs and results further in a loss of the silencing effect.

This outcome might reflect an absolute requirement for the amplification to

Page 98: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

78

produce enough secondary siRNAs to achieve the critical point of silence

[ALDER et al., 2003].

Fig. 16 Amplification mechanism in worms and plants

In mammals and fruitflies the antisense strand enters directly RISC to guide the mRNA degradation. Inworms and plants, the antisense strand is used first for an amplification step to generate secondarysiRNAs by binding to RdRP and used afterwards to guide the mRNA degradation.modified from [NOVINA & SHARP, 2004]

Page 99: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

79

4.3.4 Activation of transitive RNA interference under the generation of secondary small interfering RNAs

In C. elegans, after introduction of dsRNAs, small interfering RNAs are found

during the RNAi process, which origin definitively not from the original trigger

dsRNA. Those siRNAs contain complementary regions outside of the initial

trigger dsRNA and are termed secondary siRNAs [SIJEN et al., 2001]. Target

RNAs are not targets only; instead they are employed in the amplification of the

silencing signal. Because of that, secondary siRNAs can be seen as co-

products of the mRNA degradation [TIJSTERMAN et al., 2002]. In contrast to

the secondary siRNAs, primary siRNAs are produced from the introduced

trigger (dsRNA) through the cleavage step by dicer. The double-stranded

primary siRNAs are converted into the single-stranded forms. If they cannot find

homologous target mRNAs to basepair, they are going to be degraded. The

other option is to follow their faith and get incorporated into RISC, basepair to

target mRNA, then the amplification process could get started [PLASTERK,

2002]. Via the degradative PCR mechanisms, the siRNAs are de novo

generated. The antisense strand of the introduced siRNA binds to the

complementary target RNA and primes the amplification process. After the

synthesis, and the cleavage step by dicer, the newly secondary siRNAs are

ready to guide the next round of target degradation, referred to as transitive

RNAi. This results in the possibility, to target more mRNAs for degradation, and

leads to an extended duration, and the enormous potency of the RNAi effect

[CHI et al., 2003].

As already mentioned in the previous chapter, the generation of secondary

siRNAs requires RdRPs and might be tissue-specific [PLASTERK, 2002]. The

early suggestions that the newly synthesized secondary siRNAs run through the

same cycle as introduced dsRNAs including the dicer cleavage step, proved not

entirely correct. RRF-1 complex is the main factor for RdRP activity and

generates secondary siRNAs in a dicer-independent manner. This suggestion

derives from studies in dcr-1 deficient mutants. Secondary siRNAs are by far

more efficient to cleave the target mRNAs than the original primary single-

stranded or double-stranded siRNAs. This discovery implicates that rather

Page 100: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

80

secondary siRNAs than primary siRNAs are the important intermediates to

degrade the target during RNAi in C. elegans. It seems that the secret is the

activation of the slicer activity by the argonaute protein CSR-1. It is suggested

that secondary siRNAs and CSR-1 form a secondary RISC with a higher

efficiency to degrade the target [AOKI et al., 2007]. Suggestions indicated that

in C. elegans the newly, in the cytoplasm synthesized, secondary siRNAs have

to be transported via NRDE-3 into the nucleus, where the RNAi occurs in C.

elegans [FISCHER, 2010].

Because of their own characteristics, the secondary siRNAs are a new type of

small RNAs. Secondary siRNAs contain a triphosphate group on the 5’ end, are

always accumulated as the antisense strand, and do not have the characteristic

5’-monophosphate group of the dicer cleavage. The 3’ end formation of the new

type of small RNAs remains to be discovered [FISCHER, 2010]. It is also still

unclear if secondary siRNAs participate in other biochemical reactions [AOKI et

al., 2007]. One of the consequences of the generation of secondary siRNAs is

the spreading effect [CHI et al., 2003].

The first time, transitive silencing was observed, was in C. elegans cell cultures,

it was also displayed in C. elegans in vivo, and later on in plants as well [CHI et

al., 2003] [AHLQUIST, 2002]. Transitive RNAi is not restricted to transgene

targets, it can also silence endogenous gene expression [SIJEN et al., 2001]. In

rrf-1 mutant nematodes, transient RNAi is displayed in germline cells, but

completely inactive in somatic cells [NISHIKURA, 2001]. In rrf-1 mutant animals,

the secondary siRNA production is blocked, which further results in no

activation of the transitive silencing pathway [AHLQUIST, 2002]. Due to fact that

no RdRPs are carried in mammalian and Drosophila genomes, no secondary

siRNAs can be generated and as a result no transitive RNAi effect can be

generated [ALDER et al., 2003]. One disadvantage might be, that the de novo

synthesized siRNAs could target also other mRNAs than the original ones and

induce through those side effects [DUDLEY et al., 2005].

Page 101: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

81

4.3.5 Genes required for RNAi in Caenorhabditis elegans

One of the first identified mutants regarding the RNAi pathway were mutations

in the rde-1 through rde-4 genes [BOSHER & LABOUESSE, 2000]. Rde-1 and

rde-4 mutants (rde stands for RNAi deficient) lack an RNA interference

response, but no other defects or visible phenotypes can be scored. Under

laboratory conditions, those mutants are fertile, healthy, and grow without any

other deficiencies [TABARA et al., 1999]. Researchers found that rde-4 and rde-

1 are required in the inheritance step of the silencing signal though the

formation of a hereditary agent, which results in transitive RNAi [GRISHOK et

al., 2000]. Important is, that rde-1 is only required for the formation of the agent

and not necessary for the actual silencing effect [TUSCHL, 2001]. So rde-1 and

rde-4 are the initiators for the silencing effect, while rde-2 and mut-7 work

downstream as the effectors of the response, via targeting the specific mRNAs

for the silencing phenomena in the progeny [GRISHOK et al., 2000]. It seems,

that rde-1 and rde-4 were the only two genes essential for the RNAi response in

all tissues, somatic and germline, of the worm [SIJEN et al., 2001]. This

assumption is based on the fact, that rde-1 mutants are deficient in RNA

interference in the soma as well as in the germline [BERNSTEIN et al., 2001].

The requirement of RDE-1, RDE-4 and the later mentioned RRF-1 for dsRNA-

mediated interfering effect is now established [CERUTTI, 2003].

Rde-1 is a gene highly conserved in organisms varying from plants to

vertebrates and is nowadays counted as member of the big argonaute family

[TABARA et al., 1999]. Rde-1 contains the typical structure for the argonaute

family, the PPD domain. At the N-terminal the PAZ domain and on the C-

terminal the PIWI domain is located [NYKÄNEN et al., 2001]. It is proved, that

argonaute proteins play a key role in the RNAi mechanism and in related

silencing phenomena. They are implicated in networking with siRNAs and in

formation of different effector complexes such as RISC and miRNP [KIM, 2005].

Those two domains, PAZ and PIWI, interact also with the characteristic 3’ and

5’ ends of the siRNAs. The PIWI domain is implicated to possess slicer

characteristics (RNA cleavage) like the related RNase H. This slicer activity

might get activated during the base pairing event from siRNA with the target

Page 102: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

82

mRNA. The genome of C. elegans encodes 24 members of the argonaute

proteins. Some are predicted like rde-1 to possess the slicer activity, some

seem to display effectiveness for some certain siRNAs, and others might be

employed in other biochemical reactions of the organism [FISCHER, 2010].

Homologous of rde-1 are found in Arabidopsis t. referred to as Ago-1, qde-2 in

Neurospora crassa, and sting in Drosophila [DUDLEY et al., 2005] [TABARA et

al., 1999]. It seems that rde-1 is a gene essential for RNAi, but is not required

for other essential function in the organism [TABARA et al., 1999]. Mutations of

homologous of rde-1, alg-1, and alg-2 have shown developmental

abnormalities. This results from the fact, that those two genes are required for

the function of lin-4 and let-7 [GRISHOK et al., 2001].

Rde-4 expresses a dsRNA binding protein, which forms a complex with dicer

protein. In rde-4 mutants, no production of siRNAs was visible. In fact, rde-4 is

employed in the generation of siRNAs through the cleavage of dicer via

recognition of the trigger dsRNA. A homologous gene to rde-4 was found in the

Drosophila RNA binding protein R2D2. Also R2D2 is found in a complex with

DRC-1 and it is proved to be essential for RNAi. The differences are that R2D2

is required to bind instead of the injected dsRNA, siRNAs, and incorporate them

afterwards into the RISC complex [GRISHOK, 2005a].

Rde-1, rde-3, and rde-4 mutant strains lack completely in expressing an RNAi

effect, while it seems that alterations in rde-2 do not always result in a non-

silencing outcome. If genes are targeted, which are expressed in germline, no

RNAi response is displayed, but somatically encoded targets are sensitive to

RNA interference and silencing can be triggered [TIJSTERMAN et al., 2002].

Interestingly, rde-1 and rde-4 mutants only lack an RNAi response, but are not

defective for co-suppression or transposon silencing. But alterations of rde-2,

rde-3, mut-2, mut-7, mut-8, and mut-9 are defective in both, RNAi and co-

suppression [TUSCHL, 2001].

Some mutations found to be employed in transposon silencing. Mut-2, mut-7,

(mut stays for transposon-silencing mutants) rde-2, and rde-3 exhibit an

increased mobilization of transposons in germline, while they show a reduced

Page 103: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

83

RNAi response. Also mut-6 animals are implicated into mobilization of

transposons as well. In contrast to mut-2 and mut-7 strains, mut-6 strains are

only able to mobilize Tc1 transposons. Those results indicate a connection

between the RNAi and the transposon silencing pathways [TABARA et al.,

1999]. Here, the mutations in rde-2, rde-3, and mut-7 display also other

phenotypes than the mentioned ones. They show a decreased fertility, a

temperature-dependent sterility, and a higher rate of newborn males

[GRISHOK, 2005a]. It was discovered that some mut-strains can display a rde-

phenotype and the opposite around [KETTING & PLASTERK, 2000]. For

example, mut-2 and mut-7, those two germline specific genes display a

significant lack of RNAi response, compared to mut-6, a strain where efficient

RNAi can be triggered [TABARA et al., 1999].

Four members of RdRP-like genes were found in C. elegans, ego-1, rrf-1, rrf-2,

and rrf-3. Ego-1 and rrf-1 are closely linked genes [SIJEN et al., 2001]. For an

efficient RNAi response in C. elegans ego-1 and rrf-1 are required. This fact

results from experiments, that displayed that alterations in those two genes lost

the possibility for RNAi in both, somatically and germline expressed genes

[SIMMER et al., 2003]. Further research distinguished between those two.

There was no evidence for any employment of ego-1 during RNAi in somatic

cells [NISHIKURA, 2001]. It was figured out, that EGO-1 is employed in the

RNAi response in the germline and for the somatic RNAi response RRF-1 is

responsible. So it seems that ego-1 and rrf-1 are complementing each other

[GRISHOK, 2005b]. This in fact is very useful to identify genes working either in

somatic tissue or in the germline [GOLDEN & O'CONNELL, 2007]. Only

recently it was found, that there is no amplification mechanism of siRNAs and

further no transitive RNAi is displayed in rrf-1-mutants [FISCHER, 2010]. A

homologous of ego-1 is found in qde-1 in Neurospora crassa and sde1 in

Arabidopsis thaliana. These findings also prove the relationship of the three

different phenomena, PTGS, RNAi, and quelling [COGONI & MACINO, 2000].

No functions for rrf-2 in the RNAi pathway were found [SIMMER et al., 2003].

Page 104: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

84

Another important mutation for the RNA interference effect is the alteration in

the gene rrf-3 [SIMMER et al., 2003]. Alterations in rrf-3 enhance the sensitivity

to RNAi in the animal compared to the wild-type. This results in stronger

phenotypes, which are sometimes closely looking like a null-phenotype. Also an

advantage is, by using the rrf-3 mutated strain, that phenotypes for some genes

in neuronal tissue are possible [SIMMER et al., 2002]. It got also possible to

observe phenotypes, in the hypersensitive rrf-3 strain, which were easily

overlooked in the wild-type, because the phenotype was hardly visible

[SIMMER et al., 2003]. Some side effects of the rrf-3 strain are the high birth-

rate of males, the sensitivity of the worms to temperature, and the decreased

number of progeny, because of failures in the spermatogenesis [SIMMER et al.,

2002] [STEPHENS et al., 2005]. It occurred that rrf-3 is able to synergy with

some active genes in the germline [STEPHENS et al., 2005].

Additional to the enhancing rrf-3 strain, 19 different eri genes (eri stands for

enhanced RNAi) were found [GRISHOK, 2005b]. Eri-1, the strongest one, is

suggested to enhance RNAi by increased sensitivity to siRNAs. An increased

accumulation of the dicer products in eri-1 mutants was shown, compared to

wild-type worms. ERI-1 is mainly a cytoplasmic protein, and is especially

encoded in the gonads and in parts of neurons. This would explain the fact, why

the neuronal tissue hardly responds with RNAi after the introduction of the

trigger dsRNA. It seems that the exonuclease protein ERI-1 blocks the silencing

by increasing the stability of the duplex structure and the degradation of the 3’

overhang of the siRNAs. Without the characteristic structure, siRNAs cannot

enter RISC to guide the cleavage of the target mRNAs. Eri-1 might be one

regulation factor for the silencing phenomenon, but it is not clear, if the

regulation is limited to particular cell types or if eri-1 regulates the whole RNAi

machinery in all tissues [KENNEDY et al., 2004]. Also in LIN-15B, a protein,

enhancing the RNAi effect was found. Especially hypersensitive for RNAi is the

eri-1 and lin-15B double mutation. In that strain, it is possible to achieve results

in genes encoded in neuronal tissue. Because of that, this mutated strain is

used for large-scale screens for genes with neuronal functions [JOHNSON et

al., 2009].

Page 105: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

85

Sid stands for systemic RNAi-deficient loci, which means that animals with

mutations in those genes cannot display the earlier mentioned systemic effect.

Alterations in sid-1 result only in the lack of the spreading effect, no other

obvious phenotypes can be seen, and the fertility seems to be normal. Sid-1 is

expressed in RNAi sensitive cells and is employed in the transport of the signal

from the place of the original uptake to other tissues and into the worm’s

progeny. SID-1 is a transmembrane protein, mainly located in the cell periphery,

and implicated into the import and export of the silencing signal. It might work

as a channel for either dsRNA, siRNA, or some other still unknown silencing

signals [WINSTON et al., 2002]. As mentioned before, neuronal tissue is often

totally and often partly noncompliant to the silencing effect. It might be possible,

that most of the neurons lack the RNAi response, because of the absence of

the transporter SID-1 [JOHNSON et al., 2009]. Other genes implicated in RNAi

in C. elegans are rsd (stands for spreading defective) mutants. Different rsd

genes were found. Two independent groups worked on spreading deficient

RNAi mutations. One group published RSD-8, a transmembrane protein,

suggested to act as channel or receptor. The other group identified RSD-8 as

SID-1. Results indicated that mutants of rsd-4 and rsd-8 have no cellular uptake

of dsRNA into the animal. Compared to those two, rsd-2, rsd-3, and rsd-6, are

able to uptake the dsRNA from the gut into somatic cells, but lack in the further

distribution of the trigger into the germline [TIJSTERMAN et al., 2004].

4.4 Practical aspects and methods of RNAi in Caenorhabditis elegans

There are four possible ways to induce an RNAi effect in the nematode C.

elegans; first, by injection of dsRNA into the worm; second, by feeding bacteria

expressing dsRNA; third, by soaking them with a concentrated dsRNA solution;

and fourth, by in vitro transcription of dsRNA from transgene promoters

[ZAMORE, 2001] [TIMMONS et al., 2003].

Page 106: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

86

For a long time, it seemed that soaking and feeding results in weaker RNAi

responses than with injection, but with supplementations in the food or with

other modifications, equivalent RNAi responses can be achieved [KUMABARA

& COULSON, 2000] [MAEDA et al., 2001]. In general, certain factors can

influence the outcome of the phenotype, such as the choice of the sequence,

delivery method of dsRNA, the developmental stage at which the treatment

started, the temperature, and the stability of the target mRNA, including the

stability of the expressed proteins [MAINE, 2001].

To summarize, all methods are potent to induce a visible RNAi response, but

they all have key differences. So, it is clear that there is never just one golden

approach for all experiments. All of these four tools should be seen as

complement to each other, and not as a replacement of the previously invented

method [KAMATH et al., 2000]. The ideal method depends always on the

approach, the design of the experiment, and especially on the targeted gene.

For smaller amounts of worms or for example, maternally encoded genes,

soaking or injection are the methods of choice. For high-throughput screens, or

approaches which require a large number of treated nematodes, soaking and

feeding are amenable [FRASER & AHRINGER, 2003]. For silencing at a

specific developmental stage, feeding and soaking are recommended. To

achieve efficiency silencing in late larval stages, feeding nematodes with

Fig. 17 The four possible methods for RNA transfection

aǀ Microinjection of in vitro synthezised dsRNA bǀ Soaking into a dsRNA solution cǀ Feeding of dsRNA expressing bacteria dǀ in vivo transcription of transgene [SUGIMOTO, 2005]

Page 107: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

87

dsRNA expressing E. coli bacteria is recommended. Also for assays,

investigating the longevity or behavior, feeding is the method of choice

[SUGIMOTO, 2004]. Soaking displays in some cases stronger phenotypes than

feeding. This can be utilized, especially in phenotypes, which are only hardly

visible using the feeding method [JOHNSON et al., 2009]. It should not be

overseen that soaking and feeding are methods which are easy to perform and

no special skills are needed. Compared to injection where the scientist has to

learn how to inject the dsRNA into the nematode without hurting or killing the

worm [GOLDEN & O'CONNELL, 2007]. The transgene based delivery method

is not amenable for high-throughput approaches, because the design of the

transgenic strains requires multiple steps for each target [SUGIMOTO, 2004].

4.4.1 Microinjection

One possibility to induce RNAi in C. elegans is by microinjection of dsRNA. The

spreading effect is a big help for microinjection into C. elegans. It makes it

possible, that the injection can take place at every site of the animal. Even when

the target gene is expressed in another tissue further away, dsRNA is delivered

into that place [TABARA et al., 1998]. Also for the inheritance effect into the

next generation, the injection site is not crucial at all. All three ways, adult

germline, large intestinal cells or body cavity have no influence on the efficiency

of the RNAi response and are equally effective [HUNTER, 1999]. This offers a

very good method to study gene functions in embryos and different genes,

required for early development of the nematode [FRASER & AHRINGER,

2003].

Usually, the injections are executed in older, larger animals like larvae in stage

4 (L4) or in young adults, to score the phenotypes in the progeny. It displays

stronger phenotypes, particularly in early expressed genes, with their function in

germline development and embryogenesis [GOLDEN & O'CONNELL, 2007].

Another reason for the use of bigger animals is the easier performance of the

injection into them, than into younger and smaller ones [AHRINGER, 2006]. The

Page 108: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

88

usual injection sites are the intestinal tract or into one arm of the gonads in

hermaphrodites. This generates enough interfering response to target most of

the interested genes in the cells of the gonads and also in other tissues of the

animal [SCHEPERS, 2005]. Typically, animals should be moved to a new petri-

plate to observe the progeny. In general, the phenotypes can be scored 12 to

24 hours after the injection. It is normal, that the offspring laid during the first 24

hours displays not the strongest effects, but after that, phenotypes should

display more intense interfering response [GOLDEN & O'CONNELL, 2007]. An

effective silencing is displayed in early larval stages of hermaphrodites of the

infected progeny, while the adults displayed already an initial recovery of the

interference effect. This might be explained by the fact of dilution of the initial

trigger by cell division [TIMMONS et al., 2001]. Genes, which are expressed

more than one time during the development, must be targeted in every single

developmental stage to reduce the gene products [MAINE, 2001].

Microinjection was the first invented method for dsRNA delivery [STEPHENS et

al., 2005]. Some limitations occur with this method, which have to be calculated

in the design of every approach. It is the most labor intensive method, a lot of

equipment is required, and certainly not the method of choice for researchers

starting with RNAi experiments in C. elegans [SCHEPERS, 2005]. It takes some

time to learn the procedure. To infect 10 to 30 hermaphrodites, around half an

hour is needed; every individual animal has to be injected one after each other.

So the whole process is not adaptable for large groups, large-scale, or whole-

genome screens [FRASER & AHRINGER, 2003]. Typically, only one to ten

nematodes are treated by the microinjection-method per experiment

[JOHNSON et al., 2009]. It is believed, that injection of dsRNA into C. elegans

is the most effective way to generate an interference response [HUNTER,

1999].

Page 109: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

89

4.4.2 Feeding of dsRNA expressing bacteria

It was figured out, that not only injection of double-stranded RNA into the animal

results in an RNAi response; also the possibility of feeding dsRNA is given. In

general, not naked dsRNA is used as food source, especially generated E. coli

bacteria, which expresses the corresponding dsRNA of the gene of interest in

the host organism, serves as food source on agar plates [HUNTER, 1999]. The

phenotypes are scored either in the treated nematode or in the F1 generation

[JOHNSON et al., 2009].

For a long time, researchers were not of the same opinion about the

effectiveness of the feeding method, compared with the injection method. Some

researchers postulate the feeding method as less effective than direct

microinjection [TIMMONS & FIRE, 1998]. The silencing response is especially

less powerful during development of late embryonic and early larval stages, but

potent to trigger gene silencing in adults and early embryos [TIMMONS et al.,

2001]. The idea arose, that it might be possible that with starting the treatment

earlier, the animals are exposed for a longer period of time, and this limitation

might be overcome. Those treated worms exhibit phenotypes at least as strong

as phenotypes of nematodes treated with direct injection, and it is as sensitive

as gene silencing by dsRNA injection. Exclusive the nervous system, nerve

cells respond only hardly with a silencing effect [KAMATH et al., 2000]. Also

continuous feeding, results in a more potent outcome of the silencing effect in

postembryonic phenotypes [FRASER & AHRINGER, 2003].

It was also figured out that the feeding method might be gender-specific. Males

(him-strains) appeared to have a decreased uptake of dsRNA compared to

hermaphrodites. It can be seen in the results, that males are less affected by

RNAi. The explanation might be simple, males are not eating that much; or

could be more complex, that indeed the gender has influence on the dsRNA

uptake from the food source, or even on the RNAi mechanisms, or its

components itself are responsible [TIMMONS et al., 2001].

There are some advantages of this method over the injection. The feeding is a

method far less labor intense, easy to use, most inexpensive compared with

Page 110: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

90

injection or soaking, no special equipment is required, it is amenable for a large

number (thousands) of worms at the same time, and in a short time period

possible to test a large number of various genes at the same time, genome-

wide analysis are amenable, and it is used to detect genes participating in the

RNAi mechanism itself [KAMATH et al., 2000]. Another positive effect is the

possibility to feed at every developmental stage. So the down-regulation of the

target is obvious in every favored stage [GOLDEN & O'CONNELL, 2007]. One

drawback, which is also an advantage too, is the design of the E. coli bacteria to

express the corresponding dsRNA to the target gene. It is quite an amount of

work to clone the desired DNA structure into a transport plasmid vector and

transform it into the specific E. coli. On the other hand, once the strain is

generated, it is possible to reuse it and replicate it for very little cost [KAMATH &

AHRINGER, 2003]. Libraries of the designed E. coli strains are developed,

containing 16,757 bacterial strains. This means, 86% of the back then predicted

19,427 genes of C. elegans are covered [KAMATH et al., 2003]. To execute an

RNAi experiment by feeding the ready-made, commercially available E. coli

strains, is a very easy method to perform, also for beginners in this field

[JOHNSON et al., 2009]. Only through the development of the RNAi feeding-

libraries, gene identifications regarding chromosome morphogenesis, lifespan,

nucleotide excision repair, fat storage, and transposon silencing became

possible [STEPHENS et al., 2005].

For the plasmid design, there are two different possibilities. Either a single

fragment of the sequence of interest is cloned into a vector, flanked by two T7

polymerase promoters, called bi-directional promoter configuration, or a hairpin

structure is designed with a single T7 promoter and an inverted repeat structure

broken up by a non-homologous sequence [TIMMONS et al., 2001]. In the host,

double-stranded RNA will be expressed, further processed by dicer into

siRNAs, and after the cleavage step the siRNAs will guide the target mRNA

degradation [SCHEPERS, 2005].

Important facts to consider before starting the experiment are first, the starting

point of the first exposure to the dsRNA-containing food. Recommended is the

Page 111: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

91

use of L1 larvae, to achieve the longest exposure time, but certainly it depends

also on the gene that should be silenced. Second, there should be some tests

on the temperature before starting, because the temperature can have a great

influence on the resulting phenotypes [SCHEPERS, 2005]. Low temperature is

recommended, so bacteria cannot find their optimal living conditions. Third,

whether the liquid culture or the culture using agar plates is preferred.

Thousands of nematodes can be cultured on RNAi plates, but millions of worms

can be grown in liquid culture. The difference is that the expressed phenotypes

are more visible on agar plates. Fourth, to avoid contaminations, plates as well

as liquid cultures should be supplemented with carbenicillium. Worms favor

eating the contaminant instead of the bacterial strains, Escherichia coli strain

HT115, bearing the dsRNA [JOHNSON et al., 2009].

4.4.3 Soaking of Caenorhabditis elegans in dsRNA solution

Soaking nematodes in a dsRNA solution proved also to be an effective method

to induce RNA interference [BOSHER & LABOUESSE, 2000]. The worms are

soaked for 24 hours in a high concentrated dsRNA solution without any other

food source. After this process, the worms are transferred to agar plates to

recover. Now, the phenotypes can be observed and scored [SUGIMOTO,

2004]. It should be mentioned that the silencing effect, based on the soaking

method, is dependent on the concentration. The minimum concentrations to

produce a visible phenotype are variable and depend on the targeted gene itself

[SUGIMOTO, 2005]. In general, soaking the nematodes in the high-

concentrated dsRNA (1-5mg/ml / 24h) solution can be performed at any

developmental stage. Results in the changed phenotypes can be counted in the

parent generation as well as in the progeny [FRASER & AHRINGER, 2003]. It is

suggested that the intake of dsRNA occurs though the pharynx. With

supplementation of serotonin, the pharyngeal pumping can be stimulated, which

is possible, but not required [JOHNSON et al., 2009]. Soaking was also

believed to be not equally potent as microinjection. But it was figured out later,

that with supplementation of spermidine into the dsRNA solution, the potency

Page 112: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

92

and reproducibility is significantly improved. The method is effective for targets

employed in early embryogenesis, germline development, and muscle function.

As mentioned earlier, soaking is not potent enough to efficiently induce

silencing in nervous cells [SCHERER & ROSSI, 2005].

Advantages of the feeding and soaking methods are that younger nematodes in

their larvae stages 1 to 4 can already be used, for microinjection they would be

too small [MAINE, 2001]. Soaking is by far not that labor-intensive as

microinjection and less time-consuming [MAEDA et al., 2001]. It is easy to

perform and no special equipment is needed [SUGIMOTO, 2005]. Usually,

between 10 and 100 worms are treated per experiment. With the development

of dsRNA libraries, high-throughput analysis became approachable [JOHNSON

et al., 2009]. It is a great method to investigate genes, which play multiple roles,

and which are expressed in more than one developmental stage [SUGIMOTO,

2005]. A huge disadvantage is the high cost and amount of time for the large

amount of in vitro synthesized and purified double-stranded RNA. Another

drawback is that the exposure time is limited [GOLDEN & O'CONNELL, 2007].

And it could be possible that the worms get intoxicated by the treatment

[JOHNSON et al., 2009]. Several groups in different labors performed analysis

via soaking nematodes in dsRNA. The results were largely consistent, but there

were some in-coherences as well. This might be explained by different

definitions of phenotypes and different culture conditions [SUGIMOTO, 2005].

Depending at the developmental stage, in which the gene of interest is

expressed, there are different approaches to reach the gene knock-down. To

silence genes employed in early embryogenesis, L4 worms are soaked for 24

hours. Over the period of time, the larvae grow into adults, but the oogenesis

stays in the initial condition, and it seems that the RNAi effect is executed

during this time [SUGIMOTO, 2005]. For detecting genes function in

embryogenesis and post-embryonic development silencing, the soaking method

is ideal. L1 larvae are soaked. During the period of soaking there occurs no

development, the L1 stays arrested, but the silencing continues. In contrast,

Page 113: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

93

injection or soaking of L4 worms results in embryonic lethality and as a result no

post-embryonic phenotypes can be scored [MAEDA et al., 2001].

4.4.4 Multiple gene interactions

Experiments proved that multiple gene interactions are not very feasible.

Feeding bacteria expressing two different genes resulted in a significant

reduced displayed phenotype of both genes [KAMATH et al., 2000]. The

approach to target multiple genes in one approach displayed even weaker

phenotypes, than when two genes were silenced at the same time [JOHNSON

et al., 2009].

4.5 Knockout of the reduced folate carrier

Folate is an essential water-soluble B-vitamin, which cannot be synthesized by

humans. As a result, humans and other mammals as well, are dependent on

exogenous sources of the vitamin. It is employed in the normal cell function and

development of the organism. Deficiency of folate leads to various deficiency

symptoms, such as megaloblastic anemia, growth retardation, and neurological

disorders [BALAMURUGAN et al., 2007]. Supplementation of folate during

pregnancy is highly recommended to prevent neural tube defects. It was figured

out, that reduced folate carrier (RFC) in humans (hRFC) is responsible for the

folate uptake, additional to folate receptors, and the proton coupled folate

transporter [AUSTIN et al., 2010]. Surprisingly, two orthologs of the hRFC were

found in the nematode C. elegans, called folt-1 and folt-2, which share a 40%

and a 30% identity. RNA interference proved now the existence of an active

folate-uptake system through folt-1 in the worm [BALAMURUGAN et al., 2007].

Page 114: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

94

Folt-1 encodes an around 46.5kDA heavy protein, composed out of 410 amino

acids and ten transmembrane domains, including a loop between the

transmembrane domain 5 and 6 of about 58 amino acids. Folt-1 is differentially

expressed in the tissues. The highest level is seen in the pharynx and the

intestine of the nematode. Suggestions implicate the intestine to secret different

kinds of digestive enzymes, which further lead to the assumption that the

intestine is involved in the folate uptake of C. elegans. Folt-1 proved to be Na+-

independent, but displayed a higher activity rate at a pH of 5.5 than at 7.4.

Evidence displayed the sensitivity to anti-inflammatory agent sulfasalazine as

well as to the anion transport inhibitors DIDS and SITS [BALAMURUGAN et al.,

2007]. C. elegans knockout mutations displayed a significant inhibition of folate

uptake, which seems to lead to infertility through defects in germline

proliferation, decreased life span and metabolism, and defective oogenesis in

hermaphrodites. Additional in males, a decreased production of sperm is

displayed. Those effects can be seen universally and are not limited to C.

elegans. In mice embryos and Drosophila melanogaster, a lack of folate results

in embryonic lethality. And as mentioned before, folate deficiency in humans

during pregnancy can lead to neural tube defect [AUSTIN et al., 2010].

Fig. 18 Predicted structure of the folt-1 protein

modified from [BALAMURUGAN et al., 2007]

Page 115: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

95

5. Conclusion

After a year, reading and writing about the RNAi mechanisms, I am absolutely

the same opinion as A. Fire and C. Mello; RNA interference is a very powerful

tool. And Andy Fire and Craig Mello knew that they discovered something

incredible, but they had no idea how incredible it really was [MELLO, 2007].

Despite all the mentioned pros and contras, including the side-effects and off-

target effects, RNA interference is a very potent and specificity mechanism. I

am confident, that all those problems like the stability of the silencing agent,

modifications of the agent, sufficient uptake of the lowest possible dose, and the

activation of one arm of the immune system, can be overcome with a lot of

research.

The expectations and hopes for the development of novel drugs and individual

gene therapy are still very high, but every method is shadowed with drawbacks

[TUSCHL & BORKHARDT, 2002]. In my opinion, the biggest concern is the

usage of the RNA interference as a therapeutic tool. Through using this

mechanism, the natural system gets hijacked. It might be possible that the

endogenous function may work not with its fully capacity which could result in

an insufficient endogenous response and further might displays, still unknown,

unwanted side-effects [SHAN, 2010]. This problem has to be investigated more

rigorous, before any applications as treatments can get started.

I would like to finish with the words, quoting C. Mello:

„I think we almost always underestimate the complexity of life and of

nature” [MELLO, 2007].

Page 116: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 117: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XVII

V. SUMMARY

RNAi is a gene silencing mechanism; two decades ago, Craig Mello and

Andrew Fire discovered the knock-down phenomenon in C. elegans. In general

the mechanism starts by cleavage of long double-stranded RNA by the RNase

III enzyme dicer into 21-25nt long small interfering RNAs. After the activation of

the RISC complex through ATP and the recognition of the small interfering

RNA, the double-strand gets unwind by a helicase. The strand with the lower

thermodynamic stability at the 5’-end enters RISC (typically the antisense

strand) while the other one (the sense strand), gets degraded. By perfect

complementarities of the target mRNA and the siRNA, the target gets cleaved

and destroyed, in contrast to imperfect complementarity, where siRNA works

like microRNA and results in an translational repression by binding to the 3’UTR

region of the target mRNA.

Some specialties like the systemic effect, the inheritance, the transitive

character, and the generation of secondary siRNA appear not in every

organism. Mammals and Drosophila melanogaster lack the mechanism to

generate secondary siRNAs. This depends on the missing homologous of

RdRP enzymes, and results in the transient effect of RNAi in mammals. The

spreading mechanism is established in plants, planarias, and C. elegans, but

not in flies or mammals. This is based on the fact, that in those two organisms,

homologous of rsd or sid are missing.

RNA interference is next to the gene silencing mechanism involved in different

natural functions such as heterochromatin modification and methylation,

transposon silencing, works in lower organisms as an ancient defense system,

and can induce an immune response in mammals. This results in the activation

of the PKR pathway and the activation of the 2’-5’ oligoadenylate synthase. This

fact, as well as the possibility to induce off-target effects and cross interference,

complicates the development of therapeutic agents.

Page 118: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 119: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XIX

VI. ZUSAMMENFASSUNG

RNA Interferenz ist ein Mechanismus zur Gen-Abschaltung. Vor etwa 20 Jahren

entdeckten C. Mello und A. Fire dieses „knock-down“ Phänomen in C. elegans.

Generell startet der Mechanismus mit dem Spalten von einem langen,

doppelsträngigen DNA-Molekül durch Dicer, ein RNase III Enzym, in lauter

Bruchstücke (small interfering RNAs) von einer Länge von 21 bis 25nt. Der

RISC-Komplex muss mithilfe von ATP aktiviert werden, erst danach wird das

doppelsträngige RNA-Molekül an den Komplex gebunden und durch eine

Helicase in Einzelstränge aufgespalten. Üblicherweise wird der Strang mit der

geringeren thermischen Stabilität am 5‘ Ende an RISC gebunden (antisense)

und der andere wird abgebaut (sense). Ist das Fragment mit der Ziel-RNA

komplementär, wird dieses durch Spaltung zerstört, bei einer partiellen

Komplementarität kommt es zu keiner Spaltung, sondern zu einer Hemmung

der Translation.

Manche Besonderheiten wie der systemische Effekt, die Vererbbarkeit des

Interferenz-Signals, die Möglichkeit zur Vervielfältigung des Signals und die

Produktion von secondary siRNA sind nicht in jedem Organismus gegeben. In

Säugetieren und Drosophila melanogaster können keine secondary siRNAs

produziert werden, was auf fehlende RdRP-Enzyme zurückzuführen ist. Die

Möglichkeit zur Signalverteilung im gesamten Organismus ist in Pflanzen,

Planarien und C. elegans gegeben, jedoch nicht in Fliegen oder Säugetieren.

Dies ergibt sich aus dem Fehlen von homologen rsd- und sid-Genen.

RNA-Interferenz ist nicht nur ein Mechanismus der zur zielgerichteten

Abschaltung von Genen genutzt werden kann, sondern besitzt zudem

verschiedene natürliche Funktionen wie Modifikation und Methylierung von

Heterochromatin sowie Kontrolle der Transposons. In einfacher entwickelten

Lebewesen wirkt RNA-Interferenz als ein Abwehrsystem gegen Viren und in

Säugetieren kann es eine Immunreaktion auslösen. Zusätzlich zur Aktivierung

des Immunsystems erschweren auch die Möglichkeiten von off-target effects

sowie cross-interference die Entwicklung von neuen, individuellen Gentherapien

und Medikamenten.

Page 120: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 121: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXI

VII. REFERENCES

"The Nobel Prize in Physiology or Medicine 2006" Nobelprize.org. (2011).

Retrieved February 12, 2011, from

http://nobelprize.org/nobel_prizes/medicine/laureates/2006/.

AGRAWAL, N., DASARADHI, P., MOHMMED, A., MALHOTRA, P.,

BHATNAGAR, R. K., & MUKHERJEE, S. K. (2003). RNA Interference:

Biology, Mechanism, and Applications. Microbiol. Mol. Biol. Rev., pp.

67(4):657-685 doi:10.1128/MMBR.67.4.657-685.2003.

AHLQUIST, P. (2002). RNA-Dependent RNA Polymerase, Viruses, and RNA

Silencing. Science, pp. 296:1270-1273 doi:10.1126/science.1069132.

AHRINGER, J. (2006). Reverse genetics. In WormBook, ed. The C. elegans

Research Community. doi:10.1895/wormbook.1.47.1:

http://www.wormbook.org.

ALDER, M. N., DAMES, S., GAUDET, J., & MANGO, S. E. (2003). Gene

silencing in Caenorhabditis elegans by transitive RNA interference. RNA,

pp. 9:25-32 doi:10.1261/rna.2650903.

ALMEIDA, R., & ALLSHIRE, R. C. (2005). RNA silencing and genome

regulation. Trends Cell Biol., pp. 15:251-258

doi:10.1016/j.tcb.2005.03.006.

AMBROS, V. (2001). microRNAs: Tiny Regulators with Great Potential. Cell, pp.

107:823-826 doi:10.1016/S0092-8674(01)00616-X.

AMBROS, V. (2004). The functions of animal microRNAs. Nature, pp. 431:350-

355 doi:10.1038/nature02871.

AMBROS, V., LEE, R. C., LAVANWAY, A., WILLIAMS, P. T., & JEWELL, D.

(2003). MicroRNAs and Other Tiny Endogenous RNAs in C. elegans.

Curr. Biol., pp. 13:807-818 doi:10.1016/S0960-9822(03)00287-2.

AOKI, K., MORIGUCHI, H., YOSHIOKA, T., OKAWA, K., & TABARA, H. (2007).

In vitro analyses of the production and activity of secondary small

Page 122: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXII

interfering RNAs in C. elegans. EMBO J., pp. 26:5007-5019

doi:10.1038/sj.emboj.7601910.

APPASANI, K. (2005). Introduction. In K. APPASANI, RNA Interference

Technology: From Basic Science to Drug Development (pp. 1-13).

Cambridge: Cambridge University Press.

AULNER, N., & JAGLA, B. (2008). Validation of RNAi. In M. LATTERICH, RNAi

(pp. 31-46). New York: Taylor & Francis Group.

AUSTIN, M. U., LIAU, W.-S., BALMURUGAN, K., ASHOKKUMAR, B., SAID, H.

S., & LaMUNYON, C. W. (2010). Knockout of the folate transporter folt-1

causes germline and somatic defects in C. elegans. BMC Dev. Biol., pp.

10:46 doi: 10.1186/1471-213X-10-46.

BALAMURUGAN, K., ASHOKKUMAR, B., MOUSSAIF, M., SZE, J. Y., & SAID,

H. M. (2007). Cloning and functional characterization of a folate

transporter from the nematode Caenorhabditis elegans. Am. J. Physiol.

Cell Physiol., pp. 293:670-681 doi: 10.1152/ajpcell.00516.2006.

BARGMANN, C. I. (1993). Genetic and Cellular Analysis of Behavior in C.

elegans. Annu. Rev. Neurosci., pp. 16:47-71

doi:10.1146/annurev.ne.16.030193.000403.

BARSTEAD, R. (2001). Genome-wide RNAi. Curr. Opin. Chem. Biol., pp. 5:63-

66 doi:10.1016/S1367-5931(00)00173-3.

BARTEL, D. P. (2004). MicroRNAs: Genomics, Biogenesis, Mechanism, and

Function. Cell, pp. 116:281-297 doi:10.1016/S0092-8674(04)00045-5.

BASS, B. L. (2000). Double-Stranded RNA as a Template for Gene Silencing.

Cell, pp. 101:235-238 doi:10.1016/S0092-8674(02)71133-1.

BERNSTEIN, E., DENLI, A. M., & HANNON, G. J. (2001). The rest is silence.

RNA, pp. 7:1509-1521 doi:10.1017.S1355838201013462.

Page 123: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXIII

BOESE, Q., MARSHALL, W. S., & KHVOROVA, A. (2005). Design and

synthesis of small interfering RNA (siRNA) In Section two: Design,

synthesis of siRNAs. In K. APPASANI, RNA Interference Technology:

From Basic Science to Drug Development (pp. 103-117). Cambridge:

Cambridge University Press.

BOSHER, J. M., & LABOUESSE, M. (2000). RNA interference: genetic wand

and genetic watchdog. Nat. Cell Biol., pp. 2:E31-E36

doi:10.1038/35000102.

BRENNER, S. (1974). The Genetics of Caenorhabditis elegans. Genetics, pp.

77:71-94.

BÜRGLIN, T. R., LOBOS, E., & BLAXTER, M. L. (1998). Caenorhabditis

elegans as a model for parasitic nematodes. Int. J. Parasitol., pp. 28:395-

411 doi:10.1016/S0020-7519(97)00208-7.

CAPLEN, N. J., PARRISH, S., IMANI, F., FIRE, A., & MORGAN, R. A. (2001).

Specific inhibition of gene expression by small double-stranded RNAs in

invertebrate and vertebrate systems. PNAS, pp. 98:9742-9747

doi:10.1073/pnas.171251798.

CATALANOTTO, C., AZZALIN, G., MACINO, G., & COGONI, C. (2000).

Transcription: Gene silencing in worms and fungi. Nature, p. 404:245

doi:10.1038/35005169.

CERUTTI, H. (2003). RNA interference: traveling in the cell and gaining

functions? Trends Genet., pp. 19:39-46 doi:10.1016/S0168-

9525(02)00010-0.

CHANG, H. Y., WANG, N. N., & CHI, J.-T. (2005). High throughout RNA

Interference In Section seven: High-throughput genome-wide RNAi

analysis. In K. APPASANI, RNA Interference Technology: From Basic

Science to Drug Development (pp. 470-479). Cambridge: Cambridge

University Press.

Page 124: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXIV

CHI, J.-T., CHANG, H. Y., WANG, N. N., CHANG, D. S., DUNPHY, N., &

BROWN, P. O. (2003). Genomewide view of gene silencing by small

interfering RNAs. PNAS, pp. 11:6343-6346

doi:10.1073/pnas.1037853100.

CLAYTON, J. (2004). RNA interference: The silent treatment. Nature, pp.

431:599-605 doi:10.1038/431599a.

COGONI, C., & MACINO, G. (2000). Post-transcriptional gene silencing across

kingdoms. Curr. Opin. Genet. Dev., pp. 10:638-643 doi:10.1016/S0959-

437X(00)00134-9.

COUZIN, J. (2002). Small RNAs Make Big Splash. Science, pp. 298:2296-2297

doi:10.1126/science.298.5602.2296.

COUZIN, J. (2004). RNAi Shows Cracks in Its Armor. Science, pp. 306:1124-

1125 doi:10.1126/science.306.5699.1124.

DUDLEY, N. R., AMIN, A. Z., & GOLDSTEIN, B. (2005). Genes required for

RNA Interference In Section one: Basic RNAi, siRNA, microRNAs and

gene-silencing mechanisms. In K. APPASANI, RNA Interference

Technology: From Basic Science to Drug Development (pp. 55-68).

Cambridge: Cambridge University Press.

DYKXHOORN, D. M., & LIEBERMAN, J. (2005). The Silent Revolution: RNA

Interference as Basic Biology, Research Tool, and Therapeutic. Annu.

Rev. Med., pp. 56:401-423 doi:10.1146/annurev.med.56.082103.104606.

DYKXHOORN, D. M., NOVINA, C. D., & SHARP, P. A. (2003). Killing the

Messenger: Short RNAs that Silence Gene Expression. Nat. Rev. Mol.

Cell Biol., pp. 4:457-467 doi:10.1038/nrm1129.

EIFERT, C., KOURTIDIS, A., & CONKLIN, D. S. (2008). RNAi libraries in

dissecting molecular pathways of the human cell. In M. LATTERICH,

RNAi (pp. 47-64). New York: Taylor & Francis Group.

Page 125: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXV

ELBASHIR, S. M., HARBORTH, J., LENDECKEL, W., YALCIN, A., WEBER, K.,

& TUSCHL, T. (2001a). Duplexes of 21-nucleotide RNAs mediate RNA

interference in cultured mammalian cells. Nature, pp. 411:494-498

doi:10.1038/35078107.

ELBASHIR, S. M., LENDECKEL, W., & TUSCHL, T. (2001b). RNA interference

is mediated by 21- and 22-nucleotide RNAs. Genes Dev., pp. 15:188-200

doi:10.1101/gad.862301.

FIRE, A. (1999). RNA-triggered gene silencing. Trends Genet., pp. 15:358-363

doi:10.1016/S0168-9525(99)01818-1.

FIRE, A. (2007). Gene silencing by double-stranded RNA (Nobel Lecture).

Angew. Chem. Int. Ed. Engl., pp. 46:6966-6984

doi:10.1002/anie.200701979.

FIRE, A., XU, S., MONTGOMERY, M. K., KOSTAS, S. A., DRIVER, S. E., &

MELLO, C. C. (1998). Potent and specific genetic interference by double-

stranded RNA in Caenorhabditis elegans. Nature, pp. 391:806-811

doi:10.1038/35888.

FISCHER, S. E. (2010). Small RNA-mediated gene silencing pathways in C.

elegans. Int. J. Biochem. Cell Biol., pp. 42:1306-1315

doi:10.1016/j.biocel.2010.03.006.

FRASER, A. G., & AHRINGER, J. (2003). Total Interference: Genome-wide

RNAi Screens in C. elegans. In G. J. HANNON, RNAi A Guide to Gene

Silencing (pp. 233-242). New York: Cold Spring Harbor Laboratory

Press.

FRASER, A. G., KAMATH, R. S., ZIPPERLEN, P., MARTINEZ-CAMPOS, M.,

SOHRMANN, M., & AHRINGER, J. (2000). Functional genomic analysis

of C. elegans chromosome I by systematic RNA interference. Nature, pp.

408:325-330 doi:10.1038/35042517.

Page 126: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXVI

GOLDEN, A., & O'CONNELL, K. F. (2007). Silence is Golden: Combining RNAi

and live cell imaging to study cell cycle regulatory genes during

Caenorhabditis elegans development. Methods, pp. 41:190-197

doi:10.1016/j.ymeth.2006.07.029.

GRISHOK, A. (2005a). RNAi beginnings. Overview of the pathway in C.

elegans In Section one: Basic RNAi, siRNA, microRNAs and gene-

silencing mechanisms. In K. APPASANI, RNA Interference Technology:

from basic science to drug development (pp. 17-28). Cambridge:

Cambridge University Press.

GRISHOK, A. (2005b). RNAi mechanisms in Caenorhabditis elegans. FEBS

Lett., pp. 579:5932-5939 doi:10.1016/j.febslet.2005.08.001.

GRISHOK, A., PASQUINELLI, A. E., CONTE, D., & et al. (2001). Genes and

Mechanisms Related to RNA Interference Regulate Expression of the

Small Temporal RNAs that Control C. elegans Developmental Timing.

Cell, pp. 106:23:34 doi:10.1016/S0092-8674(01)00431-7.

GRISHOK, A., TABARA, H., & MELLO, C. C. (2000). Genetic Requirements for

Inheritance of RNAi in C. elegans. Science, pp. 287:2494-2497

doi:10.1126/science.287.5462.2494.

GROSSHANS, H., & SLACK, F. J. (2002). Micro-RNAs: small is plentiful. JCB,

pp. 156:17-22 doi:10.1083/jcb.200111033.

HADWIGER, P., & VORNLOCHER, H.-P. (2005). Chemical modifications to

achieve increased stability and sensitive detection of siRNA In Section

three: Vector development and in vivo, in vitro and in ovo delivery

methods. In K. APPASANI, RNA Interference Technology: From Basic

Science to Drug Development (pp. 194-206). Cambridge: Cambridge

University Press.

HALL, D. H., & ALTUN, Z. F. (2008). C. elegans Atlas. New York: Cold Spring

Harbor Laboratory Press.

Page 127: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXVII

HAMMOND, S. M., BERNSTEIN, E., BEACH, D., & HANNON, G. J. (2000). An

RNA-directed nuclease mediated post-transcriptional gene silencing in

Drosophila cells. Nature, pp. 404:293-296 doi:10.1038/35005107.

HANNON, G. J. (2002). RNA interference. Nature, pp. 418:244-251

doi:10.1038/418244a.

HANNON, G. J., & ROSSI, J. J. (2004). Unlocking the potential of the human

genome with RNA interference. Nature, pp. 431:371-378

doi:10.1038/nature02870.

HE, L., & HANNON, G. H. (2004). MicroRNAs: Small RNAs with a Big Role in

Gene Regulation. Nat. Rev. Genet., pp. 5:522-531 doi:10.1038/nrg1379.

HOPE, I. A. (1999 reprinted 2005). Background on Caenorhabditis elegans. In I.

A. HOPE, C. elegans: A practical approach (pp. 1-15). New York: Oxford

University Press.

HOWARD, K. (2003). Unlocking the money-making potential of RNAi. Nat.

Biotechnol., pp. 12:1441-1446 doi:10.1038/nbt1203-1441.

HUNTER, C. P. (1999). A touch of elegance with RNAi. Curr. Biol., pp. 9:R440-

R442 doi:10.1016/S0960-9822(99)80276-0.

HUNTER, C. P. (2000). Gene silencing: Shrinking the black box of RNAi. Curr.

Biol., pp. 10:R137-R140 doi:10.1016/S0960-9822(00)00325-0.

HUTVÀGNER, G., & SIMARD, M. J. (2008). Argonaute proteins: key players in

RNA silencing. Nat. Rev. Mol. Cell Biol., pp. 9:22-32

doi:10.1038/nrm2321.

HUTVÁGNER, G., & ZAMORE, P. D. (2002). RNAi: nature abhors a double-

strand. Curr. Opin. Genet. Dev., pp. 12:225-232 doi:10.1016/S0959-

437X(02)00290-3.

Page 128: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXVIII

JACKSON, A. L., & LINSLEY, P. S. (2004). Noise amidst the silence: off-target

effects of siRNAs? Trends Genet., pp. 20:521-524

doi:10.1016/j.tig.2004.08.006.

JAGLA, B., & AULNER, N. (2008). RNAi reagent design. In M. LATTERICH,

RNAi (pp. 3-20). New York: Taylor & Francis Group.

JOHNSON, N. M., FRITZ, J.-A., & BEHM, C. A. (2009). RNAi in Caenorhabditis

elegans In Part 2: RNA interference in animals. In T. DORAN, & C.

HELLIWELL, RNA Interference: Methods for Plants & Animals (pp. 85-

113). Cambridge: CABI.

KAMATH, R. S., & AHRINGER, J. (2003). Genome-wide RNAi screening in

Caenorhabditis elegans. Methods, pp. 30:313-321 doi:10.1016/S1046-

2023(03)00050-1.

KAMATH, R. S., FRASER, A. G., DONG, Y., & et al. (2003). Systemic

functional analysis of the Caenorhabditis elegans genome using RNAi.

Nature, pp. 421:231-237 doi:10.1038/nature01278.

KAMATH, R. S., MARTINEZ-CAMPOS, M., ZIPPERLEN, P., FRASER, A. G., &

AHRINGER, J. (2000). Effectiveness of specific RNA-mediated

interference through ingested double-stranded RNA in Caenorhabditis

elegans. Genome Biol., pp. 2:(I)0002.1-0002.10 doi:10.1186/gb-2000-2-

1-research0002.

KENNEDY, S., WANG, D., & RUVKUN, G. (2004). A conserved siRNA-

degrading RNase negatively regulates RNA interference in C. elegans.

Nature, pp. 427:645-649 doi:10.1038/nature02302.

KENT, O. A., & MacMILLAN, A. M. (2004). RNAi: running interference for the

cell. Org. Biomol. Chem., pp. 2:1957-1961 doi:10.1039/B404932M.

KETTING, R. F., & PLASTERK, R. H. (2000). A genetic link between co-

suppression and RNA interference in C. elegans. Nature, pp. 404:296-

298 doi:10.1038/35005113.

Page 129: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXIX

KETTING, R. F., FISCHER, S. E., BERNSTEIN, E., SIJEN, T., HANNON, G. J.,

& PLASTERK, R. H. (2001). Dicer functions in RNA interference and in

synthesis of small RNA involved in developmental timing in C. elegans.

Genes Dev., pp. 15:2654-2659 doi:10.1101/gad.927801.

KETTING, R. F., TIJSTERMAN, M., & PLASTERK, R. H. (2003). RNAi in

Caenorhabditis elegans. In G. J. HANNON, RNAi A Guide to Gene

Silencing (pp. 65-86). New York: Cold Spring Harbor Laboratory Press.

KIM, J. K., GABEL, H. W., KAMATH, R. S., & et al. (2005). Functional Genomic

Analysis of RNA Interference in C. elegans. Science, pp. 308:1164-1167

doi:10.1126/science.1109267.

KIM, N. V. (2005). Small RNAs: Classification, Biogenesis, and Function.

Retrieved September 07, 2011, from Mol. Cells, 19:1-15:

http://www.narrykim.org/MoleCellReview2005.pdf.

KIM, S. K. (2001). Functional genomics: The worm scores a knockout. Curr.

Biol., pp. 11:R85-87 doi:10.1016/S0960-9822(01)00035-5.

KOOTER, J. M., MATZKE, M. A., & MEYER, P. (1999). Listening to the silent

genes: transgene silencing, gene regulation and pathogen control.

Trends Plant Sci., pp. 9:340-347 doi:10.1016/S1360-1385(99)01467-3.

KUMABARA, P. E., & COULSON, A. (2000). RNAi - Prospects for a General

Technique for Determining Gene Function. Parasitol. Today, pp. 8:347-

349 doi:10.1016/S0169-4758(00)01677-X.

KUWABARA, P. E., & O'NEIL, N. (2001). The use of functional genomics in C.

elegans for studying human development and disease. J. Inherit. Metab.

Dis., pp. 24:127-138 doi:10.1023/A:1010306731764.

LAGOS-QUINTANA, M., RAUHUT, R., LENDECKEL, W., & TUSCHL, T.

(2001). Identification of Novel Genes Coding for Small Expressed RNAs.

Science, pp. 294:853-858 doi:10.1126/science.1064921.

Page 130: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXX

LAI, E. C. (2003). microRNAs: Runts of the Genome Assert Themselves. Curr.

Biol., pp. 13:R925-R936 doi:10.1016/j.cub.2003.11.017.

LATHAM, K., PALLOTTA, V., FORD, L., & et al. (2005). Six methods of

inducing RNAi in mammalian cells In Section three: Vector development

and in vivo, in vitro and in ovo delivery methods. In K. APPASINI, RNA

Interference Technology: From Basic Science to Drug Development (pp.

147-160). Cambridge: Cambridge University Press.

LAU, N. C., LIM, L. P., WEINSTEIN, E. G., & BARTEL, D. P. (2001). An

Abundant Class of Tiny RNAs with Probable Regulatory Roles in

Caenorhabditis elegans. Science, pp. 294:858-862

doi:10.1126/science.1065062.

LEE, R. C., & AMBROS, V. (2001). An Extensive Class of Small RNAs in

Caenorhabditis elegans. Science, pp. 294:862-864

doi:10.1126/science.1065329.

LEE, R. C., HAMMELL, C. M., & AMBROS, V. (2006). Interacting endogenous

and exogenous RNAi pathways in Caenorhabditis elegans. RNA, pp.

12:589-597 doi:10.1261/rna.2231506.

LEE, Y., JEON, K., LEE, J.-T., KIM, S., & KIM, N. V. (2002). MicroRNA

maturation: stepwise processing and subcellular localization. EMBO J.,

pp. 21:4663-4670 doi:10.1093/emboj/cdf476.

LEUNG, R. K., & WHITTAKER, P. A. (2005). RNA interference: from gene

silencing to gene-specific therapeutics. Pharmacol. Ther., pp. 107:222-

239 doi:10.1016/j.pharmthera.2005.03.004.

LIM, L. P., LAU, N. C., WEINSTEIN, E. G., & et al. (2003). The microRNAs of

Caenorhabditis elegans. Genes Dev., pp. 17:991-1008

doi:10.1101/gad.1074403.

Page 131: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXXI

LIPPMAN, Z., & MARTIENSSEN, R. (2004). The role of RNA interference in

heterochromatic silencing. Nature, pp. 431:364-370

doi:10.1038/nature02875.

LU, P. Y., & WOODLE, M. C. (2005). Delivering siRNA in vivo for functional

genomics and novel therapeutics In Section five: Drug target validation.

In K. APPASANI, RNA Interference Technology: From Basic Science to

Drug Development (pp. 235-246). Cambridge: Cambridge University

Press.

MAEDA, I., KOHARA, Y., YAMAMOTO, M., & SUGIMOTO, A. (2001). Large-

scale analysis of gene function in Caenorhabditis elegans by high-

throughput RNAi. Curr. Biol., pp. 11:171-176 doi:10.1016/S0960-

9822(01)00052-5.

MAINE, E. M. (2001). RNAi As a Tool for Understanding Germline Development

in Caenorhabditis elegans: Uses and Cautions. Dev. Biol., pp. 239:177-

189 doi:10.1006/dbio.2001.0394.

MAO, Y., MELLO, C., LAMARQ, L., & et al. (2005). Viral delivery of shRNA In

Section three: Vector development and in vivo, in vitro and in ovo

delivery methods. In K. APPASANI, RNA Interference Technology: From

Basic Science to Drug Development (pp. 161-173). Cambridge:

Cambridge University Press.

MARTINEZ, J., PATKANIOWSKA, A., URLAUB, H., LÜHRMANN, R., &

TUSCHL, T. (2002). Single-Stranded Antisense siRNAs Guide Target

RNA Cleavage in RNAi. Cell, pp. 110:563-574 doi:10.1016/S0092-

8674(02)00908-X.

McMANUS, M. T., PETERSEN, C. P., HAINES, B. B., CHEN, J., & SHARP, P.

P. (2002). Gene silencing using micro-RNA designed hairpins. RNA, pp.

8:842-850 doi:10.1017.S1355838202024032.

MEISTER, G., & TUSCHL, T. (2004). Mechanisms of gene silencing by double-

stranded RNA. Nature, pp. 431:343-349 doi:10.1038/nature02873.

Page 132: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXXII

MELLO, C. C. (2007). Return to the RNAi World: Rethinking Gene Expression

and Evolution (Nobel Lecture). Angew. Chem. Int. Ed. Engl., pp.

46:6985-6994 doi:10.1002/anie.200701713.

MELLO, C. C., & CONTE JR., D. (2004). Revealing the world of RNA

interference. Nature, pp. 431:338-342 doi:10.1038/nature02872.

MILHAVET, O., GARY, D. S., & MATTSON, M. P. (2003). RNA Interference in

Biology and Medicine. Pharmacol. Rev., pp. 55:629-648

doi:10.1124/pr.55.4.1.

MITTAL, V. (2004). Improving The Efficiency Of RNA Interference In Mammals.

Nat. Rev. Genet., pp. 5:355-365 doi:10.1038/nrg1323.

MONOHARAN, M. (2004). RNA interference and chemically modified small

interfering RNAs. Curr. Opin. Chem. Biol., pp. 8:570-579

doi:10.1016/j.cbpa.2004.10.007.

MONTGOMERY, M. K., & FIRE, A. (1998). Double-stranded RNA as a mediator

in sequence-specific genetic silencing and co-suppression. Trends

Genet., pp. 14:255-258 doi:10.1016/S0168-9525(98)01510-8.

MONTGOMERY, M. K., XU, S., & FIRE, A. (1998). RNA as a target of double-

stranded RNA-mediated genetic interference in Caenorhabditis elegans.

PNAS, pp. 95:15502-15507 doi:10.1073/pnas.95.26.15502.

MOSS, E. G. (2002). MicroRNAs: Hidden in the Genome. Curr. Biol., pp.

12:R138-140 doi:10.1016/S0960-9822(02)00708-X.

MURCHISON, E. P., & HANNON, G. J. (2004). miRNAs on the move: miRNA

biogenesis and the RNAi machinery. Curr. Opin. Cell Biol., pp. 16:223-

229 doi:10.1016/j.ceb.2004.04.003.

MYERS, J. W., & FERREL, J. (2005). Dicer in RNAi: Its roles in vivo and utility

in vitro In Section one: Basic RNAi, siRNA, microRNA and gene-silencing

mechanisms. In K. APPASANI, RNA Interference Technology: From

Page 133: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXXIII

Basic Science to Drug Development (pp. 29-54). Cambridge: Cambridge

University Press.

NAPOLI, C., LEMIEUX, C., & JORGENSEN, R. (1990). Introduction of a

Chimeric Chalcone Synthase Gene into Petunia Results in Reversible

Co-Suppression of Homologous Genes in trans. The Plant Cell, pp.

2:279-289 doi:10.1105/tpc.2.4.279.

NIRENBERG, M. (2005). Foreword. In K. APPASANI, RNA Interference

Technology: From Basic Science to Drug Development (pp. XIII-XIV).

Cambridge: Cambridge University Press.

NISHIKURA, K. (2001). A Short Primer on RNAi: RNA-Directed RNA

Polymerase Acts as a Key Catalyst. Cell, pp. 107:415-418

doi:10.1016/S0092-8674(01)00581-5.

NOVINA, C. D., & SHARP, P. A. (2004). The RNAi revolution. Nature, pp.

430:161-164 doi:10.1038/430161a.

NYKÄNEN, A., HALEY, B., & ZAMORE, P. D. (2001). ATP Requirements and

Small Interfering RNA Structure in the RNA Interference Pathway. Cell,

pp. 107:309-321 doi:10.1016/S0092-8674(01)00547-5.

PADDISON, P. J., & HANNON, G. J. (2002). RNA interference: the new

somatic cell genetics? Cancer Cell, pp. 2:17-23 doi:10.1016/S1535-

6108(02)00092-2.

PADDISON, P. J., CAUDY, A. A., BERNSTEIN, E., HANNON, G. J., &

CONKLIN, D. S. (2002). Short hairpin RNAs (shRNAs) induce sequence-

specific silencing in mammalian cells. Genes Dev., pp. 16:948-958

doi:10.1101/gad.981002.

PAROO, Z., & COREY, D. R. (2004). Challenges for RNAi in vivo. Trends

Biotechnol., pp. 22:390-394 doi:10.1016/j.tibtech.2004.06.004.

PARRISH, S., FLEENOR, J., XU, S., MELLO, C., & FIRE, A. (2000). Functional

Anatomy of a dsRNA Trigger: Differential Requirement for the Two

Page 134: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXXIV

Trigger Strands in RNA Interference. Mol. Cell, pp. 6:1077-1087

doi:10.1016/S1097-2765(00)00106-4.

PETERS, L., & MEISTER, G. (2007). Argonaute Proteins: Mediators of RNA

Silencing. Mol. Cell, pp. 26:611-623 doi:10.1016/j.molcel.2007.05.001.

PLASTERK, R. H. (2002). RNA Silencing: The Genome's Immune System.

Science, pp. 296:1263-1265 doi:10.1126/science.1072148.

POULIN, G., NANDAKUMAR, R., & AHRINGER, J. (2004). Genome-wide RNAi

screens in Caenorhabditis elegans: impact on cancer research.

Oncogene, pp. 23:8340-8345 doi:10.1038/sj.onc.1208010.

QIU, S., ADEMA, C. M., & LANE, T. (2005). A computational study of off-target

effects of RNA interference. Nucl. Acids Res., pp. 6:1834-1847

doi:10.1093/nar/gki324.

QUERFELLI, O. (2008). RNAi - a chemical perspective. In M. LATTERICH,

RNAi (pp. 21-30). New York: Taylor & Francis Group.

RAMOS-NINO, M. E., & MOSSMAN, B. T. (2005). Microarray analysis and RNA

silencing to determine genes functionally important in mesothelioma In

Section seven: High-throughput genome-wide RNAi analysis. In K.

APPASANI, RNA Interference Technology: From Basic Science to Drug

Development (pp. 447-469). Cambridge: Cambridge University Press.

SCHEPERS, U. (2005). RNA Interference in Practice: Principles, Basics, and

Methods for Gene Silencing in C. elegans, Drosophila, and Mammals.

Weinheim: WILEY-VCH Verlag GmbH & Co. KGaA.

SCHERER, L., & ROSSI, J. J. (2005). RNAi applications in living animal

systems In Section six: Therapeutic and drug development. In K.

APPASANI, RNA Interference Technology: From Basic Science to Drug

Development (pp. 406-415). Cambridge: Cambridge University Press.

Page 135: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXXV

SCHLEE, M., HORNUNG, V., & HARTMANN, G. (2006). siRNA and isRNA:

Two Edges of One Sword. Mol. Ther., pp. 14:463-470

doi:10.1016/j.ymthe.2006.06.001.

SCHWARZ, D. S., HUTVÁGNER, G., DU, T., XU, Z., ARONIN, N., & ZAMORE,

P. D. (2003). Asymmetry in the Assembly of the RNAi Enzyme Complex.

Cell, pp. 115:199-208 doi:10.1016/S0092-8674(03)00759-1.

SCHWARZ, E. M. (2005). Genomic classification of protein-coding gene

families. In WormBook, ed. The C. elegans Research Community.

doi:10.1895/wormbook.1.29.1: http://www.wormbook.org.

SEMIZAROV, D., FROST, L., SARTHY, A., KROEGER, P., HALBERT, D. H., &

FESIK, S. W. (2003). Specificity of short interfering RNA determined

through gene expression signatures. PNAS, pp. 11:6347-6352

doi:10.1073/pnas.1131959100.

SHAN, G. (2010). RNA interference as a gene knockdown technique. Int. J.

Biochem. Cell Biol., pp. 42:1243-1251 doi:10.1016/j.biocel.2009.04.023.

SHAN, G., LI, Y., ZHANG, J., & et al. (2008). A small molecule enhances RNA

interference and promotes microRNA processing. Nat. Biotechnol., pp.

26:933-940 doi:10.1038/nbt.1481.

SHARP, P. A. (1999). RNAi and double-strand RNA. Genes Dev., pp. 13:139-

141.

SHARP, P. A. (2001). RNA interference - 2001. Genes Dev., pp. 15:485-490

doi:10.1101/gad.880001.

SHI, Y. (2003). Mammalian RNAi for the masses. Trends Genet., pp. 19:9-12

doi:10.1016/S0168-9525(02)00005-7.

SIJEN, T., & KOOTER, J. M. (2000). Post-transcriptional gene silencing: RNAs

on the attack or on the defense? BioEssays, pp. 22:520-531

doi:10.1002/(SICI)1521-1878(200006)22:6<520::AID-BIES5>3.0.CO;2-

W.

Page 136: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXXVI

SIJEN, T., FLEENOR, J., SIMMER, F., & et al. (2001). On the Role of RNA

Amplification in dsRNA-Triggered Gene Silencing. Cell, pp. 107:465-476

doi:10.1016/S0092-8674(01)00576-1.

SIMMER, F., MOORMAN, C., van der LINDEN, A. M., & et al. (2003). Genome-

Wide RNAi of C. elegans Using the Hypersensitive rrf-3 Strain Reveals

Novel Gene Functions. PLoS Biol., pp. 1:077-084

doi:10.1371/journal.pbio.0000012.

SIMMER, F., TIJSTERMAN, M., PARRISH, S., & et al. (2002). Loss of the

Putative RNA-Directed RNA Polymerase RRF-3 Makes C. elegans

Hypersensitive to RNAi. Curr. Biol., pp. 12:1317-1319

doi:10.1016/S0960-9822(02)01041-2.

SOHAIL, M., & DORAN, G. (2005). Enzymatic production of small interfering

RNAs In Section two: Design, synthesis of siRNAs. In K. APPASANI,

RNA Interference Technology: From Basic Science to Drug Development

(pp. 139-143). Cambridge: Cambridge University Press.

STEPHENS, K. E., ZUGASTI, O., O'NEIL, N. J., & KUWABARA, P. E. (2005).

Practical applications of RNAi in C. elegans In Section four: Gene

silencing in model organisms. In K. APPASINI, RNA Interference

Technology: From Basic Science to Drug Development (pp. 235-246).

Cambridge: Cambridge University Press.

STIERNAGLE, T. (1999 reprinted 2005). Maintenance of C. elegans. In I. A.

HOPE, C. elegans: A practical approach (pp. 51-67). New York: Oxford

University Press.

SUGIMOTO, A. (2004). High-throughput RNAi in Caenorhabditis elegans:

genome-wide screens and functional genomics. Differentiation, pp.

72:81-91 doi:10.1111/j.1432-0436.2004.07202004.x.

SUGIMOTO, A. (2005). High-throughput RNAi by soaking in Caenorhabditis

elegans In Section seven: High-throughput genome-wide RNAi analysis.

In K. APPASANI, RNA Interference Technology: From Basic Science to

Page 137: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXXVII

Drug Development (pp. 419-432). Cambridge: Cambridge University

Press.

TABARA, H., GRISHOK, A., & MELLO, C. C. (1998). RNAi in C. elegans:

Soaking in the Genome Sequence. Science, pp. 282:430-431

doi:10.1126/science.282.5388.430.

TABARA, H., SARKISSIAN, M., KELLY, W. G., & et al. (1999). The rde-1 Gene,

RNA Interference, and Transposon Silencing in C. elegans. Cell, pp.

99:123-132 doi:10.1016/S0092-8674(00)81644-X.

TAFT, R. J., GLAZOV, E. A., LASSMANN, T., HAYASHIZAKI, Y., CARNINCI,

P., & MATTICK, J. S. (2009). Small RNAs derived from snoRNAs. RNA,

pp. 15:1233-1240 doi:10.1261/rna.1528909.

TIJSTERMAN, M., KETTING, R. F., & PLASTERK, R. H. (2002). The Genetics

Of RNA Silencing. Annu. Rev. Genet., pp. 36:489-519

doi:10.1146/annurev.genet.36.043002.091619.

TIJSTERMAN, M., MAY, R. C., SIMMER, F., OKIHARA, K. L., & PLASTERK,

R. H. (2004). Genes Required for Systemic RNA Interference in

Caenorhabditis elegans. Curr. Biol., pp. 14:111-116

doi:10.1016/j.cub.2003.12.029.

TIMMONS, L., & FIRE, A. (1998). Specific interference by ingested dsRNA.

Nature, p. 395:854 doi:10.1038/27579.

TIMMONS, L., COURT, D. L., & FIRE, A. (2001). Ingestion of bacterially

expressed dsRNAs can produce specific and potent genetic interference

in Caenorhabditis elegans. Gene, pp. 263:103-112 doi:10.1016/S0378-

1119(00)00579-5.

TIMMONS, L., TABARA, H., MELLO, C. C., & FIRE, A. Z. (2003). Inducible

Systemic RNA Silencing in Caenorhabditis elegans. MBoC, pp. 14:2972-

2983 doi:10.1091/mbc.E03-01-0858.

Page 138: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXXVIII

TUSCHL, T. (2001). RNA Interference and Small Interfering RNAs.

ChemBioChem, pp. 2:239-245 doi:10.1002/1439-

7633(20010401)2:4<239::AID-CBIC239>3.0.CO;2-R.

TUSCHL, T. (2002). Expanding small RNA interference. Nat. Biotechnol., pp.

20:446-448 doi:10.1038/nbt0502-446.

TUSCHL, T., & BORKHARDT, A. (2002). Small Interfering RNAs: A

Revolutionary Tool for the Analysis of Gene Function and Gene Therapy.

Mol. Interventions, pp. 2:158-167 doi:10.1124/mi.2.3.158.

TUSCHL, T., ZAMORE, P. D., LEHMANN, R., BARTEL, D. P., & SHARP, P. A.

(1999). Targeted mRNA degradation by double-stranded RNA in vitro.

Genes Dev., pp. 13:3191-3197.

University Of Minnesota. (2011). Caenorhabditis Genetics Center. Retrieved

February 12, 2011, from http://www.cbs.umn.edu/CGC/.

VANHECKE, D., & JANITZ, M. (2005). Functional genomics using high-

throughput RNA interference. Drug Discov. Today, pp. 10:205-212

doi:10.1016/S1359-6446(04)03352-5.

VASTENHOUW, N. L., & PLASTERK, R. H. (2004). RNAi protects the

Caenorhabditis elegans germline against transposition. Trends Genet.,

pp. 20:314-319 doi:10.1016/j.tig.2004.04.011.

WADHWA, R., KAUL, S. C., MIYAGISHI, M., & TAIRA, K. (2004). Know-how of

RNA interference and its applications in research and therapy. Rev. Mut.

Res., pp. 567:71-84 doi:10.1016/j.mrrev.2004.06.002.

WIANNY, F., & ZERNICKA-GOETZ, M. (2000). Specific interference with gene

function by double-stranded RNA in early mouse development. Nat. Cell

Biol., pp. 2:70-75 doi:10.1038/35000016.

WILKINS, C., DISHONGH, R., MOORE, S. C., WHITT, M. A., CHOW, M., &

MACHACA, K. (2005). RNA interference is an antiviral defence

Page 139: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XXXIX

mechanism in Caenorhabditis elegans. Nature, pp. 436:1044-1047

doi:10.1038/nature03957.

WINSTON, W. M., MOLODOWITCH, C., & HUNTER, C. P. (2002). Systemic

RNAi in C. elegans Requires the Putative Transmembrane Protein SID-

1. Science, pp. 295:2456-2459 doi:10.1126/science.1068836.

WormBook, e. (2010). WormBook, The Online Review of C. elegans Biology.

Retrieved February 12, 2011, from http://wormbook.org/.

ZAMORE, P. D. (2001). RNA interference: listening to the sound of silence.

Nature Structural Biology, pp. 8:746-750 doi:10.1038/nsb0901-746.

ZAMORE, P. D. (2002). Ancient Pathways Programmed by Small RNAs.

Science, pp. 296:1265-1269 doi:10.1126/science.1072457.

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, pp. 101:25-33 doi:10.1016/S0092-

8674(00)80620-0.

Page 140: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 141: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XLI

VIII. CURRICULUM VITAE

Family and first name Liberda Julia Katharina

Address Ziegelofengasse 41/30 1050 Wien

Phone +43 669 114 171 72

E-Mail [email protected]

Nationality Austrian

Date of Birth 13. February 1984

WORK EXPERIENCE

07.1999 to 12.2009 Jacoby Pharmazeutika AG, Hallein (pharmaceutical concern)

Traineeship Partime job in accounting department and merchandise warehouse

01.2006 to 02.2009 Apotheke zur Kaiserkrone, Wien (pharmacy Temporary employment at the accounting department

07.2005 to 06.2008 Österreichisches Verkehrsbüro AG / Verkehrsbüro Kulinarik GmbH / Kulinarik Gastronomie GmbH, Wien Catering at various sports and concert events

06.2007 to 08.2007 Salzburg Schokoladen GmbH, Grödig

Traineeship at the department of quality control

07.2006 to 08.2006 St. Ehrhard Apotheke, Salzburg (pharmacy)

Traineeship, Duties: bottling of pharmaceuticals and other substances like teas and herbs, production of drug specialties of the house like ointments

09.2006 Nelson Mandela Academic Hospital, Mthatha, Südafrika

Traineeship as dietician

PERSONAL

INFORMATION

Page 142: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in

XLII

EDUCATION

05.2010 Berlitz Language School, Phoenix, USA Advanced level training

2002 to date Studies of Nutritional Sciences at the Department of Nutritional Sciences, University of Vienna Master degree Master thesis: The RNA-Interference mechanism in molecular research in general, detailed in the model organism Caenorhabditis elegans as well as in terms of nutritional research

1994 to 2002

High School Bundesrealgymnasium Hallein Graduated in top 10%

1990 to 1994 Elementary School Hallein Burgfried

PERSONAL SKILLS

Native Language German

Other Languages English (advanced) Spanish (spoken and written)

IT-Knowledge Common software programs (MS Office)

working experience with kids

Leader of 6 to 14 year old girl and boy scouts

Travel Various trips through Europe, North America, India, and South Africa

Driver License for cars

Hobbies Reading, Hiking, Movie theater Cooking, Skiing and Piano playing

Page 143: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in
Page 144: The RNA interference mechanism in molecular …othes.univie.ac.at/18421/1/2012-02-10_0207600.pdf2012/02/10  · DIPLOMARBEIT The RNA interference mechanism in molecular research in