Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống
1
/ 171 trang
THÔNG TIN TÀI LIỆU
Thông tin cơ bản
Định dạng
Số trang
171
Dung lượng
3 MB
Nội dung
ELUCIDATING THE ROLE OF AUTOPHAGY IN
ZEBRAFISH MODELS OF LIVER CANCER
SIM HUEY FEN, TINA
(B.Sc.(Hons.)), NUS
NATIONAL UNIVERSITY OF SINGAPORE
2011
ELUCIDATING THE ROLE OF AUTOPHAGY IN
ZEBRAFISH MODELS OF LIVER CANCER
SIM HUEY FEN, TINA
(B.Sc.(Hons.)), NUS
A THESIS SUBMITTED
FOR THE DEGREE OF MASTER OF SCIENCE
DEPARTMENT OF BIOLOGICAL SCIENCES
NATIONAL UNIVERSITY OF SINGAPORE
2011
Acknowledgements
Acknowledgements
I would like to take this opportunity to express my greatest gratitude to my
honorific supervisors, Professor Gong Zhiyuan (Department of Biological Sciences,
Faculty of Science, NUS), and Associate Professor Shen Han Ming (Department
of Epidemiology and Public Health, Yong Loo Lin School of Medicine, NUS) for
taking me under their wing, as well as for their patience, and invaluable advice and
encouragement offered throughout the entirety of my research. I would also like to
express my gratitude to fellow laboratory mates from Prof Gong’s laboratory, both
past and present, namely, Weiling, Grace, Li Zhen, Zhou Li, Cai Xia, Anh Tuan,
Lili, Sahar, Xiao Qian, Hongyan, Xiaoyan, Shen Yuan, Ti Weng, Yin Ao,
Myintzu, Yan Tie, Huiqing and Zhengyuan for their advice and encouragement.
Special thanks to Jianzhou for the engaging brainstorming sessions and assistance
rendered during my research; and Lora for making my stay in the lab a very
memorable one. I also appreciate the favours from the members of A/P Shen’s
laboratory, especially Zhou Jing, even though I am not a full member of the
laboratory. I would also like to take this opportunity to thank staff from the fish
facility in the department for their efforts in maintaining quality fish stocks as well as
general assistance rendered. In addition, I would like to take this opportunity to show
my appreciation to the National University of Singapore for providing me with the
graduate research scholarship during these few years.
Last but not least, I would like to express my love and gratitude to my dearest
family for their love and unwavering support while I pursue my dreams and passion in
research.
i
Table of contents
Table of contents
Acknowledgements
i
Table of Contents
ii-iv
Summary
v-vi
List of Tables
vii
List of Figures
viii-x
List of Common Abbreviations
xi-xii
Introduction
1. The use of zebrafish as a model organism for the study of cancer
1.1 The use of zebrafish as a model organism in vertebrate developmental
biology studies
1.2 The use of zebrafish as a model organism in cancer studies
1.3 Generation of inducible Xmrk and cmyc driven liver cancer models
1.3.1 The use of inducible promoter systems to control transgene
expression
1.3.2 The use of transposable elements to generate transgenic
zebrafish
1.3.3 Xmrk and cmyc driven liver cancer models
2. An introduction to autophagy and its involvement in cancer
2.1 Autophagy as a basal and inducible cellular process
2.2 Molecular machinery of autophagy
2.3 Regulation of autophagy
2.4 The use of EGFP-LC3 and mRFP-EGFP-LC3 to study autophagy in
vivo
2.5 Autophagy in cancer
3. Rationale of current study
3.1 Establishing Tg(fabp10:egfp-lc3) and Tg(fabp10:mrfp-egfp-lc3)
transgenic zebrafish for studying autophagy
3.2 Establishing the role of autophagy in liver cancer
3.3 Objectives of study
Materials and Methods
1. Zebrafish care and maintenance and tissue collection
1.1 Zebrafish maintenance
1.2 Zebrafish spawning and embryo collection
1.3 Microinjection into one-cell stage embryos
1
2
2
4
7
7
8
9
11
11
13
18
23
25
29
29
30
31
33
34
34
34
35
ii
Table of contents
1.4 Larval screening and photo-documentation
1.5 Liver tissue collection from adult zebrafish
2. RNA applications
2.1 Total RNA isolation from embryos and larvae
2.2 RNA quantification
2.3 Ac mRNA preparation
3. DNA applications
3.1 Total DNA isolation
3.2 cDNA synthesis
3.3 Construction of plasmid DNA
3.3.1 Restriction endonuclease digestion of DNA
3.3.2 Recovery of DNA fragments from DNA gel
3.3.3 Ligation
3.3.4 Competent cell preparation
3.3.5 Transformation and re-transformation reactions
3.3.6 Colony screening
3.3.7 Bacterial cell culture and plasmid amplification
3.3.8 Plasmid isolation and purification
3.4 Polymerase chain reaction
3.4.1 Standard PCR
3.4.2 2-step reverse transcription polymerase chain reaction (2-step
RT-PCR)
3.5 DNA vectors
3.5.1 pLF3.0-EGFP
3.5.2 pEGFP-LC3
3.5.3 pmRFP-EGFP-LC3
3.5.4 pMDS6
3.5.5 pAc-SP6
4. Protein applications
4.1 Total protein isolation and extraction
4.2 Protein quantification
4.3 Protein detection
4.3.1 SDS-PAGE gel casting
4.3.2 Running SDS-PAGE
4.3.3 Protein transfer from PAGE gel to Polyvinylidene fluoride
(PVDF) membrane
4.3.4 Immunoblotting
4.3.5 Protein band detection
4.3.6 Calculating band intensity from immunoblots
5. Starvation and chemical treatments
5.1 Starvation treatments
5.2 Chemical treatments
5.3 Doxycycline induction of Xmrk and cmyc transgene expression
36
36
37
37
38
38
39
39
40
41
41
43
43
44
45
46
46
46
47
48
50
52
52
53
54
55
56
57
57
57
58
58
59
60
60
61
61
62
62
63
64
iii
Table of contents
6. External photo-documentation of adult treated transgenic fish
7. Cryosectioning
7.1Cryostat sections of larvae
7.2 Cryostat sections of adult tissues
7.3 Hoechst staining
8. Confocal imaging
8.1 Live confocal imaging
8.2 Confocal imaging of sections
9. Bioinformatics and sequence analyses
10. Analyses of liver transcriptome data by deep sequencing
64
65
65
66
66
66
66
67
67
68
Results
1. Conserved autophagic machinery in zebrafish
1.1 Atg gene expression in zebrafish
1.2 Conservation of Atg proteins in zebrafish
1.3 Expression of autophagic genes and proteins in zebrafish
1.4 Conserved autophagic machinery in zebrafish
2. Establishment and characterisation of Tg(fabp10:egfp-lc3)
transgenic line
2.1 Establishing Tg(fabp10:egfp-lc3) transgenic line
2.2 Characterisation of Tg(fabp10:egfp-lc3) transgenic line
3. Establishment and characterisation of Tg(fabp10:mrfp-egfp-lc3)
transgenic line
3.1 Establishing Tg(fabp10:mrfp-egfp-lc3) transgenic line
3.2 Characterisation of Tg(fabp10:mrfp-egfp-lc3) transgenic line
4. Establishing the role of autophagy in liver cancer
4.1 Preliminary data implicating autophagy inhibition in liver cancer
4.2 Establishing double transgenic lines with liver-specific oncogene overexpression and EGFP-LC3 expression to study the role of autophagy in
liver cancer
4.2.1 Tg(fabp10:TA; TRE:Xmrk; krt4:GFP)
4.2.2 Tg(fabp10:TA; TRE:cmyc; krt4:RFP)
69
70
70
76
77
81
85
85
93
99
99
104
108
110
112
115
116
Discussion
1. Conserved autophagic machinery in zebrafish
2. Establishment and characterisation of Tg(fabp10:egfp-lc3)
transgenic line
3. Establishment and characterisation of Tg(fabp10:mrfp-egfp-lc3)
transgenic line
4. Establishing the role of autophagy in liver cancer
5. Future work and conclusions
124
125
130
References
143
134
135
140
iv
Summary
Summary
Autophagy is an evolutionarily conserved cellular process that involves
autophagosomal sequestration of cytoplasm, long-lived proteins and organelles, and
subsequent lysosomal degradation, in response to cellular stress such as starvation. It
is also involved in the degradation of superfluous organelles, proteins and protein
aggregates. In addition to its main role in regulating basal cellular homeostasis, it has
also been implicated in a number of diseases. Presently, there are evidences
implicating autophagy induction and inhibition during carcinogenesis; however the
exact role of autophagy in carcinogenesis remains to be elucidated. With the zebrafish
gaining popularity as a model organism in the study of human diseases including
cancers, we aim to investigate the role of autophagy in liver cancer using our
established zebrafish liver cancer models. As little study is so far conducted on
autophagy in zebrafish, we first characterized autophagy in zebrafish, and found the
autophagic machinery and function are well-conserved between human and zebrafish.
We also established and characterized a transgenic line, Tg(fabp10:egfp-lc3), with
constitutively liver-specific egfp-lc3 expression, in order to visualize the autophagic
flux in the liver. We found that this transgenic line is able to produce EGFP-LC3
puncta under starvation and everolimus treatment, both processes inducing autophagy.
Preliminary studies using our liver cancer transgenic zebrafish with inducible
expression of cmyc or Xmrk oncogene in the liver suggested that autophagy may be
inhibited during liver carcinogenesis. To further study this phenomenon, we crossed
Tg(fabp10:egfp-lc3) with these oncogene transgenic lines in order to visualize the
autophagic flux during tumour initiation, progression and regression in vivo, and we
obtained evidences suggesting autophagy inhibition during liver carcinogenesis. We
further observed that autophagy may have been de-repressed during tumour
v
Summary
regression, although more studies need to be performed to validate our observations.
Finally, we also developed a transgenic line with constitutively liver-specific mrfpegfp-lc3 expression. This transgenic line was developed based on the concept of
lysosomal quenching of GFP fluorescence, with the mRFP-EGFP-LC3 reporter
labelling autophagosomes yellow and autolysosomes red, thus allowing autophagic
flux to be measured by comparing the ratio between yellow and red puncta. This will
be the first attempt in generating a transgenic organism expressing this novel reporter.
We hope that this second transgenic line will be useful and complement the first
transgenic line in elucidating autophagic flux during liver carcinogenesis as well as
other liver diseases.
vi
List of tables
List of tables
Table 1
Restriction enzymes used for sub-cloning
42
Table 2
Primers used in PCR
49
Table 3
Primers used in 2-step RT-PCR
51
Table 4
atg gene expression in the zebrafish
72
Table 5
Comparison of zebrafish and human Atg proteins
73
vii
List of figures
List of figures
Figure 1
Schematic illustration of the autophagy pathway and its core
machinery in mammalian cells.
17
Figure 2
Regulation of mammalian autophagy.
22
Figure 3
Autophagy in tumourigenesis and anti-cancer therapy.
28
Figure 4
Construct map of pLF3.0-EGFP.
52
Figure 5
Construct map of pEGFP-LC3.
53
Figure 6
Construct map of pmRFP-EGFP-LC3.
54
Figure 7
Construct map of pMDS6.
55
Figure 8
Construct map of pAC-SP6.
56
Figure 9
Conservation of LC3 protein in zebrafish.
74
Figure 10
Conservation of Atg5 protein in zebrafish.
75-76
Figure 11
Temporal expression patterns of lc3 and atg5 from 1 dpf to 5
dpf zebrafish larvae.
79
Figure 12
LC3-I and LC3-II protein levels from 1 dpf to 5 dpf
zebrafish larvae.
80
Figure 13
Autophagy induction by rapamycin.
83
Figure 14
Autophagy induction is necessary for larvae to survive
starvation.
84
Figure 15
The pDS-FABP10-EGFP-LC3 plasmid used
generation of Tg(fabp10:egfp-lc3) transgenic line.
the
86
Figure 16
Sequence identity exists between rat and zebrafish LC3
proteins.
87
Figure 17
Liver-specific EGFP-LC3 expression after microinjection of
pDS-FABP10-EGFP-LC3.
89
Figure 18
EGFP-LC3 expression in the progeny of the five founders.
90
in
viii
List of figures
Figure 19
EGFP-LC3 positive larvae in Line 1 harbours germline
transmission of the fabp10:egfp-lc3 expression cassette.
91-92
Figure 20
Autophagy induction via starvation.
Figure 21
EGFP-LC3 puncta formation in the presence of everolimus
and chloroquine.
Figure 22
LC3-I and LC3-II after chemical exposure for 48 hours.
Figure 23
The pDS-FABP10-mRFP-EGFP-LC3 plasmid used in the
generation of Tg(fabp10:mrfp-egfp-lc3) transgenic line.
100
Figure 24
Liver-specific
mRFP-EGFP-LC3
expression
after
microinjecting the pDS-FABP10-mRFP-EGFP-LC3 plasmid
into one-cell stage embryos.
101
Figure 25
Transgenic larvae harbour germline transmission of the
fabp10:mrfp-egfp-lc3 expression cassette.
102-103
Figure 26
Starvation induces autophagy.
Figure 27
Red and yellow puncta formation in the presence of
everolimus and chloroquine.
106-107
Figure 28
Over-expression of cmyc and Xmrk led to liver tumour
formation in stable transgenic zebrafish induced using 60
µg/ml doxycycline.
109
Figure 29
mTOR activation may cause autophagy inhibition in fish
with Xmrk over-expression.
111
Figure 30
Schematic showing the genotypes of progeny resulting from
crossing Tg(fabp10:TA; TRE:Xmrk; krt4:GFP) with
Tg(fabp10:egfp-lc3) and doxycycline exposure and control
groups.
113
Figure 31
Schedule for doxycycline exposure and sampling time
points.
114
Figure 32
Tumour progression manifested as increasingly severe gross
and cellular morphology in transgenic fish over-expressing
Xmrk.
118-119
95
96-97
98
105
ix
List of figures
Figure 33
Immunoblot analyses of Xmrk over-expressing fish showing
increased free EGFP.
120
Figure 34
Tumour progression in fish over-expressing cmyc,
manifesting as liver overgrowth and disruption to cell
morphology.
121-122
Figure 35
Immunoblot analyses if cmyc over-expressing fish showing
increased free EGFP and reduced p62 levels during tumour
regression.
123
x
List of common abbreviations
List of common abbreviations
4EBP1
Ac/Ds
AMP
AMPK
APS
Atg
ATP
BSA
cDNA
cmyc
CTP
CQ
DAPk
DEPC
DMSO
DNA
dNTP
DTT
Dox
dpf
EDTA
EGFP
EGFR
eIF2α
ENU
ER
ERK
EVE
FABP
Gly
GTP
HA
HCC
HRP
kb
kD
krt4
LB
LC3
MAPK
MCS
eukaryotic initiation factor 4E binding protein 1
Activator/Dissociation
adenosine monophosphate
5’-AMP-activated protein kinase
ammonium persulphate
autophagy related
adenosine triphosphate
bovine serum albumin
DNA complementary to RNA
cellular homologue of v-myc oncogene
cytidine triphosphate
chloroquine
death-associated protein kinase
diethyl pyrocarbonate
dimethyl sulfoxide
deoxyribonucleic acid
deoxyribonucleotide
dithiothreitol
doxycycline
days post fertilisation
ethylenediaminetetraacetic acid
enhanced green fluorescent protein
epidermal growth factor receptor
eukaryotic initiation factor 2 α
ethylnitrosourea
endoplasmic reticulum
extracellular signal-regulated kinase
everolimus
fatty acid binding protein
glycine
guanosine triphosphate
hepatocelluar adenoma
hepatocellular carcinoma
horse radish peroxidase
kilobase
kilo Dalton
keratin 4 promoter
Luria Bertani
light chain 3
mitogen activated kinase
multiple cloning site
xi
List of common abbreviations
mpf
mRFP
mRNA
mTORC1
NCBI
NTC
NTP
p-S6
PBS
PCR
PE
PFA
PI3K
PTEN
PVDF
RAP
RNA
rpm
RT-PCR
S6K
SDS-PAGE
T-PER
TBS
TEMED
tetR
TGF-α
TILLING
TOR
TPM
TRE
TSC
TTP
Ulk1
UTP
VEGF
VHL
Xmrk
ZFIN
months post fertilisation
monomeric red fluorescent protein
messenger RNA
mammalian TOR complex 1
National Centre for Biotechnology Information
no template control
nucleoside triphosphate
phosphorylated S6
phosphate buffered saline
polymerase chain reaction
phosphotidylethanolamine
paraformaldehyde
phosphatidylinositol 3-kinase
phosphatase and tensin homologue
polyvinylidene fluoride
rapamycin
ribonucleic acid
revolutions per minute
reverse-transcriptase PCR
p70 ribosomal S6 kinase
sodium dodecyl sulphate polyacrylamide gel electrophoresis
tissue protein extraction reagent
tris buffered saline
tetramethylethylenediamine
tetracycline repressor
transforming growth factor α
targeting-induced local lesions in genomes
target of rapamycin
transcript per million
tetracycline responsive element
tuberous sclerosis complex
thymidine triphosphate
Uncoordinated 51-like kinase
uridine 5’-triphosphate
vascular endothelial growth factor
von Hippel-Lindau
Xiphophorus melanoma receptor kinase gene
zebrafish information network
xii
Introduction
Introduction
1
Introduction
1. The use of zebrafish as a model organism for the study of cancer
1.1 The use of zebrafish as a model organism in vertebrate developmental
biology studies
The zebrafish (Danio rerio) is an excellent model organism for the study of
vertebrate developmental biology and genetics due to its superior physical attributes
when compared with rodent models. External fertilisation and development as well as
optically transparent embryos allow early developmental processes to be studied
easily under a dissecting microscope. In addition, traits such as high fecundity with
the ability of a mating pair producing hundreds of embryos per week and short
generation times of approximately three months further make possible the use of
zebrafish for genetics analyses. Moreover, its ease of breeding and small size facilitate
animal husbandry, and its low maintenance costs make zebrafish an ideal organism to
complement the rodents in developmental biology and genetics studies (Dooley and
Zon, 2000; Wixon, 2000).
In addition to its physical attributes, the popularity of zebrafish as an
experimental organism has also led to the development of various experimental
methods and techniques. Whole-mount in situ hybridisation and the use of anti-sense
morpholino knockdown in transparent embryos facilitate the study of developmental
processes in vivo. Forward genetic screens employing ethylnitrosourea (ENU)induced point mutations allow the zebrafish to be used for screening developmental
mutants and identifying genes involved in early development for further analyses
(Driever et al., 1996; Haffter et al., 1996). Although other model organisms such as
Drosophila and Caenorhabditis elegans are also amendable to large scale screens, the
absence of vertebrate-specific organs such as kidney and neural crest cells make them
inferior to the zebrafish in the study of specific developmental issues involving these
2
Introduction
organs. The zebrafish is also amendable to reverse genetics via an approach known as
targeting-induced local lesions in genomes, or TILLING, which is adapted from plant
genomics studies (McCallum et al., 2000). DNA from progeny derived from ENUmutagenised parents is sequenced to look for mutations that are present in a gene-ofinterest. Progeny identified to carry the desired mutations are then propagated to
maintain the line to study the function of the gene-of-interest. Furthermore,
fluorescent proteins can be expressed in specific organs in order to study their
development over time by placing the gene encoding fluorescent proteins under the
control of organ-specific promoters (Higashijima et al., 1997; Long et al., 1997). This
is especially useful in early development when the larvae are largely transparent
which facilitates the observation of the organ under study. This method will also
allow specific genes-of-interest to be expressed under the control of a specific
promoter to study their function.
Moreover, efforts have been made to map zebrafish genes onto chromosomes
by both genetic and physical mapping (Geisler et al., 1999; Hukriede et al., 1999;
Postlethwait et al., 1998). With advances in sequencing technologies, the zebrafish
genome sequence is also made available as well for positional cloning. In spite of the
large evolutionary distance between zebrafish and human, the syntenic relationship
between their genomes further allow the use of the zebrafish in the identification of
gene functions of unknown genes in both zebrafish and humans (Barbazuk et al.,
2000).
In all, these attributes of the zebrafish has positioned it to overcome the
shortcomings of mouse models as well as made it relevant in developmental biology
and genetics studies.
3
Introduction
1.2 The use of zebrafish as a model organism in cancer studies
Fish has been used to study cancer since the early 1900s (Stern and Zon, 2003).
Early studies involving Xiphophorus, or swordtail fish, ascertained the existence of an
oncogene which is the causative agent of malignant melanoma in the species
(Wittbrodt et al., 1989). The oncogene, now known as the Xiphophorus melanoma
receptor kinase gene or Xmrk, is homologous to the human Epidermal Growth Factor
(EGF) receptor, and is found to be masked by its tumour-suppressor at a separate
locus and studies have shown that it plays an important role in elucidating the
functional roles of receptor tyrosine kinases in tumourigenesis (Wittbrodt et al., 1989;
Wittbrodt et al., 1992). Over the years, other fishes such as the medaka (Oryzias
latipes) and zebrafish are also used in carcinogenicity testing (Law, 2001).
The zebrafish has been most appropriately used as a model organism for the
study of cancer (Amatruda et al., 2002; Feitsma and Cuppen, 2008; Stoletov and
Klemke, 2008). First used as a chemical carcinogenesis model, the zebrafish
developed
mainly
hepatic
neoplasms
after
exposure
to
the
carcinogen
diethylnitrosamine (Stanton, 1965). More recent studies further showed that the
zebrafish is capable of developing other types of neoplasms depending on the types of
chemical carcinogens used and developmental stage at which they were exposed to
(Spitsbergen et al., 2000a, b). Furthermore, it has been shown that the tumours that
developed from such exposure were very similar to human tumours at a histological
level, lending support to the use of zebrafish as a model to study chemically-induced
cancer (Amatruda et al., 2002; Spitsbergen et al., 2000a). Apart from histological
similarities between zebrafish and human tumours, comparisons between human and
zebrafish liver cancer gene signatures revealed significant conservation between the
two, suggesting the conservation of mechanisms that are involved in liver
4
Introduction
carcinogenesis between the two (Lam and Gong, 2006; Lam et al., 2006). The ease of
housing and processing zebrafish in large numbers for such studies enables these
studies to generate data with greater statistical powers than mammalian cancer studies
involving mice and rats (Stern and Zon, 2003). Despite the low cost and technical
simplicity of chemical carcinogenesis, the low incidence of tumour occurrence
coupled with late tumour onset and heterogeneity and variability of tumour genetic
background and location make chemical carcinogenesis a less popular means of
studying cancer in zebrafish.
As the zebrafish is amendable to both forward and reverse genetics in the
study of vertebrate developmental biology and genetics, these techniques can also be
adapted to study cancer, and discover and analyse existing and novel oncogenes and
tumour suppressors in zebrafish. Forward genetic screens were carried out to look for
mutants displaying phenotypes related to cancer, such as proliferation defects which
identified mutants with loss-of-function in bmyb and separase (Shepard et al., 2007;
Shepard et al., 2005). Tp53 mutants were also isolated in reverse genetic screens
utilising ENU-induced mutagenesis and were found to be involved in the
development of malignant peripheral nerve sheath tumours (Berghmans et al., 2005).
Xenotransplantation of mammalian cancer cells from various sources into
zebrafish of different developmental stages were also performed successfully with
excellent results. Such studies were useful in delineating the dynamics of
microtumour formation, angiogenesis and vasculature pruning, as well as cellular
invasion and metastasis in vivo (Feitsma and Cuppen, 2008; Stoletov and Klemke,
2008). Stoletov et al. studied how expression of RhoC and VEGF by human cancer
cells aid in their metastasis, with RhoC expression inducing an amoeboid-type of
invasion characterised by the formation of a rounded cell morphology and extensive
5
Introduction
and dynamic membrane blebbing and protrusions, and VEGF expression inducing
vessel permeabilisation and vessel remodelling to aid the intravasation of cancer cells
into the remodelled blood vessels (Stoletov et al., 2007).
The technique of generating transgenic zebrafish with the ability to control the
expression of various genes in the study of gene function can also be extended to the
study of oncogenes and tumour suppressors. Through the use of tissue-specific
promoters, one can control the expression of oncogenes or mutant tumour suppressors
in a tissue-specific manner. Transgenic zebrafish expressing gene fusions of these
genes-of-interest with fluorescent proteins can be generated in order to select stable
transgenic progeny, and more importantly, to facilitate in vivo monitoring and
imaging of cells expressing these gene fusions (Stoletov and Klemke, 2008).
Langenau et al. made use of this strategy to over-express human cmyc under the
control of the zebrafish rag2 promoter, and obtained transgenic zebrafish overexpressing cmyc in T-cells with the concomitant development of T-cell acute
lymphoblastic leukaemia (Langenau et al., 2003). Other transgenic zebrafish overexpressing activated BRAF, zebrafish bcl2 and activated human kRASG12D among
others were also developed and they provided ample opportunities for studying the
mechanisms of oncogene-driven carcinogenesis and the cross-talk among different
signalling pathways (Amatruda and Patton, 2008; Langenau et al., 2005b; Langenau
et al., 2007; Patton et al., 2005).
Moreover, these models are amendable to pharmacological testing and novel
drug discovery as the zebrafish embryos and larvae are permeable to many watersoluble compounds and they can be arrayed in a manner that facilitates highthroughput screening. Moreover, screens for compounds and chemicals that can exert
their effects in zebrafish larvae are more powerful compared to screens using cell
6
Introduction
lines as they are able to suppress the disease phenotype in a whole organism in vivo
and they do not harbour any target bias (Stern and Zon, 2003). Compounds that are
active in suppressing cancer phenotypes can be identified for further testing, as
evidenced by screens for anti-angiogenic compounds (Wang et al., 2010; Zhang et al.,
2009).
1.3 Generation of inducible Xmrk and cmyc driven liver cancer models
1.3.1 The use of inducible promoter systems to control transgene expression
Langenau et al. generated the first transgenic zebrafish expressing human
cmyc under the control of a zebrafish tissue-specific mitfa promoter which made for
an excellent model to study the mechanism of cmyc-driven T-cell acute lymphoblastic
leukaemia (Langenau et al., 2003). However, the authors found that the resultant
transgenic zebrafish showed rapid onset of cancer that displayed efficient invasion
and metastasis to other tissues and organs, leading to early lethality and requiring in
vitro fertilisation for line maintenance. To overcome this problem, the same authors
made use of the Cre/lox and heat shock promoter systems to control the expression of
the cmyc oncogene (Feng et al., 2007; Langenau et al., 2005a). Initially the Cre/lox
system displayed low disease penetrance when the conditional transgenic progeny
carrying germline insertion of the construct rag2-loxP-dsRED2-loxP-EGFP-mMyc
were injected with the Cre mRNA; with the complementation of the heat shock
promoter controlling Cre expression, a higher rate of recombination events took place
after heat shock which resulted in more progeny expressing the rag2-EGFP-mMyc
cassette and thus, developing T-cell acute lymphoblastic leukaemia. Thus the spatiotemporal expression of transgenes can be controlled in conditional transgenic lines
and they also allow the maintenance of the line by either in-crossing or out-crossing.
7
Introduction
To follow tumour initiation, progression and even regression in the zebrafish,
one must be able to control the switching on or off of oncogene expression (Feitsma
and Cuppen, 2008). The tetracycline-responsive systems, which include the Tet-on
and Tet-off systems, can exert such control over transgene expression. The
tetracycline-responsive system was first established and demonstrated in Hela cells to
control transgene expression (Gossen and Bujard, 1992). In the early versions of the
system, the tetracycline repressor (tetR) from the tetracycline-resistant operon in
Escherichia coli was fused to the activating domain of the virion protein 16 from the
herpes simplex virus to form a tetracycline-controlled transactivator. tetR then
activates transcription from a minimal promoter generated by the combination of
human cytomegalovirus promoter and tet operator sequences. The isolation of a
mutant tetR, known as rtTA, that requires the presence of doxycycline (Dox) in order
to bind to and activate transcription of the minimal promoter and further refinements
to the promoter system increase its efficiency and reduce its background activities and
further gave birth to the Tet-on and Tet-off systems (Urlinger et al., 2000). This
system will allow us to control not only the expression of the gene, but also the degree
of expression by controlling Dox concentration.
1.3.2 The use of transposable elements to generate transgenic zebrafish
The standard method of generating transgenic zebrafish is via the injection of
DNA constructs containing gene expression cassettes consisting of a promoter, a
transgene and polyA signals into one-cell stage embryos in the hope that the construct
will be taken up and integrated into the genome of the injected embryos for germline
transmission. This method has been the sole method in early transgenic fish studies
(Higashijima et al., 1997; Long et al., 1997). However, the rate of transgenesis
8
Introduction
employing this method is often less than satisfactory, with germline transmission rates
of less than 10%. Hence there is a need to increase transgenesis rates to facilitate
transgenic zebrafish generation. The reported use of the Tol2 transposition system
from medaka and Sleeping Beauty transposition system from salmonid provided a
new impetus in generating transgenic zebrafish with significantly higher transgenesis
and gene expression rates (Davidson et al., 2003; Kawakami et al., 2000).
Transposable elements can be found in genomes of most vertebrates and are
composed of transposon DNA and a recombinase-coding DNA sequence that is
involved in transposon trans-activation (Ivics et al., 2004). Recently, the Activator
(Ac)/Dissociation (Ds) transposition system first isolated in maize by McClintock was
found to work in zebrafish with comparable transgenesis and gene expression rates
(Emelyanov et al., 2006; McClintock, 1950). This study also showed that the Ac/Ds
transposable element is versatile enough to be active in zebrafish and human cells,
eliminating the requirement for host cell-specific factors for transposition to occur.
Furthermore, our laboratory recently reported the successful use of the Ac/Ds
transposable element in the generation of transgenic zebrafish expressing zebrafish
kRASV12 under the control of the liver-specific fabp10 promoter in modelling
oncogene-driven liver cancer (Her et al., 2003; Nguyen et al., 2011).
1.3.3 Xmrk and cmyc driven liver cancer models
Our laboratory has also successfully made use of the Tet-on system to
generate transgenic zebrafish with inducible, liver-specific expression of oncogenes to
model liver cancer. In our zebrafish, a liver-specific fabp10 promoter was used to
drive the constitutive expression of the rtTA transactivator, which will activate
transcription of the oncogene found downstream of the minimal promoter found in a
9
Introduction
separate construct in the presence of Dox. We used two different oncogenes to model
liver cancer, namely the Xiphophorus melanoma receptor kinase gene or Xmrk, an
Epidermal Growth Factor receptor (EGFR) homologue, and mouse cmyc (Huang et al.,
2011). Xmrk was first isolated in Xiphophorus, which was the main causative agent
for melanoma development in the fish species. Xmrk was found to harbour activating
mutations in the extracellular domain, explaining its tumourigenicity in vitro and in
vivo (Winnemoeller et al., 2005). The suitability of using Xmrk to drive liver
carcinogenesis was further supported by studies implicating dysregulated EGFR
signalling in human hepatocellular carcinoma or HCC (Berasain et al., 2009). It is
noteworthy to point out that the EGFR pathway is chronically stimulated during
inflammation in the liver following injury and that tumourigenesis is favoured in the
backdrop of inflammation. Further studies implicated the over-expression of EGFR
and its ligands in human HCC (Avila et al., 2006; Berasain et al., 2007; Breuhahn et
al., 2006; Castillo et al., 2006). Thus it would be interesting to uncover the role
chronic Xmrk over-expression plays in liver carcinogenesis.
We also generated Dox-inducible liver-specific over-expression of mouse
cmyc. Initially isolated from chicken DNA, cmyc is the cellular homologue of the vmyc oncogene found in avian myelocytomatosis retrovirus MC29 (Vennstrom et al.,
1982). The oncogenic potential of cmyc was first demonstrated by its involvement in
the progression of human Burkitt’s lymphoma, due to a translocation event between
chromosome 8 and one of the three chromosomes containing genes encoding
immunoglobulins (Spencer and Groudine, 1991). cmyc encodes a transcription factor
that heterodimerises with its partner protein MAX via interaction with its carboxyterminal basic-helix-loop-helix-zipper domain. The heterodimer binds to E-boxcontaining DNA and facilitates transcription and gene expression (Blackwood and
10
Introduction
Eisenman, 1991; Eilers and Eisenman, 2008; Pelengaris et al., 2002). cmyc expression
is positively correlated with cell growth and proliferation by promoting cell-cycle
progression; it also inhibits cellular terminal differentiation of most cell types as well
as sensitises cells to apoptosis (Amati, 2001; Amati et al., 1998; Dang, 1999; Eilers,
1999). In addition, cmyc expression is also involved in cellular reprogramming to
form induced pluripotent stem cells (Eilers and Eisenman, 2008). In light of these, it
is of interest to uncover links that connect dysregulated cmyc expression and
carcinogenesis. Early studies using mouse models provided evidence linking
hepatocyte-specific cmyc over-expression with chronic hepatic proliferation and
increased incidence of cancer, and it potentiates transforming growth factor (TGF)-α
in the development of hepatocellular carcinoma (Calvisi and Thorgeirsson, 2005).
However, the role of dysregulated cmyc expression in carcinogenesis is still elusive,
although several hypotheses have been proposed (Dang et al., 2005; Eilers and
Eisenman, 2008; Pelengaris et al., 2002).
These two models that we have generated will provide us with ample material
to study the roles of oncogene-driven liver carcinogenesis, and to study the intricate
cross-talk between various signalling pathways. They also represent good models for
chemical and drug screens that target both EGFR and cmyc signalling pathways in a
whole organism.
2. An introduction to autophagy and its involvement in cancer
2.1 Autophagy as a basal and inducible cellular process
Autophagy is a ubiquitous cellular process that involves the delivery of
cellular cytoplasmic components for degradation in the lysosome. Early studies led to
the identification of at least three forms of autophagy in mammalian cells: chaperone-
11
Introduction
mediated autophagy, microautophagy and macroautophagy. They differ with respect
to the method of sequestering and delivering cytoplasmic components to the lysosome
for degradation and their physiological functions (Levine and Kroemer, 2008). In
microautophagy, cytoplasmic components are taken up by protrusion, septation or
invagination of the vacuolar membrane in the yeast or the lysosomal membrane in
eukaryotes; whereas chaperone-mediated autophagy requires chaperones to facilitate
the translocation of unfolded, soluble proteins across the lysosomal membrane (Wang
and Klionsky, 2003; Yang and Klionsky, 2009). In macroautophagy, doublemembrane vesicles called autophagosomes sequester cytoplasmic components, which
then fuse with lysosomes with the concomitant delivery of the inner single-membrane
vesicles for lysosomal degradation and recycling of resultant nutrients (Yang and
Klionsky, 2009). We will only focus on macroautophagy (referred to as autophagy
hereafter) in this dissertation.
Autophagy is an evolutionarily conserved cellular process involved in the
degradation of a cell’s cytoplasm, long-lived proteins and organelles in response to
cellular stressors such as starvation. Autophagy is also involved in the degradation of
damaged or superfluous organelles and proteins and protein aggregates, in addition to
its role in regulating cellular homeostasis at a basal level (Klionsky, 2007; Mizushima
et al., 2008). The ability of autophagy to carry out large-scale degradation of cellular
components requires it to be tightly controlled, as unregulated autophagy can have
catastrophic consequences. Due to its far-reaching effects in regulating cellular
turnover and homeostasis, autophagy deregulation is found to be implicated in various
diseases such as neurodegeneration, myopathies, infection and immunity, as well as
cancer (Levine and Kroemer, 2008; Mizushima et al., 2008).
12
Introduction
Autophagy was first identified in mammalian cells by Christine de Duve in the
1950s as part of a lysosomal degradative mechanism (Klionsky, 2007). Subsequent
genetic screens for yeast mutants affecting protein turnover led to the identification of
autophagy-related (Atg) genes and their protein products that are crucial for the
autophagy process (Yang and Klionsky, 2009). To date, more than 30 Atg genes have
been identified in yeast, and more than a dozen orthologues in higher organisms have
also been identified (Kourtis and Tavernarakis, 2009; Rubinsztein et al., 2007).
2.2 Molecular machinery of autophagy
Autophagy is a multi-step process. Autophagy induction leads to the formation
of a phagophore that undergoes nucleation and elongation to form a doublemembrane vesicle called the autophagosome which sequesters cytoplasmic
components in the process. The autophagosome subsequently fuses with endosomes
forming amphisomes, or with lysosomes forming autolysosomes where the
sequestered materials are then released into the lysosome to be degraded by lysosomal
enzymes and the resultant amino acids and other nutrients are subsequently reused for
use by the cell (Klionsky, 2007; Levine and Kroemer, 2008). This section serves to
briefly summarise the current knowledge of the molecular mechanisms of autophagy.
Induction of autophagy requires the activity of Atg1, a serine/theronine kinase,
which is found in a complex with Atg13 and Atg17 (Kabeya et al., 2005; Kamada et
al., 2000; Matsuura et al., 1997). Studies have demonstrated that the formation of this
protein kinase complex is positively correlated with an increase in autophagic activity.
Uncoordinated 51-like kinases (Ulk) 1 and 2 are mammalian homologues of Atg1,
while FIP200 has been recently identified to form a complex with Ulk1 and was
proposed to be an Atg17 homologue (Chan et al., 2007; Hara and Mizushima, 2009;
13
Introduction
Hara et al., 2008; Yan et al., 1998). Furthermore, studies by Jung et al. provided
evidence of the existence of a functional mammalian homologue of Atg13, proving
that the induction machinery is well conserved in higher vertebrates (Jung et al.,
2009).
Vesicle nucleation requires the activity of a protein complex comprising a
class III phosphatidylinositol 3-kinase (PI3K) Vps34, Vps15, Atg6 and Atg14 (Levine
and Kroemer, 2008). In yeast, Vps34 activity is regulated by its partner Vps15, and
Atg14 serves to specify the complex for autophagy (Obara et al., 2006; Stack et al.,
1995). Vps34 activity is crucial for autophagy and it is thought that the production of
PI(3)P by Vps34 serves to recruit downstream effectors to the complex for nucleation
events (Yang and Klionsky, 2009). Similar to yeast, mammalian class III PI3K,
hVps34 forms a complex with p150, a homologue of Vps15, as well as Beclin1, the
mammalian homologue of Atg6, and the complex is required for autophagy, proving
that autophagy is a highly conserved process from yeast to mammals (Liang et al.,
1999; Panaretou et al., 1997; Volinia et al., 1995).
Expansion of the double-membrane structure requires the activity of two
ubiquitin-like protein conjugation systems, namely, the Atg12 and Atg8 systems
(Yang and Klionsky, 2009).
The Atg12 system is found to be essential for
preautophagosomal membrane formation, while Atg8 conjugation is required for
formation of complete autophagosomes by mediating membrane tethering and
hemifusion (Kabeya et al., 2000; Mizushima et al., 2001; Nakatogawa et al., 2007).
Atg12 is first activated by the E1-like enzyme, Atg7, which is then transferred to the
E2-like enzyme, Atg10, before being conjugated to Atg5 (Mizushima et al., 1998a).
Atg12-Atg5 complex then interacts with Atg16 to form a Atg12-Atg5-Atg16
multimeric complex facilitated by homo-oligomerisation of Atg16 that is essential for
14
Introduction
autophagy (Kuma et al., 2002). Similarly, Atg8 is first cleaved by Atg4, exposing a
terminal glycine, it is then activated by Atg7, before being transferred to another E2like enzyme Atg3 for conjugation to a membrane lipid, phosphotidylethanolamine
(PE) (Ichimura et al., 2004; Ichimura et al., 2000). Similar to other Atg proteins,
Atg12 and Atg8 systems remain highly conserved and are functional in mammalian
autophagy and their mammalian counterparts have been identified and characterised
such as the mammalian counterpart of Atg16, Atg16L (Mizushima et al., 2003;
Mizushima et al., 1998b; Mizushima et al., 2002). Similarly, Atg8 has several
mammalian homologues, namely MAP1LC3 or LC3, GATE16, GABARAP, and
Atg8L, and they are processed similarly to the yeast Atg8 with LC3 being the most
abundant in autophagosomal membranes and therefore used as a marker to monitor
autophagic activity (Hemelaar et al., 2003; Kabeya et al., 2000; Kabeya et al., 2004;
Tanida et al., 2006). During autophagosomal membrane expansion, both Atg12-Atg5Atg16L and LC3-PE conjugates can be found to decorate the autophagosomal
membrane, with the Atg12-Atg5-Atg16L mostly localised on the outer membrane and
released from the membrane shortly before or after autophagosome formation while
LC3-PE was found on both inner and outer membranes (Kabeya et al., 2000; Kirisako
et al., 1999; Mizushima et al., 2003; Mizushima et al., 2001). LC3-PE found on the
outer leaflet of the autophagosomal membrane is released from the autophagosomal
membrane by Atg4 cleavage, while those found on the inner leaflet remains in the
autophagosome and is delivered together with the sequestered materials to the
lysosome for degradation. The reversible process of Atg8-PE conjugation, where
Atg4 can also liberate Atg8 from PE, allows Atg8 to be recycled and used in another
conjugation reaction, allowing efficient autophagy to occur (Kirisako et al., 2000). It
is also shown that the Atg12-Atg5-Atg16L complex is required for efficient LC3
15
Introduction
lipidation and for specifying the site of LC3 lipidation in mammalian autophagy
(Fujita et al., 2008).
Other Atg proteins not mentioned above play other roles that are equally
important to autophagy. An integral membrane protein Atg9 is thought to play the
role of trafficking membrane to the forming autophagosome (He et al., 2006; Noda et
al., 2000). Atg9 movement to the forming autophagosome requires Atg23 and Atg27,
which function in a heterotrimeric complex, while retrieval of Atg9 from the forming
autophagosome requires the combined action of Atg2 and Atg18 in addition to the
Atg1-Atg13-Atg17 complex (Legakis et al., 2007; Reggiori et al., 2004; Yen et al.,
2007). Atg11 and Atg17 may act as scaffold proteins in the recruitment of Atg
proteins (Yang and Klionsky, 2009). Atg15 is thought to exert lipase activity due to
the presence of a lipase active site motif (Teter et al., 2001). Yeast Atg22 was found
to aid in the efflux of amino acids resulting from autophagic activity (Yang et al.,
2006).
16
Introduction
Figure 1 Schematic illustration of the autophagy pathway and its core machinery
in mammalian cells. Autophagy is a multi-step process with the involvement of
several Atg proteins at each step. Figure reproduced from Levine and Kroemer
(Levine and Kroemer, 2008).
17
Introduction
2.3 Regulation of autophagy
The Target of Rapamycin (TOR) kinase is the key regulator of autophagy in
eukaryotes. Apart from regulating autophagy, it also regulates various cellular
processes involved in cell growth such as transcription, translation, cell size and
cytoskeletal organisation (Schmelzle and Hall, 2000). Mammalian TOR (mTOR)
signalling is crucial in the integration of various cellular signals and cell stressors
(Corradetti and Guan, 2006). mTOR can be incorporated into two TOR complexes,
mTORC1 and mTORC2, where they phosphorylate different cellular targets and
possess different functions and different sensitivities to rapamycin (Jacinto et al.,
2004; Loewith et al., 2002). mTORC1 is rapamycin-sensitive and acts as a potent
inhibitor of autophagy, where it inhibits autophagy in the presence of growth factors
and nutrients (Levine and Kroemer, 2008). Growth factors stimulate receptor tyrosine
kinases, where they activate mTORC1 via PI3K-Akt signalling which in turn, inhibit
autophagy (Esclatine et al., 2009; Levine and Kroemer, 2008; Lum et al., 2005).
mTOR senses changes in cellular energy via the 5’-AMP-activated protein kinase
(AMPK). Low energy status in the cell is represented by a high AMP to ATP ratio,
and activates AMPK where it inhibits mTOR signalling to activate autophagy
(Corradetti et al., 2004; Meijer and Codogno, 2011). mTOR inhibits autophagy by
phosphorylating Atg13 in the Atg1-Atg13-Atg17 complex; hyper-phosphorylated
Atg13 has reduced affinity for Atg1 and Atg17, leading to reduced Atg1-Atg13-Atg17
complex formation and concomitantly, reduced autophagy induction (Klionsky, 2005).
Other regulatory molecules that exert influence on autophagy also include the
eukaryotic initiation factor 2α (eIF2α) which responds to stressors like nutrient
deprivation, and endoplasmic reticulum (ER) stress, mitogen-activated (MAP) kinases,
extracellular signal-regulated kinases (ERK1/2), death-associated protein kinase
18
Introduction
(DAPk), the heterotrimeric
G protein Gi3, tumour suppressor p53 and others
(Esclatine et al., 2009).
Pharmacological agents that target mTOR as well as the phosphotidylinositol
3-kinases (PI3K) involved in autophagy regulation can be used to either induce or
inhibit the autophagic process. As mentioned above, rapamycin and its analogues
such as everolimus can be used to induce autophagy by relieving mTOR-induced
inhibition (Rubinsztein et al., 2007). 3-Methyladenine, a potent PI3K inhibitor, can
also be used to inhibit autophagy by inhibiting the activity of class III PI3K
(Blommaart et al., 1997; Petiot et al., 2000; Seglen and Gordon, 1982). Lysosomal
proton pump inhibitors such as bafilomycin A1 and lysosomotropic alkalines such as
chloroquine can also be used to inhibit autophagy effectively (Levine and Kroemer,
2008).
Apoptosis is a programmed cell death process discovered during insect
metamorphosis and during embryogenesis and development of both vertebrates and
invertebrates (Glucksmann, 1965; Lockshin and Williams, 1965a, b, c; Saunders,
1966). It is characterised by caspase activation, poly (ADP-ribose) polymerase I
(PARP1) proteolysis and DNA fragmentation cuminating in cell shrinkage, chromatin
condensation and apoptotic body formation (Giansanti et al., 2011). It operates via
two pathways, extrinsic and intrinsic, with the extrinsic pathway activated via
receptor binding of molecules belonging to the Tumour Necrosis Factor (TNF) family,
and the intrinsic pathway activated by stimuli converging and acting on the
mitochondria. Both pathways culminate in the activation of caspases, which serves to
degrade cellular components at the final step of apoptosis, following which the
degraded components are engulfed by phagocytes to prevent an inflammatory
response at the end of the process.
19
Introduction
The intimate relationship between apoptosis and autophagy was discovered
when Boya et al. reported that mammalian cells were observed to undergo apoptosis
when they were devoid of nutrients and when autophagy was inhibited through the
use of chemical inhibitors or genetic silencing by siRNAs (Boya et al., 2005). Boya et
al. further observed that cells with autophagic vesicles were able to recover when
cultured under optimal conditions following nutrient deprivation, although cells with
disrupted mitochondrial transmembrane potential were still committed to die when
cultured under the same conditions, strongly suggesting a pro-survival role of
autophagy under cellular stress. However, Notte et al. noted that in the event that the
stress is too severe or the duration of the stress is too long, autophagy may also
participate in cell death on observations that numerous autophagic vesicles were
found in dying cells (Notte et al., 2011). More studies need to be performed to
determine if autophagy does indeed participate actively in cell death either on its own,
or together with other cell death mechanisms, or if these observations merely a
consequence of a failed attempt to preserve cell viability in times of stress (Levine
and Yuan, 2005).
Further studies revealed that Bcl-2 family members were found to regulate
both pathways: anti-apoptotic Bcl-2 and Bcl-XL were found to bind to and inhibit
Beclin1 through the Beclin1 BH3 domain possibly to inhibit autophagy (Maiuri et al.,
2007; Zhou et al., 2011). The involvement of Atg5 in both autophagy and apoptosis
further supports the intricate cross-talk between the two processes: tumour cells with
atg5 over-expression were found to be more susceptible to apoptotic stimuli, while
silencing of atg5 results in partial resistance to chemotherapy (Yousefi et al., 2006).
This intricate relationship between autophagy and apoptosis is further convoluted by
findings where autophagy abrogation re-sensitised tumour cells to apoptogenic stimuli
20
Introduction
as well as enforced autophagy leading to cell demise in apoptosis-resistant cells either
through cooperation with other cell death mechanisms or massive autophagy-induced
cell death (Chen et al., 2010; Scarlatti et al., 2009; Shen and Codogno, 2011; Xie et al.,
2011; Zhivotovsky and Orrenius, 2010). Thus it would be interesting to delve more
deeply into the relationship of autophagy and apoptosis and to make use of this
newly-acquired knowledge in designing new cancer therapeutics.
21
Introduction
Figure 2 Regulation of mammalian autophagy. The intricate relationship between
various signalling pathways and autophagy regulation is depicted in the figure above.
Figure reproduced from Yang and Klionsky (Yang and Klionsky, 2010).
22
Introduction
2.4 The use of EGFP-LC3 and mRFP-EGFP-LC3 to study autophagy in vivo
The past decade has seen an exponential increase in the research on autophagy,
especially its implication in diseases hence there is a need to establish techniques to
study autophagy both in vitro and in vivo. The gold standard of analysing autophagic
activity requires the use of electron microscopy to identify autophagosomes and
autolysosomes (Mizushima et al., 2010). However such a method is both time
consuming and requires expertise in the correct identification of autophagic vesicles.
Thus there is a need to establish a more reliable and accessible method of analysing
autophagy status. The finding that LC3, the mammalian homologue of Atg8,
undergoes PE lipidation for its localisation to autophagosomes provides an
opportunity to detect autophagic activity. LC3 can be found in mammalian cells in
two forms, LC3-I and the PE-conjugated LC3-II. LC3-I is found in the cytosol while
LC3-II is usually found to be associated with autophagosomal membranes and its
formation is positively correlated with autophagosome formation and thus autophagic
activity (Kabeya et al., 2000). Thus immunoblotting of LC3-I and LC3-II provide an
easy read-out of autophagic activity in both in vitro and in vivo experimental
conditions (Klionsky et al., 2008; Mizushima et al., 2010).
The introduction of protein fusions to fluorescent proteins further allow
autophagic analyses to be easily performed using fluorescent microscopes in real time.
The use of EGFP-LC3 fusion protein was first established by Kabeya et al. where
they reported the association of LC3-II with autophagosomal membranes forming
discrete EGFP puncta. They further established that LC3-I was freely available in the
cytosol forming a diffuse cytoplasmic distribution (Kabeya et al., 2000). This study
establishes the use of EGFP-LC3 to analyse autophagic activity, where an upregulation of autophagy will see a concomitant increase in GFP puncta representing
23
Introduction
autophagosomes. The intracellular localisation of EGFP-LC3 thus becomes a
complementary method for the analyses of autophagic flux in addition to
immunoblotting for LC3 (Kabeya et al., 2000). Since then, EGFP-LC3 is used to
study autophagy dynamics in a variety of in vitro conditions using various cell lines as
well as in the generation of transgenic mice to study autophagic dynamics in a variety
of conditions such as starvation (Mizushima et al., 2004).
As autophagosome formation is an intermediate step in a highly dynamic
cellular process, EGFP-LC3 puncta observed at any specific time point usually
reflects the delicate balance between the rate of autophagosome formation and the rate
at which they are converted to autolysosomes (Mizushima et al., 2010). As such, it
would be more appropriate to measure autophagic flux to determine cellular
autophagic activity. Autophagic flux refers to the dynamic process of autophagosome
formation, delivery of engulfed cellular substrates to the lysosome and subsequent
degradation of the substrates within the lysosome. One method of measuring
autophagic flux is to quantify EGFP-LC3 puncta numbers present in cells, as
increased autophagic activity often leads to increased autophagosome formation and
therefore, EGFP-LC3 puncta formation. However, a major pitfall exists as increased
puncta numbers may also be due to reduced autophagic degradation that leads to an
accumulation of autophagosomes. Therefore the number of EGFP-LC3 puncta that
can be quantified does not correlate to cellular autophagic activity per se and several
other assays are required in addition to observing EGFP-LC3 puncta formation to
determine autophagic flux.
Another useful assay to measure autophagic flux was developed based on the
concept
of
lysosomal
quenching
of
GFP
fluorescence.
As
mentioned,
autophagosomes fuse with lysosomes as part of its maturation process to degrade
24
Introduction
engulfed cellular substrates. However, the low pH achieved through autophagosomelysosome fusions quench GFP fluorescence, making it difficult to trace EGFP-LC3
delivery to lysosomes; this also hampers efforts to demonstrate the co-localisation of
EGFP-LC3 puncta with lysosomes (Bampton et al., 2005; Kabeya et al., 2000). On
the other hand, RFP exhibits stable fluorescence in acidic compartments making it
useful in labelling acidic autophagic vesicles. A novel reporter consisting of
monomeric RFP (mRFP) and EGFP fused to LC3 was developed to study
autophagosome-lysosome fusion as well as to provide a means of measuring
autophagic flux (Kimura et al., 2007). Using this novel construct, autophagosomes are
labelled yellow (due to presence of both GFP and RFP) and autolysosomes are
labelled red and autophagic flux can be measured simply by observing the ratio of
both yellow and red signals. Increases in autophagic flux are indicated by increases in
both yellow and red puncta while blockages in fusion and maturation of
autophagosomes into autolysosomes are indicated by increases in yellow puncta
without concomitant increases in red puncta, making it easier to study autophagic flux
in real time. We can accurately study autophagic activity with the use of the mRFPEGFP-LC3 reporter together with other assays.
2.5 Autophagy in cancer
Dysregulated autophagy is implicated in various diseases such as
neurodegeneration, myopathies, infection and immunity, as well as cancer (Levine
and Kroemer, 2008; Mizushima et al., 2008). Among them, the role of autophagy in
cancer still remains elusive. The first evidence linking autophagy as a tumour
suppression mechanism came from the discovery that Atg6/Beclin1 is a
haploinsufficient tumour suppressor (Karantza-Wadsworth et al., 2007; Liang et al.,
25
Introduction
1999; Qu et al., 2003; Yue et al., 2003). Other studies implicating Atg4C, Atg5 and
Atg7 deficiencies to increased rates of tumourigenesis as well as the findings that
Beclin1 cofactors act as tumour suppressors further demonstrated the tumour
suppressive function of autophagy (Brech et al., 2009; Marino et al., 2007; Mathew et
al., 2007b; Takamura et al., 2011). Further studies revealed that most oncogenes like
class I PI3K and Akt inhibit autophagy while tumour suppressors such as phosphatase
and tensin homologue (PTEN) and tuberous sclerosis complex 1 and 2 (TSC1 and
TSC2) induce autophagy (Maiuri et al., 2009). Moreover, autophagy may also be
required to kill tumour cells efficiently in certain circumstances as shown by Giaccia
and colleagues where they demonstrated that addition of a compound STF-62247
induced autophagy and vacuolisation in von Hippel-Lindau (VHL)-deficient renal cell
carcinoma cells (Turcotte et al., 2008).
On the other hand, autophagy induction may represent a means for tumour
cells to survive various cellular insults such as hypoxic conditions, metabolic stress
and chemotherapeutic stress (Chen and Debnath, 2010; Degenhardt et al., 2006).
Although autophagy may have tumour suppressive functions, autophagy is still
required for the survival of established tumour cells in response to various stresses. As
a result of their higher proliferation rates, tumour cells have higher demands for
nutrients and oxygen and they increase their autophagic activity in order to survive
this metabolic stress (Degenhardt et al., 2006). Similarly, tumour cells found in the
interior of poorly vascularised tumours survive hypoxic conditions by up-regulating
their autophagic activity, in turn protecting them from apoptosis and necrosis.
Furthermore, autophagy may confer pro-survival benefits in metastasising tumour
cells by protecting them from anoikis (Fung et al., 2008). Autophagy also serves to
help tumour cells survive radio- or chemo-therapeutic stress. A study by Apel et al.
26
Introduction
provided evidence that siRNA-induced inhibition of autophagy re-sensitised cancer
cells to radiation-induced cell death, suggesting that cancer cells co-opt autophagy as
a cell survival mechanism, which is in line with its function of promoting cell survival
during periods of stress (Apel et al., 2008). Thus autophagy is likened to a doubleedged sword, as autophagy has both tumour suppressive and pro-tumourigenic
properties (Mathew et al., 2007a; White and DiPaola, 2009). There is a consensus that
autophagy as a tumour suppressive mechanism exerts its effects via damage
mitigation and limiting metabolic stress and genomic instability (KarantzaWadsworth et al., 2007; Mathew et al., 2007b). Tumourigenesis is favoured in an
inflammatory environment, and autophagy serves to limit inflammation and therefore
exerts its tumour suppressive function (Degenhardt et al., 2006). However, autophagy
is also co-opted as a survival mechanism by established tumour cells facing hypoxic
conditions and metabolic stress, enabling long-term cell survival against a backdrop
of apoptotic defects (Degenhardt et al., 2006; Lum et al., 2005). Autophagy may also
help tumour cells establish dormancy in the face of severe stress, and the ability to
regenerate and form viable tumour cells when favourable environmental conditions
return (White and DiPaola, 2009). Lastly, autophagy may also be up-regulated in
tumour cells facing therapeutic stress, as demonstrated by Apel et al. (Apel et al.,
2008). Thus it is hypothesised that autophagy be up-regulated during early
carcinogenesis in order for it to exert its anti-cancer function, while inhibited in extant
tumours to re-sensitise them to stress and chemo- and radio-therapy. As such, more
work needs to be done to establish this paradigm as well as to determine the efficacy
of such an approach.
27
Introduction
Figure 3 Autophagy in tumourigenesis and anti-cancer therapy. Autophagy is
likened to a double-edged sword possessing both tumour suppressive and protumourigenic properties. Autophagy can either promote tumour cell survival or kills
tumour cells as a result of anti-cancer therapy; hence it is a novel target in anti-cancer
therapy. Figure reproduced from Maiuri et al. (Maiuri et al., 2009).
28
Introduction
3. Rationale of current study
3.1 Establishing Tg(fabp10:egfp-lc3) and Tg(fabp10:mrfp-egfp-lc3)
transgenic
zebrafish for studying autophagy
The study of autophagy in a model organism has been made possible by the
use of EGFP-LC3 fusion protein and this was demonstrated by Mizushima et al. when
they generated the first transgenic mouse with ubiquitous and constitutive expression
of EGFP-LC3 (Mizushima et al., 2004). Therefore, it is envisioned that the
establishment of a transgenic zebrafish expressing egfp-lc3 will be useful in the study
of autophagy in this popular vertebrate model, where little study has been conducted.
A zebrafish transgenic line with liver-specific egfp-lc3 expression will be generated in
this study in order to study autophagy in the liver. This makes sense as autophagy was
first studied in rat livers following glucagon injection and that autophagy plays an
important role in nutrient and energy metabolism and homeostasis in the liver (Yin et
al., 2008). In addition, reduced autophagic activity was implicated in liver
inflammation and carcinogenesis involving alpha-1-antitrypsin deficiency, suggesting
that functional autophagy is required for proper protein aggregate disposal (Perlmutter,
2009).
To generate a transgenic line with constitutively liver-specific egfp-lc3
expression, the liver-specific fabp10 promoter will be used to restrict egfp-lc3
expression to the liver tissue. In order to increase transgenesis and germline
transmission rates, the Ac/Ds transposition system will be used as well. This
transgenic line will be useful in studying autophagy in liver development as well as in
various liver injuries. It will be useful for studying the role of autophagy in liver
cancer using our liver cancer models, and will also be useful for chemical and drug
screens that target the autophagy pathway.
29
Introduction
As a proof of concept, we will also generate a transgenic line with
constitutively liver-specific mrfp-egfp-lc3 expression. This will be the first attempt in
generating a transgenic organism expressing this novel reporter. We hope to employ
this transgenic line in liver cancer study as well as other studies involving liver
development or liver diseases in the zebrafish. Similar methodology used in the
generation of the EGFP-LC3 transgenic line will be applied here.
3.2 Establishing the role of autophagy in liver cancer
Studies have also demonstrated both pro-tumourigenic and anti-tumourigenic
functions of autophagy in liver cancer. Mice heterozygous for Beclin1 have a higher
propensity to develop spontaneous hepatocellular carcinoma (HCC), suggesting an
anti-tumourigenic function (Qu et al., 2003). Ding et al. demonstrated in their study
that HCC cell lines have lowered levels of Atg gene expression as well as autophagic
activity compared to normal hepatic cell lines; they also found reduced levels of
Beclin1 mRNA and protein levels in HCC tissue samples compared to adjacent nontumourigenic tissues from the same patient. They also demonstrated that lowered
autohagic activity in the malignant cell lines and HCC tissues was especially
prominent when anti-apoptotic proteins such as Bcl-XL was over-expressed, further
suggesting that autophagy defects synergised with apoptotic defects in promoting
liver carcinogenesis, suggesting that antophagy has an anti-tumourigenic function
(Ding et al., 2008). In contrast, Longo et al. demonstrated that autophagy functions as
a survival mechanism in liver cancer cells and protected them from anthocyanininduced apoptosis, thus suggesting a pro-tumorigenic role of autophagy (Longo et al.,
2008). As can be seen, the role of autophagy in liver cancer is currently not well
established, although it is proposed that autophagy mainly exerts a tumour
30
Introduction
suppressive function during early stages of carcinogenesis while it is co-opted by
extant tumour cells as a means to survive metabolic and chemo- or radio-therapeutic
stress. Thus we would like to examine this phenomenon using our zebrafish liver
cancer models. In order to do this, we will examine the autophagic flux during liver
tumour initiation, progression and regression using our zebrafish liver tumour models.
We hope to derive more detailed information on the involvement of autophagy in
liver cancer, and hope that this information will help in designing combinatorial
chemotherapy for liver cancer patients using autophagy inducers or inhibitors at the
appropriate stages of treatment together with established chemotherapeutic drugs for
liver cancer.
3.3 Objectives of the study
We would like to propose the following objectives for this study:
1. Validation of conservation of autophagic machinery in the zebafish
2. Establishment and characterisation of a zebrafish transgenic line with
constitutively liver-specific egfp-lc3
3. Establishment and characterisation of a zebrafish transgenic line with
constitutively liver-specific mrfp-egfp-lc3 expression
4. Analysing autophagic flux in zebrafish transgenic lines with inducible liver
tumour zebrafish models by crossing egfp-lc3 and oncogene-expressing
transgenic lines
We hope that by achieving these objectives we will be able to prove our
hypothesis that autophagy serves an anti-cancer function in the early stages of cancer
and it may be co-opted by cancer cells as a cell survival mechanism at later stages.
This will greatly impact the field and provide information for combinatorial drug
31
Introduction
therapy using existing drugs and autophagy inducers or inhibitors at appropriate
stages of cancer to treat liver cancer patients.
32
Materials and Methods
Materials and Methods
33
Materials and Methods
1. Zebrafish care and maintenance and tissue collection
1.1 Zebrafish maintenance
Wild type and transgenic adult zebrafish were maintained in the National
University of Singapore Aquarium at Block S2 Level 2 according to the method as
described in the Zebrafish Book available online at the Zebrafish Information
Network (ZFIN) (http://zfin.org/zf_info/zfbook/zfbk.html) (Westerfield, 2000) and in
compliance with the Institutional Animal Care and Use Committee (IACUC)
guidelines. Fish were kept in the aquarium with a photoperiod of 14 h light, 10 h dark.
Fish were fed brine shrimp (World Aquafeeds, USA) twice a day, with additional
flakes being fed in the late afternoon during the spawning period.
1.2 Zebrafish spawning and embryo collection
To collect embryos, breeding adult fish were separated into pairs with each
pair being separated using a divider in a tank the night before. Spawning was induced
by the removal of the divider and allowing the fish to mate on the following morning.
Following that, the breeding pair was removed and the embryos that were found on
the bottom of the tank were drained using a sieve and placed in a petri dish containing
egg water (final concentration of 60 µg/ml sea salt dissolved in RO water) and
methylene blue (final concentration of 0.0002 %), and staged accordingly (Kimmel et
al., 1995). The required developmental stages were presented as hours postfertilisation (dpf). Embryos were placed in an incubator with a constant temperature
of 28.5 °C to facilitate staging.
To rear embryos, embryos were kept in the incubator with daily changes of
egg water and removal of dead embryos and larvae until they were approximately 4
dpf, before they were transferred into larger tanks containing egg water only. The
34
Materials and Methods
larvae were transferred into tanks in the aquatic system when they were
approximately 21 dpf.
1.3 Microinjection into one-cell stage embryos
Needles were prepared from glass capillaries using the P-97 Flaming/Brown
Micropipette puller (Sutter Instrument, USA) using optimised heat and pull time
conditions. Microinjection was carried out using the FemtoJet microinjector
(Eppendorf, Germany) using optimised pressure and microinjection time settings.
Embryos were placed on a glass slide with excess egg water drained off using
KimWipes tissues (Kimberly-Clark Co., USA) before microinjecting under a
dissecting microscope. Microinjected embryos were then returned to the petri dishes
to be reared.
To generate the Tg(fabp10:egfp-lc3) transgenic line, 50 ng/µl of the plasmid
pDS-FABP10-EGFP-LC3 was mixed with 500 ng/µl of Ac mRNA, and the mixture
was microinjected into one-cell stage embryos. A total volume of 1 nl was injected
into each embryo. A total of 25 pg of plasmid and 250 pg of Ac mRNA were microinjected into each embryo.
To generate the Tg(fabp10:mrfp-egfp-lc3) transgenic line, 50 ng/µl of the
plasmid pDS-FABP10-mRFP-EGFP-LC3 was mixed with 50 ng/µl Ac mRNA, and
the mixture was micro-injected into one-cell stage embryos. A total volume of 1 nl
was injected into each embryo. A total of 25 pg of plasmid and 25 pg of Ac mRNA
were micro-injected into each embryo.
35
Materials and Methods
1.4 Larval screening and photo-documentation
Transgenic larvae were screened based on fluorescence expression, embryos
were reared until approximately 3 dpf before being screened using a fluorescence
microscope (Carl Zeiss AG, Germany). Larvae expressing desired fluorescence were
siphoned with a plastic pipette and reared in a separate petri dish containing egg water.
Larvae were photo-documented using the Carl Zeiss Axiovert 200M
microscope (Carl Zeiss, Germany), using the 2.5X, 5X and 10X objectives after
anaesthetised using 0.1 % phenoxy-ethanol (Sigma Aldrich, USA). GFP signals were
detected using the blue filter while RFP signals were detected using the green filter.
Images were taken using the attached Axiocam HRC camera mounted on the
microscope and accompanying software. Images were further processed using Adobe
Photoshop software (Adobe, USA).
1.5 Liver tissue collection from adult zebrafish
Adult transgenic fish were sacrificed to collect their liver tissue for histology
and protein analyses. Briefly, each fish was first anaesthetised in ice water, after
which an incision was made from the anus across the belly along the midline, up
across the pectoral fins and finally along the trunk ending at the anus, with the skin
removed to expose the liver tissue. Liver tissue was then removed from the trunk
using a pair of sterile forceps and placed in sterilised microcentrifuge tubes.
Tumourigenic livers from 3 transgenic fish were pooled together in a single tube,
while up to 10 non-tumourigenic livers were pooled together in 1 tube. All tubes were
snapped frozen using liquid nitrogen and subsequently stored at -80 °C.
36
Materials and Methods
2. RNA applications
2.1 Total RNA isolation from embryos and larvae
Embryos and larvae at different stages of development were collected in
sterilised microcentrifuge tubes and snap frozen using liquid nitrogen after draining
off excess egg water. Total RNA was then extracted using TRIZOL reagent
(Invitrogen, USA) according to manufacturer’s instructions with some modifications.
All centrifugation steps were performed at 4 °C. 300 µl of TRIZOL reagent was first
added to each sample and homogenised using a motorised pestle before adding the
remaining 700 µl of the reagent. Each sample was then incubated at room temperature
for 10 minutes to allow nucleoproteins to dissociate; this was followed by the addition
of 200 µl of chloroform. The mixture was shaken vigorously by hand for 20 seconds
before incubating at room temperature for an additional 5 minutes. Samples were
centrifuged at 12,000 g for 15 minutes, after which the aqueous layer was carefully
transferred to a separate sterilised tube. 500 µl isopropanol was added to each tube to
precipitate RNA following which the tubes were inverted 10 times by hand and
incubated at room temperature for 20 minutes. The mixtures were vortexed for 5
seconds before undergoing centrifugation at 12,000 g for 10 minutes. Excess
isopropanol was removed and 1 ml of 75 % cold ethanol was added to wash the
resulting pellet. The pellets were centrifuged again at 7,500 g for 5 minutes before
draining the ethanol. The pellets were left to air-dry for approximately 5 minutes
before the addition of diethyl pyrocarbonate (DEPC)-treated water to fully dissolve
the pellet. Aqueous RNA samples were heated at 55 °C for 10 minutes.
37
Materials and Methods
2.2 RNA quantification
The
aqueous
RNA
was
quantified
using
Nanodrop
ND-1000
spectrophotometer (Thermo Fisher Scientific, USA). The RNA quality was verified
by running the samples in 1 % agarose gel between 80 to 100 V for approximately 20
minutes, upon which the gel was photo-documented using the Gel Doc XR Gel
Documentation System (Bio-Rad, USA). RNA samples were then aliquot into
separate tubes and stored at -80 °C.
2.3 Ac mRNA preparation
The pAc-SP6 plasmid was first linearised using BamHI, and gel purified to
yield linearised DNA. The linearised DNA was used to generate capped mRNA in
vitro using the mMessage mMachine SP6 kit (Ambion, USA). Briefly, approximately
2 µg of linearised template was mixed with 2 µl of 10X Reaction Buffer (containing
unspecified concentrations of salts, buffer, DTT and other ingredients), 10 µl of 2X
NTP/CAP (10 mM ATP; 10 mM CTP; 10 mM UTP; 2 mM GTP; 8 mM cap
analogue), 2 µl of SP6 enzyme mix (buffered 50 % glycerol containing RNA
polymerase, RNase inhibitor, and other components) and nuclease-free water to give a
20 µl reaction volume. The mixture was thoroughly mixed and incubated at 30 °C for
2 hours, following which 1 µl of TURBO DNase (2 units/µl) was added to the
mixture and incubated at 37 °C for an additional 15 minutes. The resultant capped
RNA was precipitated by lithium chloride. Briefly, 1 µl 0.5 M EDTA pH 8, 2.5 µl 4
M lithium chloride and 75 µl 100 % cold ethanol was added to the mixture and left to
precipitate on ice for approximately 30 minutes, following which the mixture was
centrifuged at 4 °C at 14,000 rpm for 20 minutes. The supernatant was carefully
removed and the pellet was washed using 500 µl of 75 % cold ethanol and vortexed.
38
Materials and Methods
The resultant mixture was then centrifuged at 4 °C at 14,000 rpm for 10 minutes. The
ethanol was decanted, and the pellet was air-dried for approximately 5 minutes before
being dissolved in DEPC-treated water. The aqueous capped mRNA was then
quantified using the Nanodrop ND-1000 spectrophotometer (Thermo Fisher Scientific,
USA), and the quality was verified by running a 1 % agarose gel between 80 to 100 V
for approximately 20 minutes. The gel was later photo-documented using the Gel Doc
XR Gel Documentation System (Bio-Rad, USA) to ensure only a single band of size
between 1 kb and 1.5 kb was seen. mRNA was then aliquot into separate tubes and
stored at -80 °C.
3. DNA applications
3.1 Total DNA isolation
Larvae at different stages of development were collected and DNA extraction
buffer (10 mM Tris-HCl pH8.2; 10 mM EDTA; 200 mM NaCl; 0.5 % SDS),
proteinase K and RNase were added according to established protocol. Larvae were
homogenized and incubated at 56 °C for approximately 2 hours, with vortexing every
30 minutes during incubation. An equal volume of phenol-chloroform was added and
mixed and centrifuged at 13,000 rpm for 10 minutes. The supernatant was then
transferred to a separate sterilized microcentrifuge tube and 0.1 volume of 3 M
sodium acetate (pH 5.2) and 2 volumes of 100 % ethanol were added, mixed and
incubated at room temperature for 15 minutes, following which the mixture was
centrifuged at 13,000 rpm for 5 minutes to remove ethanol. The resulting pellet was
rinsed with 200 µl of 70 % ethanol and centrifuged at 13,000 rpm for 5 minutes,
following which the excess ethanol was drained off. The pellet was air-dried for
39
Materials and Methods
approximately 5 minutes and TE buffer was added to fully dissolve the pellet. The
aqueous DNA samples were aliquot into separate tubes and stored at -20 °C.
For genotyping Polymerase Chain Reaction (PCR), individual embryos or
larvae were placed in sterilised microcentrifuge tubes, and covered with 20 µl of
extraction buffer consisting of 1X PCR Buffer from the GoTaq Flexi DNA
Polymerase kit (Promega, USA) and proteinase K (1:500 ratio). The mixture was
subsequently incubated at 60 °C for at least 2 hours, and vortexed after 1 hour of
incubation. The mixture was quickly centrifuged to pellet the mixture and heated at
95 °C for 10 minutes to eliminate proteinase K activity. The mixture was
subsequently stored at -20 °C. 1 µl of the mixture was used to run 1 reaction of
genotyping PCR.
3.2 cDNA synthesis
cDNA synthesis was carried out using isolated total RNA using the
SuperScript II Reverse Transcriptase kit (Invitrogen, USA). Briefly, total RNA was
pre-treated with DNase to remove contaminating DNA. 20 µl of aqueous total RNA
containing 2 µg of RNA, and 2 µl of 0.5 µg/µl oligo-dT and 10 mM dNTPs were
mixed in a sterilised microcentrifuge tube and incubated at 65 °C for 5 minutes. 8 µl
of 5X First Strand Buffer (250 mM Tris-HCl, pH 8.3; 375 mM KCl; 15 mM MgCl2)
were added together with 4 µl of 0.1 M DTT and 2 µl of RNaseOUT (40 units/µl) into
the mixture and incubated at 42 °C for 2 minutes, following which 2 µl of SuperScript
II Reverse Transcriptase was added. The entire reaction mixture was incubated at
42 °C for 50 minutes, and then at 70 °C for 15 minutes to inactivate reverse
transcriptase activity. The resultant cDNA was diluted 10 times using autoclaved
40
Materials and Methods
MilliQ water and quantified using the Nanodrop ND-1000 spectrophotometer
(Thermo Fisher Scientific, USA).
3.3 Construction of plasmid DNA
3.3.1 Restriction endonuclease digestion of DNA
Restriction endonuclease digestion of plasmid DNA was performed to release
desired fragments from a particular plasmid for sub-cloning purposes, or to linearise a
plasmid in order to insert a DNA fragment into the said plasmid. All restriction
enzymes used were purchased from New England Biolabs, and digestion reactions
were performed at 37 °C for 2 hours or according to manufacturer’s recommendations.
Typically 10 µg of plasmid were used in restriction digestion reactions. All digestion
reactions were performed in a total volume of either 60 µl or 100 µl, with the volume
of restriction enzymes not exceeding 10 % of total reaction volume to prevent star
activity. To stop the reaction, the mixture was immediately placed on ice, following
which they were mixed with 6X Loading Dye (Fermentas, USA) and run on a 1 %
agarose gel to separate the fragments. DNA electrophoresis was run at a constant
voltage of between 80 to 100 V for 20 to 45 minutes, depending on the estimated size
of fragment and linearised plasmid. Table 1 shows the restriction enzymes used for
subcloning in this study:
41
Materials and Methods
Table 1. Restriction enzymes used for sub-cloning
Plasmid used
Released fragment
pLF-3.0 EGFP
EGFP
pEGFP-LC3
EGFP-LC3
pmRFP-EGFP-LC3
mRFP-EGFP-LC3
pMDS6
Nil, linearise the plasmid
for sub-cloning
Restriction enzymes used
MluI, NheI
MluI, NheI
MluI, NheI
MluI, NotI
42
Materials and Methods
3.3.2 Recovery of DNA fragments from agarose gel
Gel purification is one of the most effective methods to recover and purify
DNA fragments that were released or digested by restriction enzymes. Gel
purification reactions in this study were performed using the QIAquick Gel Extraction
Kit (Qiagen, USA) according to manufacturer’s instructions. Briefly, gel slices
containing the DNA band of interest were cut from the agarose gel, weighed, and 3
volumes of Buffer QG (containing guanidinium thiocyanate) were added to 1 volume
of gel. The resultant mixture was incubated at 50 °C for 10 minutes with occasional
vortexing to completely melt the gel. This was following by the addition of 10 µl of 3
M sodium acetate, pH 5.0. For fragments larger than 4 kb, 1 gel volume of 100 %
isopropanol was added to the mixture. The entire mixture was then loaded into the
QIAquick column and centrifuged at 14,000 rpm for 1 minute. To completely remove
traces of agarose gel, an additional 500 µl of Buffer QG was added and centrifuged
using the same conditions. The column was washed using 750 µl of Buffer PE and
centrifuged. The column was centrifuged for an additional minute after decanting to
remove residual Buffer PE. 30 µl of autoclaved MilliQ water was added to the column
and the column was left to incubate at room temperature for approximately 2 minutes
before centrifuging at 14,000 rpm for 1 minute to elute the DNA in the column. The
DNA was quantified and stored at -20 °C.
3.3.3 Ligation
Ligation reactions between DNA fragments and vector DNA were typically
carried out in a 10 µl volume reaction consisting of 1 µl of 10X ligation buffer (0.3 M
Tris-HCl, pH 7.8; 0.1 M MgCl2; 0.1 M DDT and 5 mM ATP) (NEB, USA), 1 µl T4
DNA ligase (NEB, USA) and variable volumes of vector DNA, insert DNA and
43
Materials and Methods
autoclaved MilliQ water. Vector DNA and insert DNA were added according to a
molar ratio of approximately 1:3 or 1:4. The ligation reaction was mixed well and left
to incubate at 4 °C overnight. Occasionally, 1:10 molar ratios were attempted if
reactions using molar ratios of 1:3 or 1:4 did not yield successful ligation.
3.3.4 Competent cell preparation
Competent bacterial cells are usually used for transformation and retransformation reactions. To prepare competent cells, One Shot MAX Efficiency
DH5α-T1 competent cells (Invitrogen, USA) or any other types of bacterial cells were
first streaked onto blank Luria Bertani (LB) (Invitrogen, USA) agar plates and
incubated at 37 °C overnight. One colony was picked from the plate and inoculated
into 25 ml of LB media and incubated at 37 °C with constant agitation at 250 rpm. 2.5
ml of the culture was re-innoculated into another 25 ml of LB media and incubated at
37 °C overnight with constant agitation. The next morning, 0.5 ml of the overnight
culture was inoculated into 50 ml of LB media and incubated at 37 °C with constant
agitation for 2 to 3 hours, until the OD600 reached between 0.5 and 0.6. The culture
was then chilled on ice for 15 minutes after transferring the culture into 50 ml Falcon
tubes. The competent cells were pelleted by centrifuging at 2,500 rpm at 4 °C for 15
minutes, following which the supernatant was decanted and the tubes were inverted
and air-dried to completely remove the supernatant. Cells were suspended in 17 ml of
CCMB solution (80 mM CaCl2.2H2O; 20 mM MnCl2.4H2O; 10 mM MgCl2.6H20, 10
mM CH3COOK, 100 ml glycerol), following which the solution was chilled on ice for
20 minutes. The solution was further centrifuged at 3,000 rpm at 4 °C for 15 minutes,
following which the supernatant was decanted. The cells were subsequently resuspended in 4 ml of CCMB, and incubated on ice for 30 minutes, after which they
44
Materials and Methods
were aliquot into sterilised pre-chilled microcentrifuge tubes in aliquots of 100 µl.
The tubes were snap frozen using liquid nitrogen and stored at -80 °C.
3.3.5 Transformation and re-transformation reactions
Transformation and re-transformation reactions can be prepared using the
freshly made competent cells. For a transformation reaction, a tube of competent cells
was first thawed on ice before adding 5 µl of ligation product. The mixture was then
incubated on ice for 20 minutes before being subjected to 2 minutes of heat shock at
37 °C. The mixture was then incubated on ice for an additional 5 minutes, after which
1 ml of LB broth was added to the mixture. The entire mixture was then incubated at
37 °C for 1 hour with constant agitation at 250 rpm. The mixture was centrifuged at
4,000 rpm for 4 minutes following incubation, and excess LB solution was decanted,
leaving approximately 100 µl of LB solution in the tube. The competent cells were
suspended using the 100 µl of LB solution, following which the competent cell
mixture was spread on a LB agar plate containing appropriate antibiotics (30 µg/ml
Kanamycin or 100 µg/ml Ampicillin) using glass beads. The LB agar plate was
incubated at 37 °C overnight.
Re-transformation reactions were performed in order to amplify a given
plasmid for other experiments. For a re-transformation reaction, a tube of competent
cells was first thawed on ice before adding 0.1 µl of plasmid DNA. The mixture was
then incubated on ice for 5 minutes, subjected to 2 minutes of heat shock at 37 °C,
and then further incubated on ice for an additional 5 minutes. Subsequently, the
mixture was spread on a LB agar plate containing appropriate antibiotics (30 µg/ml
Kanamycin or 100 µg/ml Ampicillin) using glass beads. The LB agar plate was
incubated at 37 °C overnight.
45
Materials and Methods
3.3.6 Colony screening
To verify successful ligation and transformation reactions, colony screening
PCR was performed. Colonies were first marked in numerical order and picked using
white sterile pipette tips and inoculated into PCR tubes containing 10 µl of antibioticcontaining LB broth. The PCR tubes were further incubated at 37 °C for 30 minutes
prior to colony screening PCR using various primers, of which one primer was
designed to anneal to the vector DNA while its complementary primer was designed
to anneal to the insert DNA. Such a design allows us to determine the presence of
insert as well as the orientation of insertion from a single PCR reaction. PCR
reactions were performed according to section 3.4.1.
3.3.7 Bacterial cell culture and plasmid amplification
After selecting for colonies containing the desired inserts in the correct
orientations, 1 µl of the culture from the PCR tubes were inoculated into tubes not
containing more than 5 ml of antibiotic-containing LB broth. The cultures were
incubated at 37 °C overnight with constant agitation at 250 rpm.
3.3.8 Plasmid isolation and purification
Following an overnight incubation, the plasmid was isolated and purified
using the Wizard Plus SV Miniprep DNA Purification System (Promega, USA).
Overnight culture was centrifuged at 4,000 rpm at 4 °C for 5 minutes, and the
supernatant decanted. The pellet was re-suspended in 250 µl of Cell Re-suspension
solution (100 mg/ml RNase A; 10 mM EDTA; 25 mM Tris-HCl, pH 7.5), following
which 250 µl of Cell Lysis solution (0.02 M NaOH; 1 % SDS) was added to the
bacterial suspension. The tubes were inverted 6 times by hand to mix the bacterial
46
Materials and Methods
suspension and incubated at room temperature for 2 minutes. 10 µl of Alkaline
Protease solution was subsequently added and the suspension was mixed by inverting
the tube 5 times by hand, following which the tube was further incubated at room
temperature for 5 minutes. 350 µl of Neutralisation solution (4.09 M guanidine
hydrochloride; 0.759 M CH3COOK; 2.12 M glacial CH3COOH, pH 4.2) was then
added and the tubes were inverted 5 times by hand. The mixture was centrifuged at
14,000 rpm for 10 minutes to pellet the bacterial cell debris, and the supernatant was
transferred to a fresh spin column provided by the kit. The spin column containing the
supernatant was further centrifuged at 14,000 rpm for 1 minute, following which the
flow-through was discarded and 750 µl of Column Wash Buffer (162.8 mM
CH3COOK; 22.6 mM Tris-HCl; 0.109 mM EDTA, pH 8.0; 75 % ethanol) was added
to wash the plasmid and the spin column was further centrifuged at 14,000 rpm for 1
minute. After discarding the flow-through, an additional 250 µl of Column Wash
Buffer was added and the spin column was centrifuged for a second wash step. The
spin column was then transferred to a new sterile microcentrifuge tube to elute the
plasmid DNA. 50 µl of autoclaved MilliQ water was added to the spin column and the
spin column was incubated at room temperature for 5 minutes before being
centrifuged at 14,000 rpm for 1 minute to elute the plasmid DNA. The plasmid DNA
was then quantified and 1 µl of plasmid DNA was subjected to gel electrophoresis
using a 1 % agarose gel to verify and compare migration speed with its predecessors.
3.4 Polymerase chain reaction
Polymerase chain reactions are useful to verify as well as amplify DNA
fragments in a single reaction. PCR reactions have many applications and variations
47
Materials and Methods
in a molecular biology laboratory and we highlight some of the variations that were
utilised in this study in this section.
3.4.1 Standard PCR
Standard PCR reactions were performed using GoTaq Flexi DNA Polymerase
kit (Promega, USA) with a total reaction volume of 10 µl consisting of 2 µl of 5X
Green GoTaq Flexi Buffer, 1.2 µl of 25 mM Magnesium Chloride solution, 1 µl of
dNTP mixture (2 mM ATP; 2 mM GTP; 2 mM CTP; 2 mM TTP), 0.5 µl of 10 µM
sense primer, 0.5 µl of 10 µM anti-sense primer, 0.2 µl of Taq polymerase (5 units/µl)
and 0.5 µl of template DNA, with the addition of autoclaved MilliQ water adjusted
accordingly to obtain a total reaction volume of 10 µl. For colony screening and
genotyping PCR, 1 µl of bacterial culture was usually added.
The PCR programme was typically set up with the following cycling
parameters: an initial denaturation step at 95 °C for 5 minutes, followed by varying
number of cycles comprising a three-step cycling of denaturation at 94 °C for 30
seconds, annealing at 60 °C for 30 seconds and extension at 72 °C for 1 minute. The
entire PCR programme ends typically with a final extension at 72 °C for 10 minutes.
The annealing temperature as well as the extension times can both be adjusted for
optimised PCR reactions depending on the primers used as well as the lengths of
amplicon. Typically, the annealing temperature will be set to 2 °C lower than the
lower temperature of a pair of primers, while the extension time is estimated based on
the formula of 1 min/kb of amplicon. Table 2 shows the primers used for genotyping
and other variants of standard PCR.
48
Materials and Methods
Table 2. Primers used in PCR
Gene name
Primer
Direction
name
3'LFABPF
Fragment
Forward
corresponding
to egfp
5'LC3R
Fragment
Reverse
corresponding
to egfp
Fragment
3’LFABPF Forward
corresponding
to mrfp-egfplc3
Fragment
LC3R
Reverse
corresponding
to mrfp-egfplc3
β-actin
B-actin2F
Forward
β-actin
B-actin2R
Reverse
Sequence
5'GTTCAAACAGCAGCAGGTCATTG
3'
5'GGTCTTCTCGGACGGTCTAGATCT
3'
5'GTTCAAACAGCAGCAGGTCATTG
3'
5'AAGAGCAAGGATGTTCGGGC3'
5'GAGAGAGGCTACAGCTTCAC3'
5'ACTCCTGCTTGCTAATCCAC3'
49
Materials and Methods
3.4.3 2-step reverse transcription polymerase chain reaction (2-step RT-PCR)
The 2-step RT-PCR serves to amplify DNA fragments using mRNA templates.
It usually starts with cDNA synthesis, as described in section 3.2, before the cDNA
templates are used in a PCR reaction as described in section 3.4.1. Table 3 shows the
primers used in 2-step RT-PCR.
50
Materials and Methods
Table 3. Primers used in 2-step RT-PCR
Gene
Primer
Direction
Sequence
name
name
lc3
LC3F
Forward
5'TTGTAAAGACGGACTTCACAGGACG3'
lc3
LC3R
Reverse
5'AAGAGCAAGGATGTTCGGGC3'
5'GGTGTTGTTACCAGCTCCAAA3'
atg5
Atg5F
Forward
5'TGACCATCACTCCCAATAAGG3'
atg5
Atg5R
Reverse
β-actin
B-actin2F
Forward
5'GAGAGAGGCTACAGCTTCAC3'
β-actin
B-actin2R
Reverse
5'ACTCCTGCTTGCTAATCCAC3'
51
Materials and Methods
3.5 DNA Vectors
3.5.1 pLF3.0-EGFP
This plasmid is a kind gift from Her et al. (Her et al., 2003). The plasmid
consists of the liver-type fatty acid binding protein (FABP10) promoter attached to
egfp. This construct allows egfp expression in the liver and the promoter was used to
control egfp-lc3 expression in the liver.
Figure 4 Construct map of pLF3.0-EGFP.
52
Materials and Methods
3.5.2 pEGFP-LC3
This plasmid is a kind gift from Kabeya et al. (Kabeya et al., 2000). The
plasmid contains a fusion protein between rat lc3 and egfp. The egfp-lc3 fragment was
used to generate the Tg(fabp10:egfp-1c3) transgenic line.
CMV promoter
EGFP
pEGFP-LC3
5452 bp
Kan / Neo
rLC3
poly A signals
Figure 5 Construct map of pEGFP-LC3.
53
Materials and Methods
3.5.3 pmRFP-EGFP-LC3
This plasmid is a kind gift from Kimura et al. (Kimura et al., 2007). The
plasmid contains a fusion protein between rat lc3, egfp and mrfp. The mrfp-egfp-lc3
fragment was used to generate the Tg(fabp10:mrfp-egfp-1c3) transgenic line.
Figure 6 Construct map of pmRFP-EGFP-LC3.
54
Materials and Methods
3.5.4 pMDS6
This plasmid is a kind gift from Emelyanov et al. (Emelyanov et al., 2006).
This plasmid consists of a multiple cloning site (MCS) flanked by a pair of Ds
elements to facilitate transposition in the presence of Ac mRNA. The fabp10:egfp-lc3
expression cassette was sub-cloned into the MCS and the whole construct and Ac
mRNA were co-injected into one-cell stage embryos to generate Tg(fabp10:egfp-lc3).
Similarly, the fabp10:mrfp-egfp-lc3 expression cassette was subcloned into the MCS
and the whole construct and Ac mRNA were co-injected into one-cell stage embryos
to generate Tg(fabp10:mrfp-egfp-lc3).
Amp
5'DS
pMDS6
3666 bp
MCS
3'DS
Figure 7 Construct map of pMDS6.
55
Materials and Methods
3.5.5 pAc-SP6
This plasmid was also a kind gift from Emelyanov et al. (Emelyanov et al.,
2006). This plasmid was used to generate Ac mRNA via in vitro transcription as
described in section 2.3.
Figure 8 Construct map of pAc-SP6.
56
Materials and Methods
4. Protein applications
4.1 Total protein isolation and extraction
To extract protein from larvae, larvae must first be de-chorionated using a pair
of forceps followed by gentle washing by swirling larvae in a petri dish containing
egg water to remove chorion membranes. Next, 50 larvae were placed in a sterilised
microcentrifuge tube and 100 µl of Tissue Protein Extraction Reagent (T-PER)
(proprietary detergent in 25 mM bicine; 150 mM sodium chloride, pH 7.6) (Thermo
Scientific, USA) and 1 µl of protease inhibitor (Roche Applied Science, Germany)
were added. The whole mixture was homogenised completely, following which the
mixture was centrifuged at 14,000 rpm at 4 °C for 10 minutes. The supernatant was
then transferred to a new tube and aliquots were used for protein quantification.
4.2 Protein quantification
The Protein Assay Dye Reagent Concentrate (Bio-rad, USA) was used to
quantify proteins using the microassay procedure using a microtiter plate, according
to manufacturer’s instructions. Briefly, five dilutions of a protein standard, usually
Bovine Serum Albumin (BSA) were prepared in autoclaved MilliQ water and 160 µl
of each standard was pipeted into separate microtiter plate wells. For protein samples,
2 µl of each sample was pipeted into each well containing 158 µl of autoclaved
MilliQ water, making up a total of 160 µl. This is followed by pipeting 40 µl of Dye
Concentrate to each well, where they were mixed by gentle pipeting to avoid bubble
formation. The entire microtiter plate was then incubated at room temperature for 5
minutes before the absorbance at 595 nm was measured using the xMark Microplate
Spectrophotometer (Bio-rad, USA). Protein samples were stored at -80 °C.
57
Materials and Methods
Following protein quantification, protein samples were aliquot into separate
microcentrifuge tubes, and 6X loading dye (300 mM Tris-HCl, pH 6.8; 12 % (w/v)
SDS; 0.6 % (w/v) bromophenol blue; 60 % (v/v) glycerol; 3 % beta-mecaptoethanol)
was added to each sample. The samples were then mixed by gentle pipeting before
being subjected to denaturation at 100 °C for 5 minutes. Denatured samples were
stored at -20 °C if not used to run SDS-Polyacrylamide Gel Electrophoresis (SDSPAGE) immediately after denaturation.
4.3 Protein Detection
4.3.1 SDS-PAGE gel casting
The SDS-PAGE gel must first be cast prior to running SDS-PAGE. A typical
SDS-PAGE gel consists of a dis-continuous gel made up of a stacking gel used to
compress protein samples into a thin line across the gel, and a resolving gel used to
separate proteins according to molecular sizes. Usually 12 % resolving gels are cast
and used in a typical SDS-PAGE experiment. Occasionally, 15 % resolving gels are
used to resolve proteins with small molecular sizes while 10 % gels are used to
resolve proteins with large molecular sizes. Gels were cast according to standard
protocols from the manufacturers (Bio-rad, USA). Firstly, 1 mm spacer plates and
short glass plates were wiped clean with autoclaved MilliQ water and KimWipes
(Kimberly-Clark Co., USA), after which they were used to form a glass cassette
sandwich using the casting frame. Varying amounts of autoclaved MilliQ water and
40 % Acrylamide/Bis solution, 37.5:1 mixture (Bio-rad, USA), 2.5 ml 1.5 M Tris-HCl,
pH 8.8, 0.1 ml 10 % SDS, 0.1 ml 10 % ammonium persulphate (APS) (Bio-rad, USA),
and 0.004 ml Tetramethylethylenediamine (TEMED) (Thermo Scientific, USA) were
added in the stated order to obtain 10 ml of resolving gel solution. The gel solution
58
Materials and Methods
was quickly mixed well by inverting the tube 10 times, following which the solution
was pipeted into the glass cassette sandwich until a gap of approximately 1.5 cm
between the top of the gel solution and top of the short plate was left. A layer of
autoclaved MilliQ water was used to overlay the gel. The resolving gel mixture was
then left to polymerise for 30 to 45 minutes.
To prepare a 4 ml stacking gel, 2.9 ml of autoclaved MilliQ water was
combined with 0.5 ml 40 % Acrylamide/Bis solution, 37.5:1 mixture (Bio-rad, USA),
0.5 ml 1.0 M Tris-HCl pH 6.8, 0.04 ml 10 % SDS, 0.04 ml 10 % APS, and 0.004 ml
TEMED in the stated order and mixed. Next, the gel mixture was poured over the
hardened resolving gel to the top of the short plate and the comb was placed between
the glass cassette sandwich and left to polymerise for 20 minutes.
4.3.2 Running SDS-PAGE
After both the resolving gel and stacking gel were polymerised, the entire gel
sandwich was placed in the clamping frame. Usually 2 gels were cast and run at the
same time; however, a buffer dam will be used when running only 1 gel. Once the 2
gel sandwiches were placed in perfect alignment in the clamping frame with the short
plates facing inwards, the arms of the clamping frame will be slid into place to secure
both gels. The entire set up was then placed in the Mini-Protean Tetra tank (Bio-rad,
USA) and the entire tank was filled with 1X running buffer (19 mM Tris; 144 mM
glycine; 0.075 % SDS; pH 8.3), after which samples were loaded with a pipette.
Samples were allowed to settle at the bottom of their respective wells before placing
the lid on the tank and connecting the lid’s electrical leads into the power supply (Biorad, USA). A constant voltage of 100 V was applied for approximately 1.5 hours,
depending on the size of the proteins of interest and resolving gel percentage.
59
Materials and Methods
4.3.3 Protein transfer from PAGE gel to Polyvinylidene fluoride (PVDF)
membrane
Following SDS-PAGE, the gels were removed from the tank and their gel
cassettes and the gels were prepared for transfer to a PVDF membrane. Briefly, the
fibre pads and filter papers pre-soaked in 1X transfer buffer (25 mM Tris; 192 mM
glycine; 0.01 % SDS; 20 % (v/v) methanol) were used to make a sandwich. The
cassette was first placed with the grey side down in a tray filled with 1X transfer
buffer and the sandwich was assembled in the following order: fibre pad, filter paper,
SDS-PAGE gel, PVDF membrane, filter paper and fibre pad. The PVDF membrane
was activated prior to adding to the sandwich by soaking in 100 % methanol for 5
seconds. A 15 ml Falcon tube was used to roll out air bubbles during each step in the
sandwich assembly. The sandwich was then clamped in place by the cassette and
placed in the trans-blot tank and filled to the brim with 1X transfer buffer. The entire
tank was then run at a constant voltage of 100 V for 60 minutes to 90 minutes at 4 °C.
4.3.4 Immunoblotting
All antibodies were purchased from Sigma Aldrich (secondary antibodies,
anti-β-actin), MBL International (anti-LC3), Abnova (p62) and Covance (GFP). After
transfer, the PVDF membrane was removed from the sandwich and placed in 10 ml of
5 % BSA / 1X TBST (50 mM Tris-HCl, pH 7.4; 150 mM NaCl; 0.1 % Tween 20)
solution per 2 PVDF membranes for blocking at room temperature for 2 hours,
following which the membrane was cut into various strips for incubation in primary
antibody-containing BSA solution at 4 °C overnight with gentle agitation. Primary
antibodies were usually diluted in 1: 1000 ratio in 5 % BSA/TBST solution, with the
exception of β-actin, which was diluted in a 1:2000 ratio. The following day, the
60
Materials and Methods
primary antibodies were drained out of the container and the membrane strips were
washed with 15 ml of 1X TBST solution 3 times for 10 minutes each, following
which horse radish peroxidise (HRP)-conjugated secondary antibodies were added in
a 1:2000 dilution and incubated at room temperature for 2 hours. After the 2-hour
incubation, the membranes were washed with 15 ml of 1X TBST solution 3 times for
10 minutes each.
4.3.5 Protein band detection
Subsequently, the membranes were then incubated in a 2 ml Working solution
containing equal parts of the Stable Peroxide Solution and Luminol/Enhancer
Solution from the SuperSignal West Pico Chemiluminescent Substrate (Pierce, USA)
for 1 minute. After removing the membranes from the Working solution excess
solution was removed by tapping the edge of the membrane on an absorbent tissue
before placing it in a film cassette, with the protein side facing up. A transparency
slide cut into the shape of the cassette was then placed on top of the membrane and
after ensuring that the transparency slide is clean a piece of X-ray film (Pierce, USA)
was gently placed on top of the membrane and the film was exposed for 60 seconds.
The film was then developed using appropriate developing solution and fixer using
the film developer machine (Kodak, USA). Various exposure times were attempted to
ensure sufficient exposure. A maximum of 10 minutes was used to expose a film.
4.3.6 Calculating band intensity from immunoblots
The films were scanned and the band intensities were calculated using the
Image J software (NIH, USA). Briefly, the bands were selected using the Rectangle
Selection tool and the profile plot of each lane was calculated by using the Plot Lanes
61
Materials and Methods
function. The Straight Line tool was selected and a straight line was drawn across
each peak to enclose each peak. Each peak was then selected and the Label Peaks
function was used to label each peak with its size being expressed as a percentage of
the total size of all the measured peaks. The percentage intensity of each peak was
then compared to the control peak to obtain each peak’s relative intensity after each
peak was normalised using respective peaks’ loading control. This data was then
plotted as a bar chart.
5. Starvation and chemical treatments
5.1 Starvation treatments
Chloroquine was purchased from Sigma Aldrich and dissolved in autoclaved
MilliQ water to give a stock solution 100 mM. For the set of experiments studying the
effects of chloroquine in starvation, 50 larvae were placed in a 9 cm petri dish with 25
ml of egg water. Larvae were not fed from 3 dpf to 8 dpf in order to exhaust the
nutrient supplies found in the yolk. Larvae were further starved from 8 dpf onwards
until all larvae in an experimental group died or upon reaching experimental end point,
whichever is earlier. Egg water was changed every other day during the entire period
from 3 dpf until the end of the experimental duration. Separate pipettes were used to
siphon off water in order to prevent cross-contamination from the fed groups as well
as from groups exposed to chloroquine. Larvae in the fed group were fed chicken egg
yolk suspension from 8 dpf onwards, where an entire egg yolk was ground and 50 ml
of autoclaved water was added to give a suspension. Usually 100 µl of egg yolk
suspension was fed to the larvae in the fed group daily. All larvae were reared at
28.5 °C in an incubator.
62
Materials and Methods
For the set of experiments studying intracellular EGFP-LC3 and mRFPEGFP-LC3 distribution during starvation, 15 larvae were placed in one well
containing 15 ml of egg water, using a 6-well plate. Egg water was changed every
other day and 20 µl of egg yolk suspension was given to the fed control group daily.
Larvae were starved until 12 dpf before processing for confocal microscopy imaging.
5.2 Chemical treatments
Rapamycin and everolimus were purchased from Sigma Aldrich. Each of
these was dissolved in 100 % DMSO to obtain stock solutions of 10 mM before being
used in actual chemical exposures. All stock solutions were kept in the dark by
wrapping the tubes with aluminium foil and stored at -20 °C. 2 dpf wild type larvae
and 3 dpf transgenic larvae were used in these experiments. 3 dpf transgenic larvae
were used so as to visualise intracellular GFP-LC3 distribution. Briefly, 15 larvae
were placed in each well in a 6-well plate and 10 ml of egg water was added to each
well. DMSO was used as a vehicle control and 100 % DMSO was added to yield a
final concentration of 0.01 %, while rapamycin, everolimus and chloroquine stock
solutions were added to yield a final concentration of 1 µM rapamycin or 1 µM
everolimus and 50 µM chloroquine. The 6-well plates were further wrapped using
aluminium foil to prevent light exposure and were incubated in a separate incubator at
28.5 °C for 48 hours. Fresh egg water and fresh chemicals were added after 24 hours
incubation. Larvae were rinsed with clean egg water twice before being fixed for
confocal microscopy imaging or being processed for immunoblotting. All liquid
wastes were pooled, stored and disposed of according to established chemical waste
disposal protocols in NUS.
63
Materials and Methods
5.3 Doxycycline induction of Xmrk and cmyc transgene expression
Doxycycline was purchased from Sigma Aldrich and the stock solution was
prepared by weighing and dissolving doxycycline powder in 40 ml of autoclaved
distilled water in 50 ml Falcon tubes. The tubes were wrapped in aluminium foil to
prevent light exposure and stored at -20 °C until use.
4 mpf adult transgenic and non-transgenic fish were screened, sorted and
placed in tanks containing 5 litres of egg water, with each tank containing 40 fish. All
tanks were placed in a dark room, and fish were fed twice daily with brine shrimp.
Doxycycline stock solution was added to tanks containing transgenic fish requiring
doxycycline-induced oncogene expression to yield a final concentration of 60 µg/ml
doxycycline. Water in the tanks was changed and fresh doxycycline was added every
other day until the end of the treatment period.
During the 3-week recovery period, water in the tanks was changed every
other day, but doxycycline was not added into any tank. The tanks were kept in the
same room and under dark conditions.
6. External photo-documentation of adult treated transgenic fish
Adult transgenic and non-transgenic fish were first anaesthetised in ice water
and placed in a plastic weigh boat marked with a 1 cm scale bar. To show the gross
internal morphology, the skin covering the belly was removed using a pair of scissors
and forceps before replacing the fish back in the plastic weigh boat. Photographs were
taken with a digital single-lens reflex camera, and images were processed using the
Adobe Photoshop software (Adobe, USA), with the cropped images retaining their
aspect ratio as the original images.
64
Materials and Methods
7. Cryosectioning
7.1 Cryostat sections of larvae
Larvae were fixed using 4 % paraformaldehyde in PBS (PFA/PBS) at 4 °C
overnight with gentle shaking in the dark by wrapping the tubes with aluminium foil,
following which the larvae were washed 3 times using PBST (3.2 mM Na2HPO4; 0.5
mM KH2PO4; 1.3 mM KCl; 135 mM NaCl; 0.05 % Tween 20, pH 7.4) for 10
minutes each. The fixed larvae were then transferred into molten 2 % bactoagar/5 %
sucrose in a cap detached from a microcentrifuge tube at 50 °C. Needles were used to
orientate the larvae with their heads facing upwards before the agar solidified. The
block was further trimmed using a razor after solidifying, and was transferred into 30 %
sucrose solution to be incubated overnight at 4 °C.
The block was subsequently placed on a frozen layer of Tissue Freezing
Medium (Jung, Germany) covering the flat surface of the tissue holder. It was
subsequently covered with a layer of Medium before freezing it using liquid nitrogen.
The entire assembly was then placed inside the Leica CM 1900 cryo-microtome
(Leica, USA) to equilibrate to -30 °C for 2 hours. 12 µm thick sections were made
and placed on Superfrost Plus slides (Fisher, USA) before being dried on a hot plate at
42 °C for about 1 to 2 hours. Light was excluded by making a cover using aluminium
foil and placing the cover over the slides.
Following this, slides were washed twice using PBST for 5 minutes each.
Slides were either processed for Hoechst staining or sealed immediately using 60 µl
FluorSave Reagent (Merck, USA) and covered using 22 X 60 mm cover slips. The
slides were left to dry at 4 °C overnight in slide holders.
65
Materials and Methods
7.2 Cryostat sections of adult tissues
Adult fish were sacrificed and fixed in 4 % PFA/PBS in the morning, and the
adult liver tissue and surrounding gut was removed from the fish and further fixed in
4 % PFA/PBS overnight with gentle shaking without exposing to light. The livers
were washed 3 times using PBST for 5 minutes each, before being placed on a layer
of Tissue Freezing Medium that covered the flat surface of the tissue holder. A layer
of Tissue Freezing Medium was layered on top of it and the entire assembly was
subsequently frozen using liquid nitrogen before being placed in the microtome to
equilibrate for 2 hours. Downstream procedures were identical to cyro-sectioning of
larvae.
7.3 Hoechst staining
Washed slides were immediately stained using Hoechst staining reagent. Each
slide was incubated with sufficient amount of Hoechst stain so that sections were in
contact with the staining solution, and left to incubate at room temperature for 20
minutes in the dark. Subsequently, the slides were tilted to remove the staining
solution and washed 4 times using PBST, for 10 minutes each. 60 µl of FluorSave
Reagent was then added to each slide and each slide was covered using 22 X 60 mm
cover slips. The slides were left to dry at 4 °C overnight in slide holders.
8. Confocal imaging
8.1 Live confocal imaging
Live larvae were first anaesthetised using 0.1 % phenoxy-ethanol (Sigma
Aldrich, USA) before being transferred to a glass bottom dish (MatTek Co., USA). 1
drop of molten 1.5 % low melting agarose gel was placed on top of each larva and the
66
Materials and Methods
larva was subsequently positioned using a pair of needles before the agarose solidified.
Larvae were positioned such that they were lying with their left sides on the bottom of
the glass bottom dish. Larvae were subsequently imaged for EGFP-LC3 expression
using the Zeiss LSM510 Meta scanning laser confocal microscope (Carl Zeiss Inc.,
Germany), using an excitation wavelength of 488 nm and 505-530 band-pass filters
with the Zeiss EC Plan-Neofluar 40X objectives with 2.5 times digital zoom.
Similarly, an excitation wavelength of 543 nm and a 590-620 band-pass filter were
used to image mRFP expression. Images were collected and processed using Zeiss’s
LSM Image Browser software (Carl Zeiss, Germany).
8.2 Confocal imaging of sections
Sections were imaged using the Carl Zeiss LSM510 Meta scanning laser
confocal microscope. Slides were placed with the cover slip-attached side facing
downwards towards the objectives. Images were taken in a similar manner as
described in section 8.1.
For Hoechst staining, images were taken using an excitation wavelength of
405 nm. Scanning for Hoechst staining was performed after scanning for EGFP and
mRFP signals. Images were all collected and processed using Zeiss’s LSM Image
Browser software as mentioned.
9. Bioinformatics and sequence analyses
The National Centre for Biotechnology Information (NCBI) database was
searched for Atg proteins sequences from mouse, rat, human, African clawed frog,
chicken, Caenorhabditis elegans, yeast and zebrafish. The blast2seq programme
freely
available
from
the
NCBI
website
67
Materials and Methods
(http://blast.ncbi.nlm.nih.gov/Blast.cgi?PROGRAM=blastp&BLAST_PROGRAMS=
blastp&PAGE_TYPE=BlastSearch&SHOW_DEFAULTS=on&BLAST_SPEC=blast
2seq&LINK_LOC=blasttab&LAST_PAGE=blastn&BLAST_INIT=blast2seq)
was
used to align human and zebrafish Atg proteins to produce Table 5.
Next,
the
ClustalW2
multiple
sequence
alignment
programme
(http://www.ebi.ac.uk/Tools/msa/clustalw2/) was used to generate multiple sequence
alignments of LC3 and Atg5, as well as phylogenetic trees, for sequence analyses.
10. Analysis of liver transcriptomic data by deep sequencing
Liver transcriptomic data by deep sequencing was available in the lab and
normalized data were used for analysis in this study. Only normal male liver
transcriptomic data were used. Atg genes were searched and the average transcripts
per million (TPM) were calculated and presented. Results are presented in Table 4.
68
Results
Results
69
Results
1. Conserved autophagy machinery in zebrafish
1.1 Atg gene expression in zebrafish
We first investigated atg gene expression in the zebrafish liver using available
liver transcriptomic data generated by deep sequencing. We found that the zebrafish
expresses numerous atg genes in the liver as shown in Table 4. Among them, atg13
has the highest average transcript per million (TPM) reads of 191.8964, while atg2
has the lowest average TPM reads of 0.2825. We did not find any transcripts
corresponding to atg7 and atg12.
1.2 Conservation of Atg proteins in zebrafish
To examine if the zebrafish genome encodes proteins required for the
autophagic process, we first performed a search for zebrafish Atg proteins in the
National Centre for Biotechnology Information (NCBI) database. As seen in Table 5,
the zebrafish indeed has Atg proteins that are crucial for autophagy, including a few
proteins such as Atg7 and Atg12 that are predicted from existing genomic information
available in NCBI. Moreover, the human and zebrafish Atg protein orthologues
displayed remarkable homology when they were aligned using the blast2seq
programme
found
in
the
NCBI
database
(http://blast.ncbi.nlm.nih.gov/Blast.cgi?PROGRAM=blastp&BLAST_PROGRAMS=
blastp&PAGE_TYPE=BlastSearch&SHOW_DEFAULTS=on&BLAST_SPEC=blast
2seq&LINK_LOC=blasttab&LAST_PAGE=blastn&BLAST_INIT=blast2seq), with
identity scores ranging from 50 % to 93 %.
As we are planning to generate a transgenic line expressing liver-specific
EGFP-LC3 and mRFP-EGFP-LC3, we decided to look at the LC3 protein in more
detail. Further amino acid alignments of LC3 from Mus musculus (mouse), Rattus
70
Results
norvegicus (rat), Homo sapiens (human), Xenopus laevis (African clawed frog),
Gallus gallus (chicken), Caenorhabditis elegans, and Saccharomyces cerevisiae
(yeast) with that of zebrafish showed high conservation of amino acid residues across
the entire LC3 protein (Figure 9A). Figure 9B also showed the evolutionary
relationship of LC3 from these organisms, suggesting that zebrafish LC3 is almost as
conserved as LC3 from mammalian species despite being in a different branch. Atg5
plays an important role in the regulation of autophagy, thus we are also interested to
find out if zebrafish Atg5 is similarly conserved. Amino acid alignment of Atg5
revealed that zebrafish Atg5 is highly similar to mammalian Atg5, as shown by the
highly conserved amino acid sequence as well as from phylogenetic tree analyses
(Figure 10). It can be concluded from the above evidences that zebrafish Atg proteins
are conserved and that they may play similar functions as those found in mammals.
71
Results
Table 4 atg gene expression in the zebrafish
atg gene
Refseq sequence
ulk1a
NM_001130631
atg2b
XM_001340472
atg3
NM_200022
atg4b
NM_001089352
atg4c
NM_001002103
atg4d
XM_001333057
atg5
NM_205618
beclin1 (atg6)
NM_200872
map1lc31a (lc3)
NM_214739
atg9a
NM_001083031
atg10
NM_001037124
atg13
NM_200433
atg14
NM_001024812
Average TPM
0.43
0.28
13.83
16.23
0.77
0.29
5.65
4.57
87.11
0.50
12.29
191.90
2.71
72
Results
Table 5 Comparison of zebrafish and human Atg proteins.
Name
Refseq sequence (Human)
Refseq sequence (Zebrafish)
Alignment scores
Identities Positives E-value
Atg1 (unc 51-like-kinase 1, Ulk1) NP_003556
NP_001124103
50 %
62 %
0
Atg3
NP_071933
NP_956316
82 %
90 %
2.00E-159
Atg4B
NP_037457
NP_001082821
73 %
83 %
3.00E-177
Atg5
Atg6 (Beclin1)
NP_004840
NP_003757
NP_991181
NP_957166
81 %
78 %
90 %
87 %
1.00E-138
0
Atg7
NP_006386
XP_002667343 (obsolete)
72 %
80 %
2.00E-138
Atg8 (LC3B)
Atg9
NP_073729
NP_076990
NP_955898
NP_001076500
93 %
70 %
96 %
80 %
3.00E-59
0
Atg10
NP_113670
NP_001032201
50 %
50 %
1.00E-61
Atg12
NP_004698
XP_699602
72 %
78 %
2.00E-48
Atg13
NP_001136145
NP_956727
74 %
83 %
0
Atg14
NP_055739
NP_001019983
67 %
85 %
0
Atg16
Vps34 (PI3K Class III)
NP_110430
NP_002638
NP_001017854
NP_001017550
68 %
84 %
83 %
91 %
5.00E-126
0
73
Results
A
B
Figure 9 Conservation of LC3 protein in zebrafish. (A) Amino acid sequences
from various species were compared using ClustalW2 software, with zebrafish LC3
showing evolutionarily conserved amino acid sequence. (B) A phylogram generated
using the same ClustalW2 software, showing inferred evolutionary relationships
among LC3 from various species. It also showed that zebrafish LC3 is closely related
to mammalian LC3.
74
Results
A
75
Results
B
Figure 10 Conservation of Atg5 protein in zebrafish. (A) Amino acid sequences
from various species were compared using ClustalW2 software, with zebrafish Atg5
showing evolutionarily conserved amino acid sequence. (B) A phylogram generated
using the same ClustalW2 software, showing inferred evolutionary relationships
among LC3 from various species. It also showed that zebrafish Atg5 is closely related
to mammalian LC3, and may probably be ancestral to rat, C.elegans and yeast LC3.
76
Results
1.3 Expression of autophagic genes and proteins in zebrafish
In order to investigate if the autophagic genes are expressed during zebrafish
embryogenesis, reverse-transcription polymerase chain reaction (RT-PCR) of lc3 and
atg5 using RNA isolated from wild-type 1-cell stage embryos through 96 hours post
fertilisation (hpf) larvae was performed to study their temporal expression patterns.
We detected both lc3 and atg5 transcripts from 1-cell stage embryos onwards,
suggesting maternal deposition of both transcripts (Figure 11). Activation of zygotic
expression of genes beyond 4 hpf might help maintain the expression level of both lc3
and atg5 mRNAs during early embryonic development. Such expression patterns
suggested that both lc3 and atg5 may play important roles during early embryonic
development, especially the first four hours after fertilisation, during the cleavage
stage of embryonic development.
We then investigated if expression of lc3 corresponded to protein expression
as well as phosphatidylethanolanime (PE) conjugation of LC3 by immunoblotting for
LC3 proteins using anti-rat LC3 antibodies. Cytoplasmic LC3, or LC3-I, exists as a 16
kD species, while the PE-conjugated LC3-II has a size of 14 kD due to the increased
migratory speed conferred by the PE moiety. Immunoblotting using proteins extracted
from wild-type whole larvae from 1 dpf to 5 dpf was shown in Figure 12. Results
revealed that LC3-I was expressed from 1 dpf onwards and that PE conjugation only
occurred after 1 dpf, as evidenced by the presence of the 14 kD LC3-II band in 2 dpf
samples. This result further proved that zebrafish LC3 undergoes similar posttranslational modification as its mammalian counterparts. Results further showed
increases in LC3-II levels from 2 dpf to 4 dpf, suggesting that overall autophagic flux
may be increased during this developmental period. However, there was a great
reduction of LC3-I and LC3-II levels at 5 dpf indicating increased rates of
77
Results
degradation in the lysosome from 5 dpf. We further observed that different batches of
larvae kept under similar conditions produced varying levels of LC3-I and LC3-II
throughout the first five days of development, suggesting that autophagy may be
tightly regulated by very small changes in the environment.
78
Results
A
B
Figure 11 Temporal expression patterns of lc3 and atg5. (A) Temporal expression
of lc3 transcripts revealed maternal deposition of lc3, suggesting the importance of
LC3 protein expression or autophagy function during early development. (B)
Temporal expression of atg5 transcripts revealed that, similar to lc3, maternal
deposition of atg5 occurred, highly suggesting an unknown role of Atg5 protein
function or autophagy function during early embryonic development. β-actin served
as loading control.
79
Results
Figure 12 LC3-I and LC3-II protein levels from 1 dpf to 5 dpf zebrafish larvae.
LC3-I conversion to LC3-II occurred from 2 dpf, as faint LC3-II levels can be
observed from the immunoblot. LC3-II levels increased from 2 dpf to 4 dpf,
suggesting induction of autophagy during this developmental period. Greatly reduced
LC3-I and LC3-II levels were observed at 5 dpf, suggesting increased degradation.
Results are representative of at least three independent experiments. β-actin served as
loading control.
80
Results
1.4 Conserved autophagic machinery in zebrafish
Having establishing that the zebrafish possesses atg genes and expresses Atg
proteins and that zebrafish LC3 undergoes similar post-translational modification like
its mammalian counterparts, we then assessed the functional conservation of the
zebrafish autophagic machinery and its amenability to chemical manipulation. In
mammalian and yeast cells, autophagy induction or increased autophagic flux is
manifested as an increase in LC3-II levels although a blockage at the final stage of
autophagy - lysosomal degradation - can also bring about an increase in LC3-II levels.
An increase in LC3-I with concomitant reduction in LC3-II levels are often seen when
autophagy inhibition is effected at the induction stage. As such, autophagy inducers
and inhibitors can be used to evaluate the functionality of the autophagic machinery in
the zebrafish by manipulating autophagy chemically. Rapamycin exerts its effect by
inhibiting the mTORC1 complex, relieving its inhibitory effect on the ULK1-FIP200Atg17 complex necessary for autophagy induction, thereby enhancing autophagosome
formation and LC3-II levels. However, this increase in LC3-II levels may not be
significant due to continuous lysosomal degradation of LC3-II in the autolysosome.
Thus the lysosomotropic alkaline chloroquine is also used simultaneously to discern
the changes in LC3-II levels as it effectively interferes with the final stage of
lysosomal degradation of sequestered materials. As chloroquine interferes with the
degradation of sequestered materials in the autolysosomes, it can also be used as an
autophagy inhibitor.
To assess autophagy induction, we exposed wild-type 2 dpf larvae to 1 µM
rapamycin for 8 hours. Another group of larvae were pre-treated with 50 µM
chloroquine for 1 hour before being exposed to rapamycin for 7 hours. In addition,
larvae were also exposed to 100% dimethyl sulfoxide (DMSO) vehicle in egg water
81
Results
as vehicle controls. At the end of the treatment, samples with 1 hour pre-treatment
with chloroquine followed by 7 hours of rapamycin exposure saw a more significant
increase in LC3-II levels than the samples that were exposed to rapamycin only
(Figure 13). This thus proves that the zebrafish autophagic machinery is conserved
and is also amendable to chemical treatment like its mammalian counterparts.
Autophagy is up-regulated by cells in response to cellular stress such as
starvation in order to survive the stress period. Accordingly, cells fail to survive the
stressor if they fail to induce autophagy as a means of cell survival. To evaluate this
survival function of autophagy, 8 dpf larvae were continuously exposed to 50 µM
chloroquine under both fed and starved conditions as shown in the schematic in
Figure 14, and their survival rates were noted. Continuous exposure to chloroquine
led to early deaths regardless whether the larvae were fed or starved, while larvae not
exposed to chloroquine survived better, with starved larvae surviving slightly better
than those that were fed. This may be correlated to the amount of debris present in the
water that feeding may have caused, with the fed larvae exposed to feed that may
have decayed in the duration of the treatment, leading to an accumulation of toxic
waste products and/or excess carbon dioxide in the water that caused the death of the
fed larvae. The early deaths of the larvae exposed to chloroquine may be due to an
inhibition of protein turnover that is essential for the larvae to survive starvation.
The above results led us to conclude that the autophagic machinery is highly
conserved and obviously functional in the zebrafish, and that it functions very
similarly to the mammalian and yeast autophagic machinery in ensuring cell survival
in the face of cellular and environmental stressors. These observations provide
biological basis for us to generate transgenic zebrafish expressing EGFP-LC3 and
mRFP-EGFP-LC3 fusion proteins to study autophagy in vivo.
82
Results
A
B
LC3-II
1.8
1.6
1.4
1.2
1
0.8
0.6
0.4
0.2
0
1.59
1.00
0.79
Water
DMSO
0.89
LC3-II
Rap
Rap & CQ
Figure 13 Autophagy induction by rapamycin. (A) Larvae treated with rapamycin
showed an increase in LC3-II levels, indicating increased autophagic flux, while
larvae treated with both rapamycin and chloroquine showed a significant increase in
LC3-II. Results were representative of at least three independent experiments. β-actin
served as loading control. (B) Quantification of LC3-II levels relative to β-actin
loading control from three independent experiments. DMSO: Dimethyl sulfoxide, Rap:
1 µM rapamycin, CQ: 50 µM chloroquine.
83
Results
A
B
Number of embryos survived
Survival rate of larvae exposed to
chloroquine
50
45
40
35
30
25
20
15
10
5
0
Feed, - CQ
Starve, - CQ
Feed, + CQ
Starve, +CQ
0
2 4 6 8 10 12 14 16
Days post fertilisation (dpf)
Figure 14 Autophagy induction is necessary for larvae to survive starvation.
(A) Schematic showing the experimental set-up used to investigate the relevance of
autophagy during starvation. (B) Survival rates of larvae exposed to 50 µM
chloroquine were greatly reduced regardless of whether they were fed or starved. Fed
and starved larvae in the absence of chloroquine survived better, although some
deaths still occurred. +CQ: addition of 50 µM chloroquine, -CQ: no chloroquine.
84
Results
2. Establishment and characterisation of Tg(fabp10:egfp-lc3) transgenic line
2.1 Establishing Tg(fabp10:egfp-lc3) transgenic line
As mentioned previously in the Introduction, autophagy is first studied in the
livers of animal models, and that the study of autophagy has not been extended to
zebrafish until recently by He et al. (He et al., 2009), therefore it would be of interest
to us to study autophagy in zebrafish livers. Furthermore, our lab has generated
zebrafish models of liver cancer by transgenic expression of oncogenes, thus it would
be interesting to study the role of autophagy in liver cancer. To analyse the role of
autophagy in hepato-carcinogenesis we set out to generate a zebrafish transgenic line
with constitutive liver-specific egfp-lc3 expression.
To generate the zebrafish transgenic line the egfp-lc3 expression cassette
created by Kabeya et al. (Kabeya et al., 2000) was placed downstream under the
control of a zebrafish liver-specific fabp10 promoter isolated by Her et al. (Her et al.,
2003). We incorporated the Ac/Ds transposon system to facilitate transgenesis,
generating the plasmid construct pDS-FABP10-EGFP-LC3 as shown in Figure 15.
We further compared LC3 protein sequences from rat and zebrafish to ensure high
homology between the two as the expression cassette utilises lc3 isolated from rat.
Sequence alignment in Figure 16 revealed high sequence identity (93 %) between the
two, especially at Gly120 where pre-LC3 protein is cleaved by Atg4B forming LC3-I,
before PE conjugation can take place. This thus ensures that exogenous EGFP-LC3
will also be recognised by zebrafish Atg4B and be processed in a similar manner as
endogenous zebrafish LC3.
85
Results
5'DS
poly A signals
rLC3
Amp
E GFP
pDS-FABP10-EGFP-LC3
8379 bp
3'DS
FABP 10
Figure 15 The pDS-FABP10-EGFP-LC3 plasmid used in the generation of
Tg(fabp10:egfp-lc3) transgenic line. The egfp-lc3 fragment was first subcloned into
the plasmid construct containing the fabp10 promoter, before subcloning the
fabp10:egfp-lc3 fragment into the plasmid construct containing Ds elements required
for transposition.
86
Results
Figure 16 Sequence identity exists between rat and zebrafish LC3 proteins.
ClustalW2 was used to study the amino acid sequence of zebrafish and rat LC3.
Glycine at position 120 is boxed in red.
87
Results
Microinjection of the pDS-FABP10-EGFP-LC3 plasmid into one-cell stage
embryos was performed to evaluate the ability of the plasmid in driving EGFP-LC3
expression in the liver. Faint EGFP-LC3 expression in the liver was first observed at 3
dpf after microinjection, with increasingly stronger EGFP-LC3 signals as the larvae
developed in size. Following this evaluation, the pDS-FABP10-EGFP-LC3 plasmid
was co-injected with Ac mRNA into one-cell stage embryos. As can be seen in Figure
17, injected larvae expressed EGFP-LC3 in the liver from 3 dpf and the EGFP-LC3
expression persisted until at least 6 dpf, allowing EGFP-LC3-expressing larvae to be
selected and bred to maturity for further screening.
Screening for founder fish was performed when the EGFP-LC3-positive
larvae were bred to maturity at around four months post fertilisation (mpf). Each
founder adult fish was crossed with its corresponding wild-type partner and their F1
progeny was collected and screened for EGFP-LC3 expression. In total, five out of 11
fish screened produced progeny with EGFP-LC3 expression. Out of the five founders,
only one founder (Line 1) was used for subsequent characterisation as the other four
showed ectopic EGFP-LC3 expression in the gut (Figure 18). Both EGFP-LC3
positive and negative larvae in Line 1 were processed for genotyping PCR. PCR
results showed germline transmission of the fabp10-egfp-lc3 expression cassette in
larvae with EGFP-LC3 expression in the liver, while their non-transgenic siblings did
not harbour the expression cassette in their genome (Figure 19). Progeny from Line 1
also expressed increasingly higher levels of EGFP-LC3 in the liver as evidenced from
their development from 3 dpf to 8 dpf. Immunoblotting for GFP in the liver protein
lysates from transgenic adult livers showed the expression of EGFP-LC3 fusion
protein as well as free EGFP while those from non-transgenic siblings did not (Figure
19).
88
Results
Figure 17 Liver-specific EGFP-LC3 expression after microinjection of pDSFABP10-EGFP-LC3. (A-C) Wild type (5 dpf) un-injected zebrafish larvae did not
express EGFP-LC3 in the liver. (D-F) Wild type zebrafish larva (6 dpf) with liverspecific expression after co-injecting pDS-FABP10-EGFP-LC3 and Ac mRNA.
89
Results
Figure 18 EGFP-LC3 expression in the progeny of the five founders. Five
different founders were identified producing progeny with EGFP-LC3 expression.
The founders were named according to the sequence in which they were crossed with
wild type fish. (A-D) Line 1 founder producing progeny with liver-specific EGFPLC3 expression, and was chosen for further characterisation. (E-T) Four other
founders produced progeny with ectopic EGFP-LC3 expression.
90
Results
A
B
91
Results
C
Figure 19 EGFP-LC3 positive larvae in Line 1 harbours germline transmission
of the fabp10:egfp-lc3 expression cassette. (A) Liver-specific EGFP-LC3 expression
increased in intensity from 3 dpf to 8 dpf. (B) Genotyping PCR revealed germline
transmission of the fabp10-egfp-lc3 expression cassette in transgenic progeny with
EGFP-LC3 expression, while the non-transgenic siblings without EGFP-LC3
expression did not harbour the expression cassette in their genome. β-actin served as
loading control. NTC: no template control. (C) Immunoblotting for EGFP-LC3 using
anti-GFP antibodies revealed the presence of a band for both EGFP-LC3 and free
EGFP in protein lysates from transgenic adult liver. No such bands were detected in
protein lysates obtained from non-transgenic adult liver. β-actin served as loading
control.
92
Results
2.2 Characterisation of Tg(fabp10:egfp-lc3) transgenic line
We further characterised the F1 progeny from Line 1 for their ability to
visualise the autophagic flux in vivo. Firstly, the EGFP-LC3 positive larvae were
subjected to starvation from 3 dpf onwards until 12 dpf when they have exhausted
their yolk supply in the yolk sac, while the control group were fed throughout the
entire period. Live imaging using confocal microscopy was performed to visualise
changes in the intracellular distribution of EGFP-LC3 in the liver. As expected,
punctate structures of EGFP-LC3 were observed in the livers of larvae subjected to
starvation, while the fed larvae showed diffused cytoplasmic distribution of EGFPLC3 (Figure 20). This result indicated that there was increased cellular turnover
during starvation and that the autophagic machinery was conserved in the zebrafish.
This result further proved that the autophagic flux can be studied in the zebrafish in
vivo using confocal microscopy.
We further investigated if the transgenic progeny were amendable to chemical
manipulation of autophagy using everolimus, an analogue of rapamycin, and
chloroquine. Larvae were exposed to everolimus, chloroquine and a combination of
everolimus and chloroquine for 48 hours and imaged using the confocal microscope
as shown in Figure 21. Control larvae displayed a diffused cytoplasmic distribution of
EGFP-LC3, while small EGFP-LC3 puncta were observed in larvae exposed to
everolimus, and to the combination of everolimus and chloroquine, as a result of an
induction in autophagy. However, there was no observable EGFP-LC3 puncta in
larvae exposed to chloroquine alone. This is unexpected as chloroquine exposure
should have increased the number of observable EGFP-LC3 puncta due to disrupted
lysosomal degradation of EGFP-LC3. However, immunoblotting for LC3 revealed
that chemical exposure indeed caused changes in the levels of LC3-I and LC3-II,
93
Results
similar to published data, as shown in Figure 22. The reason behind this is currently
unknown.
We also observed another phenomenon in this set of experiments. The EGFPLC3 puncta observed in larvae exposed to chemical inducers and inhibitors were
smaller than those observed when larvae were subjected to starvation. This may be
caused by different stimuli and therefore activation of subtly different signalling
pathways although they may converge on to the mTOR pathway to control autophagy
induction. Further studies are required to study this phenomenon.
94
Results
Figure 20 Autophagy induction via starvation. (A) Control larvae that did not
undergo starvation showed diffused cytoplasmic EGFP-LC3 distribution in
hepatocytes. (B) Hepatocytes from starved larvae produced EGFP-LC3 puncta under
starvation conditions, some of the autophagosomes are indicated by the white arrows.
Images were taken at 1,000X magnification.
95
Results
96
Results
Figure 21 EGFP-LC3 puncta formation in the presence of everolimus and
chloroquine. 3 dpf larvae were exposed to 1 µM of everolimus, 50 µM of
chloroquine or to a combination of both as indicated in the figure for 48 hours. 100%
DMSO was dissolved in egg water to a final concentration of 0.01% and acted as
vehicle control. (A-D) Hepatocytes showed diffused cytoplasmic EGFP-LC3
distribution in water and in vehicle control. (E-F) Small EGFP-LC3 puncta were
observed in hepatocytes of larvae exposed to 1 µM everolimus. (G-H) No significant
EGFP-LC3 puncta were observed in hepatocytes of larvae exposed to 50 µM
chloroquine. (I-J) Larvae exposed to both 1 µM everolimus and 50 µM chloroquine
had EGFP-LC3 puncta in their hepatocytes. EGFP-LC3 puncta were indicated with
white arrows. Cell nuclei were indicated by blue Hoechst staining. EVE: 1 µM
everolimus, CQ: 50 µM chloroquine. Images were taken at 1,000X magnification.
97
Results
A
B
LC3-II
11.34
12
10
8
6
LC3-II
4
2
1.00
0.93
Water
DMSO
1.38
1.80
0
EVE
CQ
EVE+CQ
Figure 22 LC3-I and LC3-II after chemical exposure for 48 hours. (A) A small
increase in LC3-II amounts were observed after larvae were exposed to everolimus,
while a small increase in LC3-I was observed in larvae exposed to chloroquine.
Larvae exposed to both had markedly increased LC3-I and LC3-II levels, indicative
of autophagy induction. β-actin served as loading control. (B) Quantification of LC3II levels relative to β-actin loading control from three independent experiments.
DMSO: Dimethyl sulfoxide, EVE: 1 µM everolimus, CQ: 50 µM chloroquine.
98
Results
3. Establishment and characterisation of Tg(fabp10:mrfp-egfp-lc3) transgenic line
3.1 Establishing Tg(fabp10:mrfp-egfp-lc3) transgenic line
As described earlier on the use of the mRFP-EGFP-LC3 fusion protein to
detect and measure autophagic flux, we also attempted to generate a transgenic line
with constitutively liver-specific mRFP-EGFP-LC3 expression. To generate this
transgenic line, we sub-cloned the mrfp-egfp-lc3 expression cassette created by
Kimura et al. (Kimura et al., 2007) downstream of the fabp10 promoter before subcloning the fabp10:mrfp-egfp-lc3 fragment into the pMDS6 plasmid construct to
generate the pDS-FABP10-mRFP-EGFP-LC3 plasmid construct as shown in Figure
23.
The plasmid was microinjected into one-cell stage embryos to evaluate the
ability of the plasmid in driving mrfp-egfp-lc3 expression in the liver. As expected,
both EGFP and mRFP signals were observed in the livers of 3 dpf microinjected
larvae. The plasmid was also co-injected with Ac mRNA into one-cell stage embryos
and larvae with both EGFP and mRFP expression as shown in Figure 24 were reared
to maturity at around 4 mpf before screening for founder fish. Two founder adult fish
were identified to produce progeny with liver-specific EGFP and mRFP fluorescence.
Genotyping PCR using genomic DNA isolated from the progeny with EGFP and
mRFP fluorescence showed germline transmission of the fabp10-mrfp-egfp-lc3
expression cassette, while their non-transgenic siblings did not (Figure 25).
99
Results
5'DS
poly A signals
rLC3
Amp
EGFP
pDS-FABP10-mRFP-EGFP-LC3
mRFP
9063 bp
3'DS
FABP -10
Figure 23 The pDS-FABP10-mRFP-EGFP-LC3 plasmid used in the generation
of Tg(fabp10:mrfp-egfp-lc3) transgenic line. The mrfp-egfp-lc3 fragment was first
subcloned into the plasmid construct containing the fabp10 promoter, before
subcloning the fabp10:mrfp-egfp-lc3 fragment into the plasmid construct containing
Ds elements required for transposition.
100
Results
Figure 24 Liver-specific mRFP-EGFP-LC3 expression after microinjecting the
pDS-FABP10-mRFP-EGFP-LC3 plasmid into one-cell stage embryos. (A-C)
Wild type (5 dpf) zebrafish larvae did not express mRFP-EGFP-LC3 in the liver. (D-F)
Wild type zebrafish larva (4 dpf) with liver-specific mRFP-EGFP-LC3 expression
after co-injecting the plasmid with Ac mRNA.
101
Results
102
Results
Figure 25 Transgenic larvae harbours germline transmission of the fabp10:mrfpegfp-lc3 expression cassette. Genotyping PCR revealed germline transmission of the
fabp10:mrfp-egfp-lc3 expression cassette in transgenic progeny with both EGFP and
mRFP expression, while the non-transgenic siblings without both EGFP and RFP
expression did not. Identical results were obtained from the two transgenic lines
isolated. β-actin served as loading control.
103
Results
3.2 Characterisation of Tg(fabp10:mrfp-egfp-lc3) transgenic line
We next proceeded to characterise the F1 progeny of Line 2 for their ability to
visualise the autophagic flux. Similar to the Tg(fabp10:egfp-lc3) transgenic line, the
two-colour transgenic progeny were also subjected to starvation from 3 dpf to 12 dpf,
while the control larvae were fed throughout the entire period. Live imaging using a
confocal microscope revealed the formation of obvious red puncta corresponding to
autolysosomes in starved larvae, although we did not observe any significant
increases in yellow puncta corresponding to autophagosomes in the starved larvae
(Figure 26). As this is an initial experiment, more studies need to be performed to
confirm this observation.
We also investigated if we can utilise the transgenic progeny to visualise the
autophagic flux under chemical treatment using everolimus and chloroquine.
Similarly, transgenic larvae were treated with everolimus, chloroquine and a
combination of everolimus and chloroquine for 48 hours and imaged using a confocal
microscope. Larvae treated with everolimus alone displayed increased numbers of red
puncta, although we did not observe a corresponding increase in yellow puncta in the
same larva (Figure 27). Larvae treated with chloroquine alone displayed more yellow
puncta corresponding to autophagosome formation without any concomitant increase
in red puncta indicative of blocked maturation of autophagosomes into autolysosomes,
while larvae treated with a combination of both everolimus and chloroquine had
increases in both red and yellow puncta, indicative of increased autophagic flux. As
this is also an initial experiment to test the validity of our transgenic line, more studies
are required to validate our results.
104
Results
Figure 26 Starvation induces autophagy. (A) Control larvae that did not undergo
starvation did not show obvious red and yellow puncta formation in the hepatocytes.
(B) Hepatocytes of starved larvae produced red puncta corresponding to
autolysosomes while there was no significant increase in the numbers of yellow
puncta corresponding to autophagosomes.
105
Results
106
Results
Figure 27 Red and yellow puncta formation in the presence of everolimus and
chloroquine. 3 dpf transgenic larvae were treated with 1 µM everolimus, 50 µM
chloroquine or a combination of both as indicated in the figure for 48 hours. Control
larvae were placed in egg water for 48 hours. (A-B) No obvious red and yellow
puncta were observed in control larvae. (C-D) Red puncta were observed without
significant yellow puncta formation when larvae were treated with everolimus. (E-F)
Slightly more yellow puncta were observed in larvae treated with 50 µM chloroquine.
(G-H) Significantly increased red and yellow puncta were observed in larvae treated
with both 1 µM everolimus and 50 µM chloroquine. Cell nuclei were indicated by
blue Hoechst staining. EVE: 1 µM everolimus, CQ: 50 µM chloroquine. Images were
taken at 400X magnification.
107
Results
4. Establishing the role of autophagy in liver cancer
With the establishment and characterisation of the Tg(fabp10:egfp-lc3)
transgenic line, we further investigated if we could extend the use of this transgenic
line to study the involvement of autophagy in liver cancer using our liver cancer
transgenic models (Huang et al., 2011; Li et al., 2011; Nguyen et al., 2011). Currently
we have developed two liver cancer models based on oncogene over-expression in the
liver. The Tg(fabp10:TA; TRE:Xmrk; krt4:GFP) and the Tg(fabp10:TA; TRE:cmyc,
krt4:RFP) transgenic lines were generated to study liver cancer, with the former line
developing hepatocellular carcinoma (HCC) and the latter line developing
hepatocellular adenoma (HA), as shown in Figure 28 (Huang et al., 2011). Moreover,
the use of the Tet-on inducible system further allows us to control the temporal
expression of the oncogene, allowing us to study tumour initiation, progression and
regression. This would also facilitate our study of the autophagy status and role in
liver tumourigenesis caused by oncogene over-expression.
108
Results
Figure 28 Over-expression of cmyc and Xmrk led to liver tumour formation in
stable transgenic zebrafish induced using 60 µg/ml doxycycline. Fish were treated
with 60 µg/ml doxycycline from 21 dpf onwards for 30 days. (A, D, G) External gross
morphology and histo-pathological analysis of a non-transgenic fish used as a control.
(B, E, H) Fish over-expressing cmyc developed an enlarged liver and developing
hepatocellular adenoma. (C, F, J) Fish over-expressing Xmrk developed a discoloured
and enlarged liver. Histo-pathological analysis revealed the fish developing
hepatocellular carcinoma. White dotted lines demarcate liver tissue from surrounding
tissue in the fish. HA: hepatocellular adenoma, HCC: hepatocellular carcinoma.
Figure adapted from Huang et al. and courtesy of Dr Zhen Li (Huang et al., 2011).
109
Results
4.1 Preliminary data implicating autophagy inhibition in liver cancer
Activation of autophagy often entails the suppression of the mTOR pathway
with a concomitant reduction in the phosphorylation of targets downstream of the
mTOR complex 1 (mTORC1) such as p70 ribosomal S6 kinase (S6K), and eukaryotic
initiation factor 4E binding protein 1 (4EBP1) (Corradetti and Guan, 2006). This
indirectly leads to reduced amounts of phosphorylated S6 ribosomal protein (p-S6), a
target of phospho-S6K. This relationship between mTORC1 activation and increased
amounts of p-S6 can be exploited as readout for mTOR activation. Immunoblotting of
lysates of both 4- and 8-month old Xmrk over-expressing zebrafish liver saw an
increase in p-S6, reflecting mTOR activation, while the untreated transgenic and nontransgenic sibling fish did not (Figure 29). An increase in LC3-I levels were also
observed in fish with elevated levels of p-S6, suggesting that autophagy may be
blocked due to activation of the mTOR signalling pathway. However, we were unable
to determine the amounts of LC3-II levels, thus it would be premature for us to
conclude that autophagy blockage was caused by mTOR activation. This observation
entails further investigation.
110
Results
Figure 29 mTOR activation may cause autophagy inhibition in fish with Xmrk
over-expression. Both 4 months and 8 months old transgenic fish treated with
doxycycline (Dox) showed elevated p-S6 levels, indicative of mTOR activation. LC3I levels were also elevated in these Dox-induced transgenic fish compared to wild
type treated or transgenic non-treated controls, suggesting autophagy inhibition. LC3II levels remained to be determined. Fish liver lysates were used for immunoblotting.
β-actin served as loading control.
111
Results
4.2 Establishing double transgenic lines with liver-specific oncogene overexpression and EGFP-LC3 expression to study the role of autophagy in liver
cancer
In order to study the autophagic flux in the zebrafish liver cancer models in
vivo, we crossed the Tg(fabp10:TA; TRE:Xmrk; krt4:GFP) and Tg(fabp10:TA;
TRE:cmyc; krt4:RFP) transgenic lines with Tg(fabp10:egfp-lc3) to produce double
transgenic fish, with Figure 30 detailing the progeny genotypes that resulted from a
cross involving Tg(fabp10:TA; TRE:Xmrk; krt4:GFP). Fish exposed to doxycycline
(treatment group) and their respective controls were grouped as in Figure 30. Progeny
resulting from crossing Tg(fabp10:TA; TRE:cmyc; krt4:RFP) and Tg(fabp10:egfp-lc3)
were treated in an identical manner as the above. The double transgenic fish were
induced to over-express Xmrk and cmyc respectively in order to develop liver cancer
upon doxycycline exposure when they were approximately 4 mpf. Fish were sampled
every two weeks to follow tumour initiation and progression, and doxycycline was
withdrawn after six and a half continuous weeks of doxycycline exposure to effect
tumour regression. Figure 31 showed the schedule for both doxycycline exposure as
well as sampling time points for the double transgenic fish. External gross
morphology, and internal gross morphology were taken note of and photodocumented, while liver tissue were taken from fish for processing for
immunoblotting and confocal microscopy.
112
Results
Figure 30 Schematic showing the genotypes of progeny resulting from crossing
Tg(fabp10:TA; TRE:Xmrk; krt4:GFP) with Tg(fabp10:egfp-lc3), and doxycycline
exposure and control groups. Fish expressing Xmrk and egfp-lc3 (L+X+) as well as
egfp-lc3 only (L+X-) were selected and grouped as shown in the schematic. Each
group were further divided into two sub-groups, one of which was treated with 60
µg/ml doxycycline (denoted by red dotted circles) while the other group were not,
therefore four different treatment groups will result. L: EGFP-LC3, X: Xmrk, D:
doxycycline.
113
Results
Adult double transgenic and control fish doxycycline treatment
1st sampling at 2.5 weeks
Continue treatment
2nd sampling at 4.5 weeks
Continue treatment
3rd sampling at 6.5 weeks
Recovery (no doxycycline added from here on)
4th sampling at 7.5 weeks
Continue recovery
5th sampling at 9.5 weeks
Figure 31 Schedule for doxycycline exposure and sampling time points.
Doxycycline treatment was continuous for six and a half weeks to allow tumour
formation and development, following which doxycycline was withdrawn and
tumours were allowed to regress. Sampling took place approximately every two
weeks, with the exception of the fourth sampling time point, where it took place one
week after doxycycline withdrawal.
114
Results
4.2.1 Tg(fabp10:TA; TRE:Xmrk; krt4:GFP)
Treated double transgenic fish over-expressing Xmrk developed liver tumours
as expected within six and a half weeks of doxycycline exposure, with the tumours
regressing following three weeks of doxycycline withdrawal (Figure 32). We
observed the double transgenic fish developing increasingly larger bellies due to
enlargement of the liver tissue as time progressed, with noticeable liver tissue
discolouration. Upon dissection, the liver tissue had a firmer consistency, unlike the
controls that had liver tissue with a softer consistency. These observations
corresponded to the severity of the liver tumours developed, with firmer consistency
positively correlated with severity of the tumours developed. The size of the liver
tissue returned to near normal when regression was induced with doxycycline
withdrawal, with the return of near normal liver consistency and colouration when
compared to the controls. Confocal imaging of double transgenic fish livers overexpressing Xmrk also revealed altered cell morphology with increasing severity as the
tumours developed. They also displayed diffused intracellular cytoplasmic EGFPLC3 and no obvious EGFP-LC3 puncta were observed. We further observed cell
morphology returning back to near normal as tumours regressed, with the reappearance of EGFP-LC3 puncta three weeks after tumours were induced to regress
(Figure 32).
We also examined the protein levels of LC3-I, LC3-II, p62 as well as EGFPLC3 and free EGFP in these fish (Figure 33). p62 levels can be used to estimate the
autophagy status as it interacts with LC3 via a LC3 recognition sequence (LRS) and is
a substrate for autophagic degradation and accumulation of p62 is correlated with
defective autophagy (Komatsu and Ichimura, 2010). We found that L+X+D+ fish had
increased amounts of EGFP-LC3 in their lysates throughout the entire experimental
115
Results
duration, despite the failure to detect significant EGFP-LC3 in the other controls. We
hypothesised that this may be due to a blockage in autophagy, causing EGFP-LC3 to
accumulate in L+X+D+ fish. However, the levels of free EGFP present did not concur
with our hypothesis as significant amounts of free EGFP could still be detected in
L+X+D+ fish during tumour induction as well as during regression. We did not detect
any changes in p62 levels, thus we could not corroborate p62 levels with autophagy
status. We were also unable to determine LC3-I and LC3-II levels accurately to arrive
at any conclusion. We could only hypothesise that autophagy may be inhibited during
liver tumour formation and development, as such, hypothesising that autophagy may
serve as an anti-cancer mechanism in Xmrk-induced liver tumour formation.
4.3.2 Tg(fabp10:TA; TRE:cmyc; krt4:RFP)
As with the Xmrk over-expressing double transgenic fish, the cmyc overexpressing double transgenic fish developed tumours that were visible due to their
severely enlarged livers which manifested as severely enlarged bellies (Figure 34).
However, we observed that the tumours that developed maintained their consistencies
as well as colouration when compared to the controls. Liver overgrowth was observed
in every fish that we sampled at the first three time points. Confocal imaging did not
reveal any differences in distribution of intracellular EGFP-LC3, thus it was difficult
for us to ascertain if there was any changes in the autophagic flux.
We also performed protein analyses on the fish livers. As shown in Figure 35,
double transgenic (L+M+D+) liver protein samples showed increased free EGFP
levels in samples during tumour regression, suggesting that there was increased
lysosomal degradation of EGFP-LC3, though the controls also had increased EGFP
levels in the fourth and fifth time points that corresponded to the tumour regression
116
Results
phase. The double transgenic protein samples also showed reduced p62 levels in the
fourth and fifth time points, highly suggesting increased lysosomal degradation or
increased autophagic flux. We also detected slight reduction in EGFP-LC3 levels
during tumour regression in L+M+D+ fish, suggesting that EGFP-LC3 was probably
degraded. This observation also corroborated with our observation and hypothesis that
autophagy may be de-repressed during tumour regression. Immunoblotting for LC3-I
and LC3-II did not provide any substantial evidence in supporting the notion that
autophagy was inhibited during tumour progression and they remain to be
investigated. Although the changes in protein levels in L+M+D+ recapitulated those
in L+M-D- controls, those from the two other control groups (L+M+D- ad L+M-D+)
were rather different, suggesting that different genotypes as well as addition of
doxycycline may have some effect on the fish and on the autophagic process, though
this phenomenon also remains to be investigated.
117
Results
118
Results
Figure 32 Tumour progression manifested as increasingly severe gross and
cellular morphology in transgenic fish over-expressing Xmrk. Fish were grouped
according to sampling time point. Liver overgrowth was monitored as treatment time
lapsed, with significant overgrowth by the third time point. Liver discolouration was
observed as well and corresponded to severity of tumour. Liver cell morphology was
also increasingly disrupted, correlating with severity of liver overgrowth and period of
doxycycline treatment. Doxycycline removal resulted in recovery and tumour
regression, as evidenced by liver size reduction. Cell morphology returned to near
normal concurrently. No significant EGFP-LC3 puncta were observed during tumour
initiation and progression. EGFP-LC3 puncta re-appeared in the fifth time point,
corresponding to recovery. Dotted lines demarcate liver tissue from surrounding
tissue. Fish were representative of three different individual fish that were sampled
per time point. Confocal images were taken at 1,000X magnification.
119
Results
Figure 33 Immunoblot analyses of Xmrk over-expressing fish showing increased
free EGFP. Increased free EGFP were observed in double transgenic fish during
tumour regression, suggesting either increased autophagic flux or increased lysosomal
degradation. Double transgenic fish also had visibly increased amounts of EGFP-LC3
compared to controls. There were no significant changes in p62 levels throughout the
entire experimental duration. Changes in LC3-I and LC3-II as well as EGFP-LC3
levels remain to be investigated. Fish liver lysates were used for immunoblotting.
Numbers refer to time point at which samples were collected. L: EGFP-LC3, X:
Xmrk, D: doxycycline.
120
Results
121
Results
Figure 34 Tumour progression in fish over-expressing cmyc, manifesting as liver
overgrowth and disruption to cell morphology. Fish were grouped according to
sampling time point. Liver overgrowth was monitored as treatment time lapsed, with
significant overgrowth by the third time point. Liver cell morphology was disrupted,
although not to the extent of fish over-expressing Xmrk. Doxycycline removal
resulted in recovery and tumour regression, as evidenced by liver size reduction and
return of near normal cell morphology. No significant EGFP-LC3 puncta were
observed in any of the fish sampled. Dotted lines demarcate liver tissue from
surrounding tissue. Fish were representative of three different individual fish that
were sampled per time point. Confocal images were taken at 1,000X magnification.
122
Results
Figure 35 Immunoblot analyses of cmyc over-expressing fish showing increased
free EGFP and reduced p62 levels during tumour regression. Increased free EGFP
and reduced p62 were observed in double transgenic fish during tumour regression,
suggesting either increased autophagic flux or increased lysosomal degradation.
Changes in LC3-I and LC3-II as well as EGFP-LC3 levels remain to be investigated.
Changes in protein levels in L+M+D+ recapitulated those of L+M-D-, however
L+M+D- and L+M-D+ did not recapitulate this trend, suggesting that doxycycline as
well as genotype may play subtle roles in autophagy regulation. Fish liver lysates
were used for immunoblotting. Numbers refer to time point at which samples were
collected. L: GFP-LC3, M: Cmyc, D: doxycycline.
123
Discussion
Discussion
124
Discussion
Autophagy is an evolutionarily conserved cellular process that involves the
degradation of a cell’s cytoplasm, long-lived proteins as well as organelles in the
lysosome; it enables cells to survive periods of stress such as starvation, and in the
case of cancer cells, metabolic and therapeutic stress (Klionsky, 2007; Mizushima et
al., 2008). As such, it is central for cells to be able to regulate autophagy for their own
survival during periods of cellular stress, and for cancer cells to co-opt autophagy for
its survival and subsequent proliferation and metastasis. Although autophagy was first
observed in mammalian hepatocytes, studies on autophagy have been carried out
mainly in yeast, until recently where more focus is directed towards its involvement in
diseases including cancer in humans (Klionsky, 2007). Autophagy has been found to
be evolutionary conserved from yeast to humans (Kourtis and Tavernarakis, 2009;
Rubinsztein et al., 2007), and in this study, we extended our analyses of autophagy to
the zebrafish, a model organism where essentially no studies on autophagy have been
done. We first characterised the autophagic machinery in zebrafish and found it to be
highly conserved in terms of function like its mammalian counterparts. We further
generated transgenic lines with constitutively liver-specific egfp-lc3 and mrfp-egfp-lc3
expression in order to study autophagic flux in the liver. In addition, with our newly
developed zebrafish models of liver cancer, we can, at the same time study the
involvement of autophagy in liver cancer caused by oncogene over-expression, and
expand our knowledge on how these oncogenes affect the regulation of autophagy and
how dysregulation of autophagy impact on tumour development.
1. Conserved autophagy machinery in zebrafish
To study autophagy in zebrafish, we first investigated the atg gene expression
in the zebrafish liver using transcriptomic data generated by deep sequencing (Table
125
Discussion
4). We found that the zebrafish indeed expresses numerous atg genes that encode the
core proteins required for functional autophagy. However, we failed to detect any
transcripts corresponding to atg7 and atg12, genes that are required for the autophagic
process. We hypothesise that lack of studies on autophagy in zebrafish may have been
the cause of this and propose the existence and nucleotide sequences of these two
genes be verified. We also found that the zebrafish expresses Atg proteins that have
high degrees of homology to human Atg proteins when their amino acid sequences
were compared (Table 5). We also found that Atg7 and Atg12 protein sequences were
derived from predicted genomic sequences, corroborating with our earlier findings
that zebrafish atg7 and atg12 genes were still currently uncharacterised. Studies can
be done to verify and characterise these proteins in detail after their nucleotide
sequences have been verified.
We further investigated the degree of similarity of LC3 and Atg5 proteins
across different mammalian and non-mammalian species, and found them to be highly
conserved in terms of amino acid sequences as well as types of amino acids at each
position (Figures 9 and 10). Phylogeny analyses further showed the close evolutionary
relationships between zebrafish and mammalian LC3 and Atg5, suggesting that the
function of zebrafish Atg proteins may be well-conserved compared to their
counterparts in other organisms. Thus, the zebrafish can be used as a complementary
organism to study autophagy as zebrafish develops externally and this feature of the
zebrafish facilitates such developmental studies involving autophagy. It can also be
used as a complementary organism to study the relationship between autophagy and
cancer.
We next set out to study the temporal lc3 and atg5 gene expression pattern
during development. We found that both lc3 and atg5 were maternally deposited as
126
Discussion
explained by the presence of their respective mRNA transcripts at the one-cell stage
(Figure 11), suggesting that autophagy may be involved in as early as during the
cleavage and blastula stages. This observation was also consistent with findings from
Tsukamoto et al. that autophagy is triggered by fertilisation and up-regulated in early
mouse embryos (Tsukamoto et al., 2008). More studies need to be performed to
confirm if the same occurs during zebrafish development. However, He et al. disputed
this hypothesis by providing evidence that other atg genes such as ulk1a, ulk1b, atg9a
and atg9b that are also crucial for autophagy are not expressed until later during
development (He et al., 2009). Hence our findings of maternal deposition of lc3 and
atg5 may also point to some unknown function or requirement for the presence of
these transcripts during development and this may be worthy of investigation.
Morpholino knockdown of lc3 and atg5 may shed light on the need for maternal
deposition of these two genes as well as their roles in embryonic development.
Furthermore, the presence of these two proteins in addition to Ulk1 and Atg9 in the
first 24 hpf should also be confirmed by immunoblotting.
Armed with this knowledge, we further investigated the temporal expression
of LC3 protein during development. We are also interested in finding out if zebrafish
LC3 undergoes conjugation with phosphatidylethanolamine (PE) like its mammalian
counterparts. We found that LC3-I was expressed in 1 dpf embryos, while PE
conjugation occurred only from 2 dpf onwards (Figure 12). This provided evidence
that LC3 undergoes identical post-translational modification like its mammalian
counterparts, and that LC3-I and LC3-II can be used as readout for autophagic flux
under appropriate experimental conditions. We also observed an increase in the levels
of LC3-II from 2 dpf to 4 dpf, highly suggesting autophagy induction and increased
autophagic flux during this period of time. Autophagy up-regulation may be required
127
Discussion
for larval development from 2 dpf to 4 dpf as zebrafish larvae undergo developmental
changes such as fin formation, pigment cell differentiation and circulatory system
formation among others from 2 dpf to 4 dpf (Kimmel et al., 1995). This observation
corroborated with findings that autophagy plays an important role in mammalian
development and cellular differentiation (Mizushima and Levine, 2010). We further
observed that LC3-II levels were greatly reduced at 5 dpf. This may be due to an
increased rate of lysosomal degradation or a reduction in the autophagic flux at 5 dpf,
and we hypothesised that this may be due to autophagic flux returning to a basal level
as larval morphogenesis comes to an end. Alternatively, this observation can be
interpreted as a blockage at the degradation step in the autophagic process from 2 dpf
to 4 dpf, resulting in an increase in LC3-II levels with a corresponding increase in
lysosomal degradation, leading to reduced LC3-II levels on 5 dpf. More studies are
proposed to understand the dynamics of autophagy during larval development,
especially during the transition from 4 dpf to 5 dpf.
We next assessed the conservation of autophagy function in the zebrafish
larvae using chemical inducers and inhibitors of autophagy (Figure 13). Increases in
LC3-II were observed after subjecting wild type larvae to rapamycin treatment,
indicative of autophagy induction, while co-incubation with chloroquine further
increased LC3-II amounts, which is consistent with published observations
(Mizushima et al., 2010). Chloroquine, a lysosomotropic agent, inhibits lysosomal
degradation, which includes degradation of materials sequestered and delivered to the
lysosome via autophagy as well as LC3-II that is found in the inner and outer leaflets
of the autophagosomes, resulting in increases in LC3-II levels. The use of chloroquine
alone can effect autophagy inhibition, which is the basis of our future set of
experiments that will be discussed in detail later.
128
Discussion
We also assessed the pro-survival function of autophagy by continuously
exposing wild type zebrafish larvae to chloroquine in the presence and absence of
food (Figure 14). We observed that continuous exposure to chloroquine lead to early
deaths regardless of whether the larvae were fed or starved, whereas larvae that were
not exposed to chloroquine had significantly higher survival rates. This observation
may be explained by chloroquine inhibiting lysosomal degradation, thereby disrupting
the intracellular turnover of amino acids and other nutrients and building blocks
required for cell survival. This set of experiments also recapitulated the observation of
reduced survival time of neonates deficient in atg5 by Kuma et al., where they
observed starved neonates deficient in atg5 did not survive as long as starved wild
type neonates (Kuma et al., 2004). They also observed reduced amino acid
concentrations in the plasma and tissues as well as signs of energy depletion in these
atg5-deficient neonates, suggesting that autophagy is required for the maintenance of
energy homeostasis during neonatal starvation.
Furthermore, Tsukamoto et al.
published the finding that autophagic degradation in the early mouse embryos was
necessary for preimplantation development in mammals and that deficient autophagy
may impair protein recycling in these embryos (Tsukamoto et al., 2008). Thus this
leads us to hypothesise that autophagy is required for cellular turnover during
embryonic development as well as survival during early neonatal starvation regardless
of nutritional availability. Furthermore, this published finding can also be correlated
to the finding that atg5 transcripts were found in 1-cell stage embryos via maternal
deposition, although we did not have evidence suggesting autophagic activity during
the cleavage stage. However we cannot refute the possibility of toxic side effects that
chloroquine exerts that can cause their early demise, thus we propose that similar
experiments using lower concentrations of chloroquine should be tried in future
129
Discussion
experiments in order to eliminate the possibility of chloroquine poisoning.
Experiments using other autophagy inhibitors can also be performed although these
inhibitors must be used with care as they are not specific for autophagy and may have
other off-target effects. Similarly, morpholino knockdown can be performed to
determine the requirement of autophagy genes, such as atg5 for zebrafish larval
development.
2. Establishment and characterisation of Tg(fabp10:egfp-lc3) transgenic line
We further went on to generate a transgenic zebrafish line with liver-specific
egfp-lc3 expression. We made use of the construct which was a kind gift from Kabeya
et al. in the generation of our transgenic line (Kabeya et al., 2000). As their construct
utilised rat lc3 gene, we first compared the degree of similarity between zebrafish and
rat LC3 amino acid sequence to ensure that rat LC3 will be correctly recognised and
processed by zebrafish Atg4. We then proceeded to sub-clone the egfp-lc3 fragment
into the plasmid containing the liver-specific fabp10 promoter so that egfp-lc3
expression is controlled by the fabp10 promoter. Finally, we utilised the Ac/Ds
transposition system to increase transgenesis rate, generating the plasmid as shown in
Figure 15. We found that the larvae had transient liver-specific EGFP expression after
microinjecting the plasmid, suggesting that the plasmid worked and that expression
was indeed restricted to the liver through the use of the fabp10 promoter. We then coinjected Ac mRNA and the plasmid and reared the larvae with EGFP-LC3 expression
(Figure 17). The use of the transposition system greatly enhanced transgenesis rates,
as five out of 11 potential founders produced larvae with EGFP expression (Figure
18). We further went on to verify the sites of EGFP-LC3 expression and found that
only one founder (named Line 1) produced progeny with liver-specific EGFP-LC3
130
Discussion
expression, while the other four founders expressed EGFP-LC3 in the gut or had nonspecific EGFP-LC3 expression in tissues surrounding the liver. This may be due to
the enhancer trap effect that is commonly observed when using transposition systems
such as the Tol2 transposition system (Kawakami et al., 2004; Parinov et al., 2004).
Thus we selected Line 1 for further characterisation. Genotyping PCR using primers
that amplify a DNA fragment containing egfp verified germline transmission of the
fabp10:egfp-lc3 expression cassette, while immunoblotting using anti-GFP antibodies
confirmed the expression of GFP-LC3 fusion protein in the transgenic line (Figure 19),
both evidences suggesting the successful integration and expression of the transgene
in the fish.
We next subjected the transgenic progeny to starvation and successfully
visualised EGFP-LC3 puncta formation in starved larvae, while the fed larvae serving
as control had diffused cytoplasmic EGFP-LC3 distribution (Figure 20). As zebrafish
larvae are able to survive without external nutrient supply for an extended period of
time, we deliberately starved them from 3 dpf to 12 dpf in order to exhaust the
nutrient supply found in the yolk to induce starvation, while the control larvae were
fed from 3 dpf onwards with egg yolk suspension. We observed fewer puncta induced
in the livers of zebrafish larvae when compared to cell culture conditions and this may
be due to obvious differences between in vitro and in vivo model systems. We also
observed that different larvae induced different degrees of EGFP-LC3 puncta
formation and attributed this to individual differences. In addition, our observations
were also consistent with those of Mizushima et al. using their GFP-LC3 transgenic
mice, where starvation-induced autophagic activity differed among individual mice
(Mizushima et al., 2004), suggesting individual differences and responses to the same
stimulus.
131
Discussion
To further prove that our transgenic line was also amendable to chemical
manipulation by autophagy inducers and inhibitors, we exposed transgenic larvae to
everolimus and chloroquine (Figure 21). Everolimus is an analogue of rapamycin with
similar potency and has been used in treating advanced renal cell carcinoma
(Garnock-Jones and Keating, 2009; Schuler et al., 1997). Thus, we are interested to
find out if everolimus can induce autophagy in vivo, and if we can adopt its use in
future experiments involving the liver cancer models. As expected, we observed
changes in intracellular EGFP-LC3 distribution when transgenic larvae were exposed
to everolimus, as well as a combination of everolimus and chloroquine for 48 hours.
We found that the EGFP-LC3 puncta that were induced in the hepatocytes were not as
numerous than those induced in in vitro systems, again reflecting the differences
between in vivo and in vitro systems. We further observed that the EGFP-LC3 puncta
that were induced by chemical treatment were significantly smaller than those
observed under starvation conditions. We hypothesised that the involvement of the
upstream AMPK pathway during starvation resulted in larger feedback to the mTOR
pathway, resulting in larger GFP-LC3 puncta formation. We further hypothesised that
as everolimus acts mainly on the mTORC1 complex, a smaller feedback may result
from such an inhibition, resulting in smaller EGFP-LC3 puncta formation. We also
hypothesised that this difference in puncta size may be due to the application of
different stimuli, with physiological stimuli like starvation resulting in larger puncta
being formed and smaller puncta formation in response to chemical stimuli.
Surprisingly, we did not observe any significant EGFP-LC3 puncta formation in
larvae treated with chloroquine. Chloroquine being a lysosomotropic agent inhibits
lysosomal degradation by increasing lysosomal pH, rendering lysosomal enzymes
inactive; hence the application of chloroquine should inhibit autophagic degradation
132
Discussion
and should result in an increase in EGFP-LC3 puncta observed. However, we did not
observe this increase and thus we put forth the hypothesis that exposure to
chloroquine alone may be insufficient to induce significant GFP-LC3 puncta
formation, although we observe increases in EGFP-LC3 puncta formation in larvae
treated with the combination of everolimus and chloroquine.
We turned to immunoblotting for LC3 proteins in an attempt to explain these
observations (Figure 22). We saw a significant increase in both LC3-I and LC3-II in
larvae exposed to both everolimus and chloroquine, suggesting that inhibition of the
lysosomal degradation by choloroquine and autophagy induction by everolimus were
successful. Although we saw an increase in LC3-II levels in larvae exposed to
everolimus alone, the slight increase can be explained by lysosomal degradation of
LC3-II in the autolysosomes. Interestingly, larvae exposed to chloroquine alone had
slightly increased amounts of LC3-II; however this does not explain why we were
unable to observe any significant GFP-LC3 puncta in the same group of larvae. More
studies need to be performed to provide a reasonable explanation for this observation.
We also further characterise our transgenic line with atg3 knockdown (Cui J,
unpublished), and proved that GFP fluorescence were absent in morphants, even
when exposed to both everolimus and cholorquine, while GFP fluorescence were still
observable in larvae microinjected with control morpholino. This set of experiments
further proved the value of our transgenic line in studying autophagy and the
relationship between different Atg proteins in the zebrafish. Our transgenic line, with
liver-specific EGFP-LC3 protein expression, finds value in studying the relevance of
autophagy in various liver diseases and pathologies.
133
Discussion
3. Establishment and characterisation of Tg(fabp10:mrfp-egfp-lc3) transgenic line
We also generated a transgenic zebrafish line with constitutively liver-specific
mrfp-egfp-lc3 expression using the pDS-FABP10-mRFP-EGFP-LC3 plasmid
construct shown in Figure 23. Microinjection of both the plasmid and Ac mRNA also
yielded larvae with liver-specific mRFP and EGFP expression (Figure 24), and two
founders producing transgenic progeny with germline transmission of fabp10:mrfpegfp-lc3 were isolated (Figure 25). One of the founders (Line 2) was randomly
selected for characterisation for its ability to measure autophagic flux under both
starvation and chemical treatment.
Starvation represents a cellular stressor that up-regulates autophagy, thus
increases in both the number of red puncta corresponding to autolysosomes and
yellow puncta corresponding to autophagosomes should be observed in the livers of
transgenic larvae under starvation conditions. We observed increased numbers of red
puncta, but we did not observe equivalent increases in the number of yellow puncta,
which is contrary to what is expected (Figure 26). We hypothesised that what we
observed may be due to increased rates of autophagosome-lysosome fusion and
maturation into autolysosomes and increased rates of lysosomal degradation, leading
to reduced numbers of autophagosomes observed. On the other hand, autophagy
levels returning to basal levels after prolonged periods of starvation may also result in
lesser number of autophagosomes compared to autolysosomes. Mizushima et al. using
their GFP-LC3 transgenic mice showed that more GFP-LC3 puncta were observed in
the livers following 24 hours starvation, whereas a lesser number of GFP-LC3 puncta
were observed in the livers after 48 hours starvation (Mizushima et al., 2004). We
hypothesised that zebrafish, similar to mice, limits the duration of autophagy upregulation to ensure that cellular integrity is not affected during starvation. This
134
Discussion
limitation may also apply to other autophagy-inducing stimuli as well. To prove this,
we intend to perform daily observations to find out if this applies to zebrafish larvae
as well.
We also subjected the transgenic larvae to identical chemical treatment
conditions to find out if they can be used to measure autophagic flux. Identical
conditions were applied to the transgenic larvae and we observed that transgenic
larvae treated with everolimus alone displayed increased numbers of red puncta
corresponding to autolysosomes without any concomitant increases in the number of
yellow puncta (Figure 27). Again, we hypothesised that autophagy levels may have
returned to basal levels after prolonged periods of induction, causing fewer
autophagosomes to be observed, or that there was an increased rate of
autophagosome-lysosome fusion and maturation, leading to reduced numbers of
yellow puncta observed. We further observed that larvae treated with chloroquine
alone displayed more yellow puncta compared to red puncta. This is indicative of
blocked maturation of autophagosomes into autolysosomes. We also observed
equivalent increases in the number of red and yellow puncta in transgenic larvae
treated with a combination of both everolimus and chloroquine, indicative of
increased autophagic flux. We intend to perform experiments involving shorter
incubation periods with everolimus alone to find out the cause behind the reduced
numbers of yellow puncta formation observed.
4. Establishing the role of autophagy in liver cancer
Our lab successfully generated two transgenic lines with inducible oncogene
over-expression, culminating in the development of liver tumours. In the
Tg(fabp10:TA; TRE:Xmrk; krt4:GFP) transgenic line, Xmrk over-expression resulted
135
Discussion
in the development of hepatocellular carcinoma (HCC), while cmyc over-expression
in the Tg(fabp10:TA; TRE:cmyc; krt4:RFP) transgenic line resulted in the
development of hepatocelluar adenoma (HA) (Figure 28). Thus we have on hand two
different transgenic lines that will result in the development of different types of liver
tumours and these two models are perfect to study the role of autophagy in these two
tumours. These two transgenic models further allow us to study the interaction
between Xmrk and cmyc over-expression and regulation of autophagy. As oncogene
over-expression in both lines are inducible by the addition of doxycycline, both
transgenic lines further allow us to study the course of tumour development and how
autophagy may have been affected at different time points in the course of tumour
development. Similarly, removal of doxycycline causes tumour regression in our liver
cancer models, and we can also study the relationship between autophagy and tumour
regression at the same time.
Our early study revealed that activation of the TOR pathway may be chronic
in Xmrk over-expressing fish as a result of Xmrk over-expression. We observed that
transgenic fish over-expressing Xmrk had elevated levels of phosphorylated S6 (p-S6)
protein, which serves as read out of TOR pathway activation, hence our conclusion
that TOR pathway may be activated in transgenic fish over-expressing Xmrk (Figure
29). Xmrk, an orthologue of the EGF receptor, was found to activate intracellular
signalling pathways such as the ras/raf/MEK/MAPK pathway, and can activate ERK
(Li et al., 2011). It has also been reported that EGFR signalling will directly and
indirectly activate the PI3K/Akt signalling pathway (Citri and Yarden, 2006) and that
the PI3K/Akt signalling pathway is a central regulator of the TOR complex and
therefore, coupled to the TOR pathway (Fabregat et al., 2007). Our results here
demonstrated that Xmrk over-expression also has an activating effect on the
136
Discussion
PI3K/Akt/TOR pathway, and this may have led to an inhibition on autophagy. We
also observe elevated levels of LC3-I in the same samples with elevated levels of p-S6;
however, we were unable to determine LC3-II levels as they seem to be non-existent
in our immunoblotting experiments, as a result, we were unable to determine if
autophagy has been inhibited in these same samples. This may be due to rapid
turnover of LC3-II in vivo. Thus more thorough investigation needs to be carried out
to validate if TOR signalling indeed has been activated and that autophagy is indeed
been inhibited in Xmrk over-expressing transgenic fish.
To determine if autophagy has been inhibited in these transgenic fish when the
oncogenes were over-expressed, we crossed the newly generated Tg(fabp10:egfp-lc3)
transgenic line with the two oncogene over-expressing transgenic lines to obtain
progeny that will express EGFP-LC3 both with and without oncogene expression. We
further separated them into groups with and without addition of doxycycline to induce
liver tumour formation, while trying to dissect the autophagic flux via confocal
microscopy. We observed severe overgrowth of liver tissue manifesting as enlarged
bellies in fish treated with doxyxycline to induce Xmrk and cmyc over-expression.
Xmrk over-expressing fish were found to develop increasingly larger liver tissue that
was gradually discoloured, probably due to enhanced proliferation and increasing
severity of the tumour developed (Figure 32). Accordingly, we observed increasingly
disrupted cell morphology when observing under the confocal microscope, again
corresponding to severity of the tumour. We did not find any EGFP-LC3 puncta
developing in the hepatocytes during doxycycline induction, instead, we found EGFPLC3 to be freely distributed in the cytoplasm, suggesting that autophagy may have
been inhibited. Upon doxycycline withdrawal, the size of the livers shrank to almost
their normal size, and the colouration returned to normal as recovery proceeded.
137
Discussion
EGFP-LC3 puncta re-emerged as recovery proceeded suggesting that autophagy may
have been re-activated once Xmrk over-expression was repressed. Our lab found that
apoptosis may have played an important role during tumour regression by eliminating
HCC cells; we also hypothesised that the newly generated normal hepatocytes may
have replaced these HCC cells and direct reversion of HCC cells to normal
hepatocytes may have occurred concurrently during tumour regression (Li et al.,
2011). We hypothesise that autophagy may have also been up-regulated during
tumour regression by both processes, to either aid apoptosis or aid cellular
differentiation to normal hepatocytes, although more studies are required to prove this.
We next examined LC3-I, LC3-II, p62, EGFP-LC3 as well as free EGFP
levels in the protein lysates (Figure 33). We observed increased amounts of EGFPLC3 in L+X+D+ protein lysates compared to controls (L+X+D-, L+X-D+ and L+XD-) throughout the entire experimental duration from tumour initiation through
tumour regression except at the last time point (corresponding to three weeks after
regression was induced). This observation may suggest that there may be an inhibition
of autophagy flux, especially at the later stages of autophagy involving lysosomal
degradation, therefore manifesting as significantly increased amounts of GFP-LC3 as
observed in immunoblots. This increase in EGFP-LC3 was also evident from confocal
microscopy imaging, where L+X+D+ livers displayed diffused intracellular GFP-LC3
distribution. The reduction in EGFP-LC3 levels three weeks after tumour regression
was induced corroborated with the re-appearance of EGFP-LC3 puncta, suggesting
the blockage of autophagic flux may have been relieved and that autophagy may have
been involved in recovering from tumourigenesis and generation of normal
hepatocytes. However, we did not observe any reduction in free EGFP in L+X+D+
lysates during tumour initiation and progression suggesting blocked autophagic
138
Discussion
degradation of EGFP-LC3, thus we were unable to validate if there was indeed a
blockage in autophagy caused by Xmrk over-expression. Increases in p62 levels are
usually associated with blocked autophagic flux since p62 is a substrate for autophagy;
however, we failed to detect any significant changes in p62 levels across samples and
across time points to further verify if autophagy was indeed blocked. We also failed to
detect LC3-II accurately in most of our samples, thus we could only hypothesise that
autophagy may have been blocked during tumour initiation and progression. More
work needs to be done in order to verify our hypothesis of autophagy being an anticancer mechanism and that it is inhibited during tumourigenesis.
We also observed severe liver tissue overgrowth manifesting as enlarged
bellies in cmyc over-expressing fish, although we did not detect any tissue
discolouration, suggesting that the tumours that developed were not as malignant as
those that arise from Xmrk over-expression, although the liver tissue overgrowth was
due to hyper-proliferation of hepatocytes (Figure 34). This was confirmed by histopathological analyses where liver tumours that arise from cmyc over-expression were
indeed HA (Figure 28). Confocal imaging did not reveal any differences in the
intracellular distribution of EGFP-LC3 in the hepatoytes either, thus we were unable
to determine the autophagy status in these tumours. We also carried out
immunoblotting for several proteins, including EGFP, to determine autophagic flux in
these fish (Figure 35). We observed increased p62 levels during tumour induction,
highly suggestive of autophagy repression. Elevated p62 levels could be a driving
force for tumourigenesis by enhancing oxidative stress and genomic instability as well
as activating the non-canonical NF-κB pathway, suggesting that autophagy downregulation may be favoured during tumourigenesis in order to elevate p62 levels
(Mathew et al., 2009). Accordingly we observed reduced p62 levels and increased
139
Discussion
levels of free EGFP in cmyc over-expressing fish during recovery, suggesting
autophagy inhibition may have been relieved to promote recovery. However, the role
of autophagy during cellular recovery remains to be investigated; although we
hypothesise that enhanced autophagy may serve to rid the cell of damaging
superfluous protein aggregates thereby relieving cells of oxidative stress. It may also
be involved in activating tumour-suppressing pathways as well as cellular remodelling
or simply aiding apoptosis in riding tumourigenic hepatocytes. Immunoblotting for
LC3-I and LC3-II were not very successful as we were unable to detect them
accurately, thus we were unable to further furnish supporting evidence involving
changes in LC3-II levels for this phenomenon.
Furthermore, we found difficulty in detecting LC3-I and LC3-II levels in
immunoblots, and we attributed this to the dynamic nature of the autophagic flux as a
whole. We suggest adding chloroquine 24 hours prior to sacrificing the fish in an
attempt to visualise and quantify LC3-II levels. We also hope to make use of the
Tg(fabp10:mrfp-egfp-lc3) transgenic line to visualise the autophagic flux and hope
that both transgenic lines will complement each other.
5. Future work and conclusions
In this study we attempted to characterise autophagy in zebrafish and found
that some autophagy genes like lc3 and atg5 were maternally deposited although no
detectable autophagic activity was detected at the early stages of embryonic
development. This presents an opportunity to study the relevance of these two
autophagy genes during early embryonic development. This also provides an
opportunity for us to learn if these proteins have other functions apart from being
involved in autophagy. Our experiments further showed that autophagy inhibition via
140
Discussion
chloroquine led to catastrophic consequences regardless of larvae being fed or starved,
further proving that autophagy plays a very important role in cell survival whether
nutrients are available, although we have to prove that larval deaths were due to
defective cellular turnover and not due to chloroquine toxicity.
We
successfully
generated
and
characterised
the
transgenic
line
Tg(fabp10:egfp-lc3) with liver-specific EGFP-LC3 fusion protein expression in order
to visualise autophagic flux in real time as well as to provide supporting evidence for
our immunoblotting findings. We further made use of this transgenic line to study the
role of autophagy in liver tumourigenesis by crossing this transgenic line with existing
transgenic lines with inducible oncogene over-expression. We found evidence that
autophagy may be inhibited during tumourigenesis, regardless of which oncogene was
being over-expressed, and that its inhibition may be relieved once oncogene overexpression ceased. To further prove our hypothesis we need to investigate LC3-I and
LC3-II levels in more detail, as we were unable to detect with accuracy LC3-I and
LC3-II levels in our samples. Our next step also entails further validation of p-S6
levels in an attempt to validate TOR signalling pathway activation, as well as to find
out which pathways were activated and how these pathways cross-talk with the
autophagy machinery to invoke autophagy inhibition during tumourigenesis. We also
intend to manipulate autophagy during liver tumourigenesis using chemical inducers
and inhibitors. To further prove our hypothesis that autophagy may be inhibited
during liver carcinogenesis, we intend to induce autophagy by exposing the double
transgenic fish to everolimus to find out if such an induction will lead to slower
tumour initiation and progression.
We also generated the transgenic line Tg(fabp10:mrfp-egfp-lc3) in order to
visualise the autophagic flux more accurately. This is the first transgenic model
141
Discussion
utilising differences in the propensity of EGFP and mRFP to fluoresce in acidic
conditions in order to visualise the autophagic flux from autophagosome formation to
maturation into autolysosomes. Although more in-depth characterisation is still
required to validate the functionality of our model, this model will in the future serve
the function of verifying our data on autophagy inhibition in our liver cancer models
as well as on studying autophagy in various liver diseases.
We believe that our study provides an impetus for more research to be carried
out in this area and that our data shed more light on the role of autophagy in liver
tumourigenesis. Moreover, our study provides initial clues that will help improve
current therapeutic options by designing combinatorial chemotherapy for liver cancer
in humans using autophagy inducers or inhibitors at the appropriate stages of
treatment together with established chemotherapeutic drugs.
142
References
References
143
References
Amati, B. (2001). Integrating Myc and TGF-beta signalling in cell-cycle control. Nat
Cell Biol 3, E112-113.
Amati, B., Alevizopoulos, K., and Vlach, J. (1998). Myc and the cell cycle. Front
Biosci 3, d250-268.
Amatruda, J.F., and Patton, E.E. (2008). Genetic models of cancer in zebrafish. Int
Rev Cell Mol Biol 271, 1-34.
Amatruda, J.F., Shepard, J.L., Stern, H.M., and Zon, L.I. (2002). Zebrafish as a cancer
model system. Cancer Cell 1, 229-231.
Apel, A., Herr, I., Schwarz, H., Rodemann, H.P., and Mayer, A. (2008). Blocked
autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res 68,
1485-1494.
Avila, M.A., Berasain, C., Sangro, B., and Prieto, J. (2006). New therapies for
hepatocellular carcinoma. Oncogene 25, 3866-3884.
Bampton, E.T., Goemans, C.G., Niranjan, D., Mizushima, N., and Tolkovsky, A.M.
(2005). The dynamics of autophagy visualized in live cells: from autophagosome
formation to fusion with endo/lysosomes. Autophagy 1, 23-36.
Barbazuk, W.B., Korf, I., Kadavi, C., Heyen, J., Tate, S., Wun, E., Bedell, J.A.,
McPherson, J.D., and Johnson, S.L. (2000). The syntenic relationship of the zebrafish
and human genomes. Genome Res 10, 1351-1358.
Berasain, C., Castillo, J., Prieto, J., and Avila, M.A. (2007). New molecular targets for
hepatocellular carcinoma: the ErbB1 signaling system. Liver Int 27, 174-185.
Berasain, C., Perugorria, M.J., Latasa, M.U., Castillo, J., Goni, S., Santamaria, M.,
Prieto, J., and Avila, M.A. (2009). The epidermal growth factor receptor: a link
between inflammation and liver cancer. Exp Biol Med (Maywood) 234, 713-725.
Berghmans, S., Murphey, R.D., Wienholds, E., Neuberg, D., Kutok, J.L., Fletcher,
C.D., Morris, J.P., Liu, T.X., Schulte-Merker, S., Kanki, J.P., et al. (2005). tp53
mutant zebrafish develop malignant peripheral nerve sheath tumors. Proc Natl Acad
Sci U S A 102, 407-412.
Blackwood, E.M., and Eisenman, R.N. (1991). Max: a helix-loop-helix zipper protein
that forms a sequence-specific DNA-binding complex with Myc. Science 251, 12111217.
Blommaart, E.F., Krause, U., Schellens, J.P., Vreeling-Sindelarova, H., and Meijer,
A.J. (1997). The phosphatidylinositol 3-kinase inhibitors wortmannin and LY294002
inhibit autophagy in isolated rat hepatocytes. Eur J Biochem 243, 240-246.
Boya, P., Gonzalez-Polo, R.A., Casares, N., Perfettini, J.L., Dessen, P., Larochette, N.,
Metivier, D., Meley, D., Souquere, S., Yoshimori, T., et al. (2005). Inhibition of
macroautophagy triggers apoptosis. Mol Cell Biol 25, 1025-1040.
144
References
Brech, A., Ahlquist, T., Lothe, R.A., and Stenmark, H. (2009). Autophagy in tumour
suppression and promotion. Mol Oncol 3, 366-375.
Breuhahn, K., Longerich, T., and Schirmacher, P. (2006). Dysregulation of growth
factor signaling in human hepatocellular carcinoma. Oncogene 25, 3787-3800.
Calvisi, D.F., and Thorgeirsson, S.S. (2005). Molecular mechanisms of
hepatocarcinogenesis in transgenic mouse models of liver cancer. Toxicol Pathol 33,
181-184.
Castillo, J., Erroba, E., Perugorria, M.J., Santamaria, M., Lee, D.C., Prieto, J., Avila,
M.A., and Berasain, C. (2006). Amphiregulin contributes to the transformed
phenotype of human hepatocellular carcinoma cells. Cancer Res 66, 6129-6138.
Chan, E.Y., Kir, S., and Tooze, S.A. (2007). siRNA screening of the kinome identifies
ULK1 as a multidomain modulator of autophagy. J Biol Chem 282, 25464-25474.
Chen, N., and Debnath, J. (2010). Autophagy and tumorigenesis. FEBS Lett 584,
1427-1435.
Chen, S., Rehman, S.K., Zhang, W., Wen, A., Yao, L., and Zhang, J. (2010).
Autophagy is a therapeutic target in anticancer drug resistance. Biochim Biophys Acta
1806, 220-229.
Citri, A., and Yarden, Y. (2006). EGF-ERBB signalling: towards the systems level.
Nat Rev Mol Cell Biol 7, 505-516.
Corradetti, M.N., and Guan, K.L. (2006). Upstream of the mammalian target of
rapamycin: do all roads pass through mTOR? Oncogene 25, 6347-6360.
Corradetti, M.N., Inoki, K., Bardeesy, N., DePinho, R.A., and Guan, K.L. (2004).
Regulation of the TSC pathway by LKB1: evidence of a molecular link between
tuberous sclerosis complex and Peutz-Jeghers syndrome. Genes Dev 18, 1533-1538.
Dang, C.V. (1999). c-Myc target genes involved in cell growth, apoptosis, and
metabolism. Mol Cell Biol 19, 1-11.
Dang, C.V., O'Donnell K, A., and Juopperi, T. (2005). The great MYC escape in
tumorigenesis. Cancer Cell 8, 177-178.
Davidson, A.E., Balciunas, D., Mohn, D., Shaffer, J., Hermanson, S., Sivasubbu, S.,
Cliff, M.P., Hackett, P.B., and Ekker, S.C. (2003). Efficient gene delivery and gene
expression in zebrafish using the Sleeping Beauty transposon. Dev Biol 263, 191-202.
Degenhardt, K., Mathew, R., Beaudoin, B., Bray, K., Anderson, D., Chen, G.,
Mukherjee, C., Shi, Y., Gelinas, C., Fan, Y., et al. (2006). Autophagy promotes tumor
cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 10,
51-64.
Ding, Z.B., Shi, Y.H., Zhou, J., Qiu, S.J., Xu, Y., Dai, Z., Shi, G.M., Wang, X.Y., Ke,
A.W., Wu, B., et al. (2008). Association of autophagy defect with a malignant
145
References
phenotype and poor prognosis of hepatocellular carcinoma. Cancer Res 68, 91679175.
Dooley, K., and Zon, L.I. (2000). Zebrafish: a model system for the study of human
disease. Curr Opin Genet Dev 10, 252-256.
Driever, W., Solnica-Krezel, L., Schier, A.F., Neuhauss, S.C., Malicki, J., Stemple,
D.L., Stainier, D.Y., Zwartkruis, F., Abdelilah, S., Rangini, Z., et al. (1996). A
genetic screen for mutations affecting embryogenesis in zebrafish. Development 123,
37-46.
Eilers, M. (1999). Control of cell proliferation by Myc family genes. Mol Cells 9, 1-6.
Eilers, M., and Eisenman, R.N. (2008). Myc's broad reach. Genes Dev 22, 2755-2766.
Emelyanov, A., Gao, Y., Naqvi, N.I., and Parinov, S. (2006). Trans-kingdom
transposition of the maize dissociation element. Genetics 174, 1095-1104.
Esclatine, A., Chaumorcel, M., and Codogno, P. (2009). Macroautophagy signaling
and regulation. Curr Top Microbiol Immunol 335, 33-70.
Fabregat, I., Roncero, C., and Fernandez, M. (2007). Survival and apoptosis: a
dysregulated balance in liver cancer. Liver Int 27, 155-162.
Feitsma, H., and Cuppen, E. (2008). Zebrafish as a cancer model. Mol Cancer Res 6,
685-694.
Feng, H., Langenau, D.M., Madge, J.A., Quinkertz, A., Gutierrez, A., Neuberg, D.S.,
Kanki, J.P., and Look, A.T. (2007). Heat-shock induction of T-cell
lymphoma/leukaemia in conditional Cre/lox-regulated transgenic zebrafish. Br J
Haematol 138, 169-175.
Fujita, N., Itoh, T., Omori, H., Fukuda, M., Noda, T., and Yoshimori, T. (2008). The
Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in
autophagy. Mol Biol Cell 19, 2092-2100.
Fung, C., Lock, R., Gao, S., Salas, E., and Debnath, J. (2008). Induction of autophagy
during extracellular matrix detachment promotes cell survival. Mol Biol Cell 19, 797806.
Garnock-Jones, K.P., and Keating, G.M. (2009). Everolimus: in advanced renal cell
carcinoma. Drugs 69, 2115-2124.
Geisler, R., Rauch, G.J., Baier, H., van Bebber, F., Bross, L., Dekens, M.P., Finger,
K., Fricke, C., Gates, M.A., Geiger, H., et al. (1999). A radiation hybrid map of the
zebrafish genome. Nat Genet 23, 86-89.
Giansanti, V., Torriglia, A., and Scovassi, A.I. (2011). Conversation between
apoptosis and autophagy: "Is it your turn or mine?" Apoptosis 16, 321-333.
146
References
Glucksmann, A. (1965). Cell death in normal development. Arch Biol (Liege) 76,
419-437.
Gossen, M., and Bujard, H. (1992). Tight control of gene expression in mammalian
cells by tetracycline-responsive promoters. Proc Natl Acad Sci U S A 89, 5547-5551.
Haffter, P., Granato, M., Brand, M., Mullins, M.C., Hammerschmidt, M., Kane, D.A.,
Odenthal, J., van Eeden, F.J., Jiang, Y.J., Heisenberg, C.P., et al. (1996). The
identification of genes with unique and essential functions in the development of the
zebrafish, Danio rerio. Development 123, 1-36.
Hara, T., and Mizushima, N. (2009). Role of ULK-FIP200 complex in mammalian
autophagy: FIP200, a counterpart of yeast Atg17? Autophagy 5, 85-87.
Hara, T., Takamura, A., Kishi, C., Iemura, S., Natsume, T., Guan, J.L., and
Mizushima, N. (2008). FIP200, a ULK-interacting protein, is required for
autophagosome formation in mammalian cells. J Cell Biol 181, 497-510.
He, C., Bartholomew, C.R., Zhou, W., and Klionsky, D.J. (2009). Assaying
autophagic activity in transgenic GFP-Lc3 and GFP-Gabarap zebrafish embryos.
Autophagy 5, 520-526.
He, C., Song, H., Yorimitsu, T., Monastyrska, I., Yen, W.L., Legakis, J.E., and
Klionsky, D.J. (2006). Recruitment of Atg9 to the preautophagosomal structure by
Atg11 is essential for selective autophagy in budding yeast. J Cell Biol 175, 925-935.
Hemelaar, J., Lelyveld, V.S., Kessler, B.M., and Ploegh, H.L. (2003). A single
protease, Apg4B, is specific for the autophagy-related ubiquitin-like proteins GATE16, MAP1-LC3, GABARAP, and Apg8L. J Biol Chem 278, 51841-51850.
Her, G.M., Chiang, C.C., Chen, W.Y., and Wu, J.L. (2003). In vivo studies of livertype fatty acid binding protein (L-FABP) gene expression in liver of transgenic
zebrafish (Danio rerio). FEBS Lett 538, 125-133.
Higashijima, S., Okamoto, H., Ueno, N., Hotta, Y., and Eguchi, G. (1997). Highfrequency generation of transgenic zebrafish which reliably express GFP in whole
muscles or the whole body by using promoters of zebrafish origin. Dev Biol 192, 289299.
Huang, X., Nguyen, A.T., Li, Z., Emelyanov, A., Parinov, S., and Gong, Z. (2011).
One step forward: The use of transgenic zebrafish tumor model in drug screens. Birth
Defects Res C Embryo Today 93, 173-181.
Hukriede, N.A., Joly, L., Tsang, M., Miles, J., Tellis, P., Epstein, J.A., Barbazuk,
W.B., Li, F.N., Paw, B., Postlethwait, J.H., et al. (1999). Radiation hybrid mapping of
the zebrafish genome. Proc Natl Acad Sci U S A 96, 9745-9750.
Ichimura, Y., Imamura, Y., Emoto, K., Umeda, M., Noda, T., and Ohsumi, Y. (2004).
In vivo and in vitro reconstitution of Atg8 conjugation essential for autophagy. J Biol
Chem 279, 40584-40592.
147
References
Ichimura, Y., Kirisako, T., Takao, T., Satomi, Y., Shimonishi, Y., Ishihara, N.,
Mizushima, N., Tanida, I., Kominami, E., Ohsumi, M., et al. (2000). A ubiquitin-like
system mediates protein lipidation. Nature 408, 488-492.
Ivics, Z., Kaufman, C.D., Zayed, H., Miskey, C., Walisko, O., and Izsvak, Z. (2004).
The Sleeping Beauty transposable element: evolution, regulation and genetic
applications. Curr Issues Mol Biol 6, 43-55.
Jacinto, E., Loewith, R., Schmidt, A., Lin, S., Ruegg, M.A., Hall, A., and Hall, M.N.
(2004). Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin
insensitive. Nat Cell Biol 6, 1122-1128.
Jung, C.H., Jun, C.B., Ro, S.H., Kim, Y.M., Otto, N.M., Cao, J., Kundu, M., and Kim,
D.H. (2009). ULK-Atg13-FIP200 complexes mediate mTOR signaling to the
autophagy machinery. Mol Biol Cell 20, 1992-2003.
Kabeya, Y., Kamada, Y., Baba, M., Takikawa, H., Sasaki, M., and Ohsumi, Y. (2005).
Atg17 functions in cooperation with Atg1 and Atg13 in yeast autophagy. Mol Biol
Cell 16, 2544-2553.
Kabeya, Y., Mizushima, N., Ueno, T., Yamamoto, A., Kirisako, T., Noda, T.,
Kominami, E., Ohsumi, Y., and Yoshimori, T. (2000). LC3, a mammalian homologue
of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J
19, 5720-5728.
Kabeya, Y., Mizushima, N., Yamamoto, A., Oshitani-Okamoto, S., Ohsumi, Y., and
Yoshimori, T. (2004). LC3, GABARAP and GATE16 localize to autophagosomal
membrane depending on form-II formation. J Cell Sci 117, 2805-2812.
Kamada, Y., Funakoshi, T., Shintani, T., Nagano, K., Ohsumi, M., and Ohsumi, Y.
(2000). Tor-mediated induction of autophagy via an Apg1 protein kinase complex. J
Cell Biol 150, 1507-1513.
Karantza-Wadsworth, V., Patel, S., Kravchuk, O., Chen, G., Mathew, R., Jin, S., and
White, E. (2007). Autophagy mitigates metabolic stress and genome damage in
mammary tumorigenesis. Genes Dev 21, 1621-1635.
Kawakami, K., Shima, A., and Kawakami, N. (2000). Identification of a functional
transposase of the Tol2 element, an Ac-like element from the Japanese medaka fish,
and its transposition in the zebrafish germ lineage. Proc Natl Acad Sci U S A 97,
11403-11408.
Kawakami, K., Takeda, H., Kawakami, N., Kobayashi, M., Matsuda, N., and Mishina,
M. (2004). A transposon-mediated gene trap approach identifies developmentally
regulated genes in zebrafish. Dev Cell 7, 133-144.
Kimmel, C.B., Ballard, W.W., Kimmel, S.R., Ullmann, B., and Schilling, T.F. (1995).
Stages of embryonic development of the zebrafish. Dev Dyn 203, 253-310.
148
References
Kimura, S., Noda, T., and Yoshimori, T. (2007). Dissection of the autophagosome
maturation process by a novel reporter protein, tandem fluorescent-tagged LC3.
Autophagy 3, 452-460.
Kirisako, T., Baba, M., Ishihara, N., Miyazawa, K., Ohsumi, M., Yoshimori, T., Noda,
T., and Ohsumi, Y. (1999). Formation process of autophagosome is traced with
Apg8/Aut7p in yeast. J Cell Biol 147, 435-446.
Kirisako, T., Ichimura, Y., Okada, H., Kabeya, Y., Mizushima, N., Yoshimori, T.,
Ohsumi, M., Takao, T., Noda, T., and Ohsumi, Y. (2000). The reversible modification
regulates the membrane-binding state of Apg8/Aut7 essential for autophagy and the
cytoplasm to vacuole targeting pathway. J Cell Biol 151, 263-276.
Klionsky, D.J. (2005). The molecular machinery of autophagy: unanswered questions.
J Cell Sci 118, 7-18.
Klionsky, D.J. (2007). Autophagy: from phenomenology to molecular understanding
in less than a decade. Nat Rev Mol Cell Biol 8, 931-937.
Klionsky, D.J., Abeliovich, H., Agostinis, P., Agrawal, D.K., Aliev, G., Askew, D.S.,
Baba, M., Baehrecke, E.H., Bahr, B.A., Ballabio, A., et al. (2008). Guidelines for the
use and interpretation of assays for monitoring autophagy in higher eukaryotes.
Autophagy 4, 151-175.
Komatsu, M., and Ichimura, Y. (2010). Physiological significance of selective
degradation of p62 by autophagy. FEBS Lett 584, 1374-1378.
Kourtis, N., and Tavernarakis, N. (2009). Autophagy and cell death in model
organisms. Cell Death Differ 16, 21-30.
Kuma, A., Hatano, M., Matsui, M., Yamamoto, A., Nakaya, H., Yoshimori, T.,
Ohsumi, Y., Tokuhisa, T., and Mizushima, N. (2004). The role of autophagy during
the early neonatal starvation period. Nature 432, 1032-1036.
Kuma, A., Mizushima, N., Ishihara, N., and Ohsumi, Y. (2002). Formation of the
approximately 350-kDa Apg12-Apg5.Apg16 multimeric complex, mediated by
Apg16 oligomerization, is essential for autophagy in yeast. J Biol Chem 277, 1861918625.
Lam, S.H., and Gong, Z. (2006). Modeling liver cancer using zebrafish: a
comparative oncogenomics approach. Cell Cycle 5, 573-577.
Lam, S.H., Wu, Y.L., Vega, V.B., Miller, L.D., Spitsbergen, J., Tong, Y., Zhan, H.,
Govindarajan, K.R., Lee, S., Mathavan, S., et al. (2006). Conservation of gene
expression signatures between zebrafish and human liver tumors and tumor
progression. Nat Biotechnol 24, 73-75.
Langenau, D.M., Feng, H., Berghmans, S., Kanki, J.P., Kutok, J.L., and Look, A.T.
(2005a). Cre/lox-regulated transgenic zebrafish model with conditional myc-induced
T cell acute lymphoblastic leukemia. Proc Natl Acad Sci U S A 102, 6068-6073.
149
References
Langenau, D.M., Jette, C., Berghmans, S., Palomero, T., Kanki, J.P., Kutok, J.L., and
Look, A.T. (2005b). Suppression of apoptosis by bcl-2 overexpression in lymphoid
cells of transgenic zebrafish. Blood 105, 3278-3285.
Langenau, D.M., Keefe, M.D., Storer, N.Y., Guyon, J.R., Kutok, J.L., Le, X.,
Goessling, W., Neuberg, D.S., Kunkel, L.M., and Zon, L.I. (2007). Effects of RAS on
the genesis of embryonal rhabdomyosarcoma. Genes Dev 21, 1382-1395.
Langenau, D.M., Traver, D., Ferrando, A.A., Kutok, J.L., Aster, J.C., Kanki, J.P., Lin,
S., Prochownik, E., Trede, N.S., Zon, L.I., et al. (2003). Myc-induced T cell leukemia
in transgenic zebrafish. Science 299, 887-890.
Law, J.M. (2001). Mechanistic considerations in small fish carcinogenicity testing.
ILAR J 42, 274-284.
Legakis, J.E., Yen, W.L., and Klionsky, D.J. (2007). A cycling protein complex
required for selective autophagy. Autophagy 3, 422-432.
Levine, B., and Kroemer, G. (2008). Autophagy in the pathogenesis of disease. Cell
132, 27-42.
Levine, B., and Yuan, J. (2005). Autophagy in cell death: an innocent convict? J Clin
Invest 115, 2679-2688.
Li, Z., Huang, X., Zhan, H., Zeng, Z., Li, C., Spitsbergen, J.M., Meierjohann, S.,
Schartl, M., and Gong, Z. (2011). Inducible and repressable oncogene-addicted
hepatocellular carcinoma in Tet-on xmrk transgenic zebrafish. J Hepatol.
Liang, X.H., Jackson, S., Seaman, M., Brown, K., Kempkes, B., Hibshoosh, H., and
Levine, B. (1999). Induction of autophagy and inhibition of tumorigenesis by beclin 1.
Nature 402, 672-676.
Lockshin, R.A., and Williams, C.M. (1965a). Programmed Cell Death--I. Cytology of
Degeneration in the Intersegmental Muscles of the Pernyi Silkmoth. J Insect Physiol
11, 123-133.
Lockshin, R.A., and Williams, C.M. (1965b). Programmed cell death. IV. The
influence of drugs on the breakdown of the intersegmental muscles of silkmoths. J
Insect Physiol 11, 803-809.
Lockshin, R.A., and Williams, C.M. (1965c). Programmed cell death. V. Cytolytic
enzymes in relation to the breakdown of the intersegmental muscles of silkmoths. J
Insect Physiol 11, 831-844.
Loewith, R., Jacinto, E., Wullschleger, S., Lorberg, A., Crespo, J.L., Bonenfant, D.,
Oppliger, W., Jenoe, P., and Hall, M.N. (2002). Two TOR complexes, only one of
which is rapamycin sensitive, have distinct roles in cell growth control. Mol Cell 10,
457-468.
150
References
Long, Q., Meng, A., Wang, H., Jessen, J.R., Farrell, M.J., and Lin, S. (1997). GATA1 expression pattern can be recapitulated in living transgenic zebrafish using GFP
reporter gene. Development 124, 4105-4111.
Longo, L., Platini, F., Scardino, A., Alabiso, O., Vasapollo, G., and Tessitore, L.
(2008). Autophagy inhibition enhances anthocyanin-induced apoptosis in
hepatocellular carcinoma. Mol Cancer Ther 7, 2476-2485.
Lum, J.J., Bauer, D.E., Kong, M., Harris, M.H., Li, C., Lindsten, T., and Thompson,
C.B. (2005). Growth factor regulation of autophagy and cell survival in the absence of
apoptosis. Cell 120, 237-248.
Maiuri, M.C., Le Toumelin, G., Criollo, A., Rain, J.C., Gautier, F., Juin, P., Tasdemir,
E., Pierron, G., Troulinaki, K., Tavernarakis, N., et al. (2007). Functional and
physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J
26, 2527-2539.
Maiuri, M.C., Tasdemir, E., Criollo, A., Morselli, E., Vicencio, J.M., Carnuccio, R.,
and Kroemer, G. (2009). Control of autophagy by oncogenes and tumor suppressor
genes. Cell Death Differ 16, 87-93.
Marino, G., Salvador-Montoliu, N., Fueyo, A., Knecht, E., Mizushima, N., and
Lopez-Otin, C. (2007). Tissue-specific autophagy alterations and increased
tumorigenesis in mice deficient in Atg4C/autophagin-3. J Biol Chem 282, 1857318583.
Mathew, R., Karantza-Wadsworth, V., and White, E. (2007a). Role of autophagy in
cancer. Nat Rev Cancer 7, 961-967.
Mathew, R., Karp, C.M., Beaudoin, B., Vuong, N., Chen, G., Chen, H.Y., Bray, K.,
Reddy, A., Bhanot, G., Gelinas, C., et al. (2009). Autophagy suppresses
tumorigenesis through elimination of p62. Cell 137, 1062-1075.
Mathew, R., Kongara, S., Beaudoin, B., Karp, C.M., Bray, K., Degenhardt, K., Chen,
G., Jin, S., and White, E. (2007b). Autophagy suppresses tumor progression by
limiting chromosomal instability. Genes Dev 21, 1367-1381.
Matsuura, A., Tsukada, M., Wada, Y., and Ohsumi, Y. (1997). Apg1p, a novel protein
kinase required for the autophagic process in Saccharomyces cerevisiae. Gene 192,
245-250.
McCallum, C.M., Comai, L., Greene, E.A., and Henikoff, S. (2000). Targeting
induced local lesions IN genomes (TILLING) for plant functional genomics. Plant
Physiol 123, 439-442.
McClintock, B. (1950). The origin and behavior of mutable loci in maize. Proc Natl
Acad Sci U S A 36, 344-355.
Meijer, A.J., and Codogno, P. (2011). Autophagy: regulation by energy sensing. Curr
Biol 21, R227-229.
151
References
Mizushima, N., Kuma, A., Kobayashi, Y., Yamamoto, A., Matsubae, M., Takao, T.,
Natsume, T., Ohsumi, Y., and Yoshimori, T. (2003). Mouse Apg16L, a novel WDrepeat protein, targets to the autophagic isolation membrane with the Apg12-Apg5
conjugate. J Cell Sci 116, 1679-1688.
Mizushima, N., and Levine, B. (2010). Autophagy in mammalian development and
differentiation. Nat Cell Biol 12, 823-830.
Mizushima, N., Levine, B., Cuervo, A.M., and Klionsky, D.J. (2008). Autophagy
fights disease through cellular self-digestion. Nature 451, 1069-1075.
Mizushima, N., Noda, T., Yoshimori, T., Tanaka, Y., Ishii, T., George, M.D.,
Klionsky, D.J., Ohsumi, M., and Ohsumi, Y. (1998a). A protein conjugation system
essential for autophagy. Nature 395, 395-398.
Mizushima, N., Sugita, H., Yoshimori, T., and Ohsumi, Y. (1998b). A new protein
conjugation system in human. The counterpart of the yeast Apg12p conjugation
system essential for autophagy. J Biol Chem 273, 33889-33892.
Mizushima, N., Yamamoto, A., Hatano, M., Kobayashi, Y., Kabeya, Y., Suzuki, K.,
Tokuhisa, T., Ohsumi, Y., and Yoshimori, T. (2001). Dissection of autophagosome
formation using Apg5-deficient mouse embryonic stem cells. J Cell Biol 152, 657668.
Mizushima, N., Yamamoto, A., Matsui, M., Yoshimori, T., and Ohsumi, Y. (2004). In
vivo analysis of autophagy in response to nutrient starvation using transgenic mice
expressing a fluorescent autophagosome marker. Mol Biol Cell 15, 1101-1111.
Mizushima, N., Yoshimori, T., and Levine, B. (2010). Methods in mammalian
autophagy research. Cell 140, 313-326.
Mizushima, N., Yoshimori, T., and Ohsumi, Y. (2002). Mouse Apg10 as an Apg12conjugating enzyme: analysis by the conjugation-mediated yeast two-hybrid method.
FEBS Lett 532, 450-454.
Nakatogawa, H., Ichimura, Y., and Ohsumi, Y. (2007). Atg8, a ubiquitin-like protein
required for autophagosome formation, mediates membrane tethering and hemifusion.
Cell 130, 165-178.
Nguyen, A.T., Emelyanov, A., Koh, C.H., Spitsbergen, J.M., Lam, S.H., Mathavan, S.,
Parinov, S., and Gong, Z. (2011). A high level of liver-specific expression of
oncogenic KrasV12 drives robust liver tumorigenesis in transgenic zebrafish. Dis
Model Mech.
Noda, T., Kim, J., Huang, W.P., Baba, M., Tokunaga, C., Ohsumi, Y., and Klionsky,
D.J. (2000). Apg9p/Cvt7p is an integral membrane protein required for transport
vesicle formation in the Cvt and autophagy pathways. J Cell Biol 148, 465-480.
Notte, A., Leclere, L., and Michiels, C. (2011). Autophagy as a mediator of
chemotherapy-induced cell death in cancer. Biochem Pharmacol 82, 427-434.
152
References
Obara, K., Sekito, T., and Ohsumi, Y. (2006). Assortment of phosphatidylinositol 3kinase complexes--Atg14p directs association of complex I to the preautophagosomal structure in Saccharomyces cerevisiae. Mol Biol Cell 17, 1527-1539.
Panaretou, C., Domin, J., Cockcroft, S., and Waterfield, M.D. (1997).
Characterization of p150, an adaptor protein for the human phosphatidylinositol
(PtdIns) 3-kinase. Substrate presentation by phosphatidylinositol transfer protein to
the p150.Ptdins 3-kinase complex. J Biol Chem 272, 2477-2485.
Parinov, S., Kondrichin, I., Korzh, V., and Emelyanov, A. (2004). Tol2 transposonmediated enhancer trap to identify developmentally regulated zebrafish genes in vivo.
Dev Dyn 231, 449-459.
Patton, E.E., Widlund, H.R., Kutok, J.L., Kopani, K.R., Amatruda, J.F., Murphey,
R.D., Berghmans, S., Mayhall, E.A., Traver, D., Fletcher, C.D., et al. (2005). BRAF
mutations are sufficient to promote nevi formation and cooperate with p53 in the
genesis of melanoma. Curr Biol 15, 249-254.
Pelengaris, S., Khan, M., and Evan, G. (2002). c-MYC: more than just a matter of life
and death. Nat Rev Cancer 2, 764-776.
Perlmutter, D.H. (2009). Autophagic disposal of the aggregation-prone protein that
causes liver inflammation and carcinogenesis in alpha-1-antitrypsin deficiency. Cell
Death Differ 16, 39-45.
Petiot, A., Ogier-Denis, E., Blommaart, E.F., Meijer, A.J., and Codogno, P. (2000).
Distinct classes of phosphatidylinositol 3'-kinases are involved in signaling pathways
that control macroautophagy in HT-29 cells. J Biol Chem 275, 992-998.
Postlethwait, J.H., Yan, Y.L., Gates, M.A., Horne, S., Amores, A., Brownlie, A.,
Donovan, A., Egan, E.S., Force, A., Gong, Z., et al. (1998). Vertebrate genome
evolution and the zebrafish gene map. Nat Genet 18, 345-349.
Qu, X., Yu, J., Bhagat, G., Furuya, N., Hibshoosh, H., Troxel, A., Rosen, J.,
Eskelinen, E.L., Mizushima, N., Ohsumi, Y., et al. (2003). Promotion of
tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin
Invest 112, 1809-1820.
Reggiori, F., Tucker, K.A., Stromhaug, P.E., and Klionsky, D.J. (2004). The Atg1Atg13 complex regulates Atg9 and Atg23 retrieval transport from the preautophagosomal structure. Dev Cell 6, 79-90.
Rubinsztein, D.C., Gestwicki, J.E., Murphy, L.O., and Klionsky, D.J. (2007).
Potential therapeutic applications of autophagy. Nat Rev Drug Discov 6, 304-312.
Saunders, J.W., Jr. (1966). Death in embryonic systems. Science 154, 604-612.
Scarlatti, F., Granata, R., Meijer, A.J., and Codogno, P. (2009). Does autophagy have
a license to kill mammalian cells? Cell Death Differ 16, 12-20.
153
References
Schmelzle, T., and Hall, M.N. (2000). TOR, a central controller of cell growth. Cell
103, 253-262.
Schuler, W., Sedrani, R., Cottens, S., Haberlin, B., Schulz, M., Schuurman, H.J.,
Zenke, G., Zerwes, H.G., and Schreier, M.H. (1997). SDZ RAD, a new rapamycin
derivative: pharmacological properties in vitro and in vivo. Transplantation 64, 36-42.
Seglen, P.O., and Gordon, P.B. (1982). 3-Methyladenine: specific inhibitor of
autophagic/lysosomal protein degradation in isolated rat hepatocytes. Proc Natl Acad
Sci U S A 79, 1889-1892.
Shen, H.M., and Codogno, P. (2011). Autophagic cell death: Loch Ness monster or
endangered species? Autophagy 7, 457-465.
Shepard, J.L., Amatruda, J.F., Finkelstein, D., Ziai, J., Finley, K.R., Stern, H.M.,
Chiang, K., Hersey, C., Barut, B., Freeman, J.L., et al. (2007). A mutation in separase
causes genome instability and increased susceptibility to epithelial cancer. Genes Dev
21, 55-59.
Shepard, J.L., Amatruda, J.F., Stern, H.M., Subramanian, A., Finkelstein, D., Ziai, J.,
Finley, K.R., Pfaff, K.L., Hersey, C., Zhou, Y., et al. (2005). A zebrafish bmyb
mutation causes genome instability and increased cancer susceptibility. Proc Natl
Acad Sci U S A 102, 13194-13199.
Spencer, C.A., and Groudine, M. (1991). Control of c-myc regulation in normal and
neoplastic cells. Adv Cancer Res 56, 1-48.
Spitsbergen, J.M., Tsai, H.W., Reddy, A., Miller, T., Arbogast, D., Hendricks, J.D.,
and Bailey, G.S. (2000a). Neoplasia in zebrafish (Danio rerio) treated with 7,12dimethylbenz[a]anthracene by two exposure routes at different developmental stages.
Toxicol Pathol 28, 705-715.
Spitsbergen, J.M., Tsai, H.W., Reddy, A., Miller, T., Arbogast, D., Hendricks, J.D.,
and Bailey, G.S. (2000b). Neoplasia in zebrafish (Danio rerio) treated with N-methylN'-nitro-N-nitrosoguanidine by three exposure routes at different developmental
stages. Toxicol Pathol 28, 716-725.
Stack, J.H., DeWald, D.B., Takegawa, K., and Emr, S.D. (1995). Vesicle-mediated
protein transport: regulatory interactions between the Vps15 protein kinase and the
Vps34 PtdIns 3-kinase essential for protein sorting to the vacuole in yeast. J Cell Biol
129, 321-334.
Stanton, M.F. (1965). Diethylnitrosamine-Induced Hepatic Degeneration and
Neoplasia in the Aquarium Fish, Brachydanio Rerio. J Natl Cancer Inst 34, 117-130.
Stern, H.M., and Zon, L.I. (2003). Cancer genetics and drug discovery in the zebrafish.
Nat Rev Cancer 3, 533-539.
Stoletov, K., and Klemke, R. (2008). Catch of the day: zebrafish as a human cancer
model. Oncogene 27, 4509-4520.
154
References
Stoletov, K., Montel, V., Lester, R.D., Gonias, S.L., and Klemke, R. (2007). Highresolution imaging of the dynamic tumor cell vascular interface in transparent
zebrafish. Proc Natl Acad Sci U S A 104, 17406-17411.
Takamura, A., Komatsu, M., Hara, T., Sakamoto, A., Kishi, C., Waguri, S., Eishi, Y.,
Hino, O., Tanaka, K., and Mizushima, N. (2011). Autophagy-deficient mice develop
multiple liver tumors. Genes Dev 25, 795-800.
Tanida, I., Sou, Y.S., Minematsu-Ikeguchi, N., Ueno, T., and Kominami, E. (2006).
Atg8L/Apg8L is the fourth mammalian modifier of mammalian Atg8 conjugation
mediated by human Atg4B, Atg7 and Atg3. FEBS J 273, 2553-2562.
Teter, S.A., Eggerton, K.P., Scott, S.V., Kim, J., Fischer, A.M., and Klionsky, D.J.
(2001). Degradation of lipid vesicles in the yeast vacuole requires function of Cvt17,
a putative lipase. J Biol Chem 276, 2083-2087.
Tsukamoto, S., Kuma, A., Murakami, M., Kishi, C., Yamamoto, A., and Mizushima,
N. (2008). Autophagy is essential for preimplantation development of mouse embryos.
Science 321, 117-120.
Turcotte, S., Chan, D.A., Sutphin, P.D., Hay, M.P., Denny, W.A., and Giaccia, A.J.
(2008). A molecule targeting VHL-deficient renal cell carcinoma that induces
autophagy. Cancer Cell 14, 90-102.
Urlinger, S., Baron, U., Thellmann, M., Hasan, M.T., Bujard, H., and Hillen, W.
(2000). Exploring the sequence space for tetracycline-dependent transcriptional
activators: novel mutations yield expanded range and sensitivity. Proc Natl Acad Sci
U S A 97, 7963-7968.
Vennstrom, B., Sheiness, D., Zabielski, J., and Bishop, J.M. (1982). Isolation and
characterization of c-myc, a cellular homolog of the oncogene (v-myc) of avian
myelocytomatosis virus strain 29. J Virol 42, 773-779.
Volinia, S., Dhand, R., Vanhaesebroeck, B., MacDougall, L.K., Stein, R., Zvelebil,
M.J., Domin, J., Panaretou, C., and Waterfield, M.D. (1995). A human
phosphatidylinositol 3-kinase complex related to the yeast Vps34p-Vps15p protein
sorting system. EMBO J 14, 3339-3348.
Wang, C., Tao, W., Wang, Y., Bikow, J., Lu, B., Keating, A., Verma, S., Parker, T.G.,
Han, R., and Wen, X.Y. (2010). Rosuvastatin, identified from a zebrafish chemical
genetic screen for antiangiogenic compounds, suppresses the growth of prostate
cancer. Eur Urol 58, 418-426.
Wang, C.W., and Klionsky, D.J. (2003). The molecular mechanism of autophagy.
Mol Med 9, 65-76.
Westerfield, M. (2000). The Zebrafish Book. A Guide for the Laboratory Use of
Zebrafish (Danio rerio). 4th edn (University of Oregon Press, Eugene).
155
References
White, E., and DiPaola, R.S. (2009). The double-edged sword of autophagy
modulation in cancer. Clin Cancer Res 15, 5308-5316.
Winnemoeller, D., Wellbrock, C., and Schartl, M. (2005). Activating mutations in the
extracellular domain of the melanoma inducing receptor Xmrk are tumorigenic in
vivo. Int J Cancer 117, 723-729.
Wittbrodt, J., Adam, D., Malitschek, B., Maueler, W., Raulf, F., Telling, A.,
Robertson, S.M., and Schartl, M. (1989). Novel putative receptor tyrosine kinase
encoded by the melanoma-inducing Tu locus in Xiphophorus. Nature 341, 415-421.
Wittbrodt, J., Lammers, R., Malitschek, B., Ullrich, A., and Schartl, M. (1992). The
Xmrk receptor tyrosine kinase is activated in Xiphophorus malignant melanoma.
EMBO J 11, 4239-4246.
Wixon, J. (2000). Featured organism: Danio rerio, the zebrafish. Yeast 17, 225-231.
Xie, B.S., Zhao, H.C., Yao, S.K., Zhuo, D.X., Jin, B., Lv, D.C., Wu, C.L., Ma, D.L.,
Gao, C., Shu, X.M., et al. (2011). Autophagy inhibition enhances etoposide-induced
cell death in human hepatoma G2 cells. Int J Mol Med 27, 599-606.
Yan, J., Kuroyanagi, H., Kuroiwa, A., Matsuda, Y., Tokumitsu, H., Tomoda, T.,
Shirasawa, T., and Muramatsu, M. (1998). Identification of mouse ULK1, a novel
protein kinase structurally related to C. elegans UNC-51. Biochem Biophys Res
Commun 246, 222-227.
Yang, Z., Huang, J., Geng, J., Nair, U., and Klionsky, D.J. (2006). Atg22 recycles
amino acids to link the degradative and recycling functions of autophagy. Mol Biol
Cell 17, 5094-5104.
Yang, Z., and Klionsky, D.J. (2009). An overview of the molecular mechanism of
autophagy. Curr Top Microbiol Immunol 335, 1-32.
Yang, Z., and Klionsky, D.J. (2010). Eaten alive: a history of macroautophagy. Nat
Cell Biol 12, 814-822.
Yen, W.L., Legakis, J.E., Nair, U., and Klionsky, D.J. (2007). Atg27 is required for
autophagy-dependent cycling of Atg9. Mol Biol Cell 18, 581-593.
Yin, X.M., Ding, W.X., and Gao, W. (2008). Autophagy in the liver. Hepatology 47,
1773-1785.
Yousefi, S., Perozzo, R., Schmid, I., Ziemiecki, A., Schaffner, T., Scapozza, L.,
Brunner, T., and Simon, H.U. (2006). Calpain-mediated cleavage of Atg5 switches
autophagy to apoptosis. Nat Cell Biol 8, 1124-1132.
Yue, Z., Jin, S., Yang, C., Levine, A.J., and Heintz, N. (2003). Beclin 1, an autophagy
gene essential for early embryonic development, is a haploinsufficient tumor
suppressor. Proc Natl Acad Sci U S A 100, 15077-15082.
156
References
Zhang, J., Sattler, M., Tonon, G., Grabher, C., Lababidi, S., Zimmerhackl, A., Raab,
M.S., Vallet, S., Zhou, Y., Cartron, M.A., et al. (2009). Targeting angiogenesis via a
c-Myc/hypoxia-inducible factor-1alpha-dependent pathway in multiple myeloma.
Cancer Res 69, 5082-5090.
Zhivotovsky, B., and Orrenius, S. (2010). Cell cycle and cell death in disease: past,
present and future. J Intern Med 268, 395-409.
Zhou, F., Yang, Y., and Xing, D. (2011). Bcl-2 and Bcl-xL play important roles in the
crosstalk between autophagy and apoptosis. FEBS J 278, 403-413.
157
[...]... for mutations that are present in a gene-ofinterest Progeny identified to carry the desired mutations are then propagated to maintain the line to study the function of the gene -of- interest Furthermore, fluorescent proteins can be expressed in specific organs in order to study their development over time by placing the gene encoding fluorescent proteins under the control of organ-specific promoters (Higashijima... pathways, extrinsic and intrinsic, with the extrinsic pathway activated via receptor binding of molecules belonging to the Tumour Necrosis Factor (TNF) family, and the intrinsic pathway activated by stimuli converging and acting on the mitochondria Both pathways culminate in the activation of caspases, which serves to degrade cellular components at the final step of apoptosis, following which the degraded... only the expression of the gene, but also the degree of expression by controlling Dox concentration 1.3.2 The use of transposable elements to generate transgenic zebrafish The standard method of generating transgenic zebrafish is via the injection of DNA constructs containing gene expression cassettes consisting of a promoter, a transgene and polyA signals into one-cell stage embryos in the hope that the. .. advances in sequencing technologies, the zebrafish genome sequence is also made available as well for positional cloning In spite of the large evolutionary distance between zebrafish and human, the syntenic relationship between their genomes further allow the use of the zebrafish in the identification of gene functions of unknown genes in both zebrafish and humans (Barbazuk et al., 2000) In all, these... to be active in zebrafish and human cells, eliminating the requirement for host cell-specific factors for transposition to occur Furthermore, our laboratory recently reported the successful use of the Ac/Ds transposable element in the generation of transgenic zebrafish expressing zebrafish kRASV12 under the control of the liver- specific fabp10 promoter in modelling oncogene-driven liver cancer (Her... stimulated during inflammation in the liver following injury and that tumourigenesis is favoured in the backdrop of inflammation Further studies implicated the over-expression of EGFR and its ligands in human HCC (Avila et al., 2006; Berasain et al., 2007; Breuhahn et al., 2006; Castillo et al., 2006) Thus it would be interesting to uncover the role chronic Xmrk over-expression plays in liver carcinogenesis... expression inducing vessel permeabilisation and vessel remodelling to aid the intravasation of cancer cells into the remodelled blood vessels (Stoletov et al., 2007) The technique of generating transgenic zebrafish with the ability to control the expression of various genes in the study of gene function can also be extended to the study of oncogenes and tumour suppressors Through the use of tissue-specific... driven liver cancer models Our laboratory has also successfully made use of the Tet-on system to generate transgenic zebrafish with inducible, liver- specific expression of oncogenes to model liver cancer In our zebrafish, a liver- specific fabp10 promoter was used to drive the constitutive expression of the rtTA transactivator, which will activate transcription of the oncogene found downstream of the minimal... found to harbour activating mutations in the extracellular domain, explaining its tumourigenicity in vitro and in vivo (Winnemoeller et al., 2005) The suitability of using Xmrk to drive liver carcinogenesis was further supported by studies implicating dysregulated EGFR signalling in human hepatocellular carcinoma or HCC (Berasain et al., 2009) It is noteworthy to point out that the EGFR pathway is chronically... attributes of the zebrafish has positioned it to overcome the shortcomings of mouse models as well as made it relevant in developmental biology and genetics studies 3 Introduction 1.2 The use of zebrafish as a model organism in cancer studies Fish has been used to study cancer since the early 1900s (Stern and Zon, 2003) Early studies involving Xiphophorus, or swordtail fish, ascertained the existence of an ... transgenic line Establishing the role of autophagy in liver cancer 4.1 Preliminary data implicating autophagy inhibition in liver cancer 4.2 Establishing double transgenic lines with liver- specific... as a model organism in the study of human diseases including cancers, we aim to investigate the role of autophagy in liver cancer using our established zebrafish liver cancer models As little study.. .ELUCIDATING THE ROLE OF AUTOPHAGY IN ZEBRAFISH MODELS OF LIVER CANCER SIM HUEY FEN, TINA (B.Sc.(Hons.)), NUS A THESIS SUBMITTED FOR THE DEGREE OF MASTER OF SCIENCE DEPARTMENT OF BIOLOGICAL