Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống
1
/ 186 trang
THÔNG TIN TÀI LIỆU
Thông tin cơ bản
Định dạng
Số trang
186
Dung lượng
2,06 MB
Nội dung
CONSTRUCTION OF NEUROTOXIN DATABASE AND SCREENING FOR
POTENTIAL THERAPUETIC AGENTS
BY
JOYCE SIEW PHUI YEE
A THESIS SUBMITTED FOR THE DEGREE OF
MASTER OF SCIENCE
DEPARTMENT OF BIOCHEMISTRY
NATIONAL UNIVERSITY OF SINGAPORE
2004
i
In loving memory
To my father, Mr. Siew Yew Chuan
i
Acknowledgements
I would like to express my sincere thanks to my supervisor, Professor Kandiah Jeyaseelan
for his dedicated supervision, constant encouragement and continued support during the
course of this project. I am also grateful for the assistance of Associate Professor
Vladimir Brusic from Institute of Infocomm Research for his kind help in the
construction of the neurotoxin database.
My sincere appreciation also goes out to Dr Arunmozhiarasi Armugam for her valuable
input and advice throughout the entire course of my studies.
I am grateful to the Department of Biochemistry, National University of Singapore for
the outstanding academic training it had provided me during my 6 years of undergraduate
and graduate studies here.
A big thank you to my labmates, past and present – Ram, Charmian, Dawn, Siaw Ching,
Sam and Yimin for all their wonderful friendship, support and kind assistance in every
possible way.
Finally, words are not enough to express my gratitude to my family, my mentor in life,
Dr. Daisaku Ikeda, and Sun, who have always been supportive and understanding of my
long absence from home and many late nights in the lab. Thank you from the bottom of
my heart!!
i
Table of contents
Acknowledgements
i
Contents
ii
Publications
vi
List of Figures
vii
List of Tables
ix
Abbreviations
x
Summary
xi
Chapter 1: Introduction
Chapter 1: Introduction
1.1
General introduction
1
1.2
Snake venom components
4
1.3
Neurotoxins
5
1.3.1
Neurotransmission
6
1.3.2
Neurotoxins and envenomation
7
1.4
Classification of neurotoxins
9
1.4.1
Presynaptic neurotoxins
9
1.4.2
β-neurotoxins
9
1.4.3
Acetylcholinesterase inhibitors
11
1.4.4
Potassium channel inhibitors
12
1.5
Postsynaptic neurotoxins
13
1.5.1
Nicotinic acetylcholine receptor
13
1.5.2
α-Neurotoxins
16
ii
1.5.3
Short- and long-chain neurotoxins
17
1.5.4
Kappa neurotoxins
18
1.5.5
Weak neurotoxins
18
1.6
Application of snake venom neurotoxins in research and therapy
19
1.6.1
Viral infection
19
1.6.2
Myasthenia gravis
21
1.6.3
Alzheimer’s disease
22
1.6.4
Thrombotic disease
22
1.6.5
Research tools
22
1.6.6
Novel functional homologs
24
1.6.7
Chimeric toxins
25
1.7
Scorpion
25
1.7.1
Buthus martensi Karsch
25
1.7.2
Scorpion venom
26
1.8
Microarray
26
1.8.1
DNA microarray
28
1.8.2
Applications of DNA microarray
29
1.9
Neurogenesis
31
1.10
Database creation
32
1.10.1
Current problem with analysis of snake toxins using bioinformatic resources
35
1.11
Aims of the project
35
iii
Chapter 2: Materials and Methods
Section A
2.1
Bioinformatics
38
2.2
Data collection and cleaning
39
2.3
Data annotation
40
2.4
Data analysis
41
2.5
Phylogenetic study
42
2.6
Bioinformatics tools used
42
2.7
Scorpion venom and venom glands
44
2.8
Purification of scorpion venom
44
2.9
Purification of snake Bcntx-4
44
2.10
Mass spectrometry determination and N-terminal amino acid sequencing
45
2.11
Protein estimation
45
2.12
Cell culture and medium
45
2.13
MTT cell viability assay
45
2.14
Microarray
46
2.15
Total RNA cleanup
46
2.16
cDNA synthesis
47
2.16.1
Primer hybridization
47
2.16.2
Temperature adjustment
47
2.16.3
First strand synthesis
48
Section B
iv
2.17
Second strand cDNA synthesis
48
2.18
Cleanup of double stranded cDNA
49
2.19
Synthesis of biotin-labeled cRNA
50
2.20
Cleaning up and quantifying in vitro transcription (IVT) products.
51
2.21
Fragmentation of cRNA for target preparation
51
2.22
Eukaryotic target hybridization
52
2.23
Probe Array washing, staining and scanning
53
2.24
Microarray data analysis
56
2.25
Isolation of total RNA
57
2.26
RNA gel electrophoresis
57
2.27
Real-time quantitative PCR
58
2.28
Purification of Torpedo californica acetylcholine receptors
59
2.29
Competitive binding assay
60
2.30
Statistical analysis
60
Section C
List of chemicals used
61
Chapter 3: Construction of protein database of snake venom neurotoxins
3.1
Introduction
67
3.2
Data acquisition
67
3.3
Database features
69
3.4
Descriptions of the svNTX individual record
72
3.5
Classification of snake venom neurotoxins
75
3.6
Receptor binding studies to test the in silico prediction tool in the database
91
3.7
Conclusion
93
v
Chapter 4: Therapuetic leads screening in BMK fraction II using toxicogenomic approach
4.1
Introduction
94
4.2
Purification of BMKII using reverse phase-high performance liquid chromatography
95
4.3
MTT test
96
4.4
Total RNA isolation
97
4.5
Microarray analysis
97
4.6
Quantitative Real-time PCR analysis of genes affecting neurogenesis and angiogenesis
107
4.7
Further purification of BmkII using reverse phase-high performance liquid chromatography
110
4.7.1
Effects of BMKII fractions 1 – 9 on gene expression of ten selected genes
4.7.2
Further separation of fractions RP5-7 using RP-HPLC
113
4.7.3
Effects of BmKII fraction RP5, 6 and 7 on six selected genes
116
4.7.4
Purification of the five fractions using RP-HPLC and determination of protein mass
using MALDI-TOF
117
4.7.5
Effects of BmKII subfractions RP5 and 6 on six selected genes
125
4.8
N-terminal sequencing of subfractions 5, 6 and 7
127
4.9
Conclusion
128
Chapter 5: Discussion
5.1
Neurotoxin database
130
vi
5.2
Conclusion and future studies
132
5.3
Screening of scorpion venom for therapeutic peptides
133
5.3.1
Galanin
134
5.3.2
Amphiregulin
136
5.3.2.1
Application
138
5.3.3
GABA (A) receptor a-5 subunit
139
5.3.4
Sensory and motor-neuron derived factor (neuregulin)
140
5.3.5
Fibroblast growth factor 2 (FGF2)
141
5.3.6
Krit1
144
5.4
Conclusions and future studies
145
References
146
vii
Summary
Snake and scorpion venoms contain many toxins that have important application as
therapeutic agents and also as research tools. As a first part of this study, a snake venom
neurotoxin (NTX) database was constructed using bioinformatics. Snake NTXs are a
large family of active peptides with considerable sequence homology, but with different
biological properties. Sequence, functional and structural information on snake venom
neurotoxins (svNTXs) are scattered across multiple sources such as journals, books and
public databases, with very limited functional annotation. Through this study, an on-line
database
of
NTX
proteins
has
been
made
available
at
http://sdmc.12r.a-
star.edu.sg/Templar/DB/snake_neurotoxin. This database can also be found under SwissProt Toxin Annotation Project website http://www.expasy.org/sprot. At present, 272
NTXs sequences are available in the database and each sequence contains fully annotated
function and literature references. An annotation tool was also incorporated to aid
functional prediction of newly identified NTXs as an additional resource for
toxinologists. In addition, a classification system based on structure-function and
phylogeny relationship derived from 272 NTXs has been proposed and discussed in detail
in the thesis.
In Traditional Chinese Medicine, the scorpion venom has been used in the treatment of
neuromuscular diseases such as paralysis, epilepsy, apoplexy, hemiplegic and facial
paralysis. The screening of these peptides have been carried out using proteomics. Hence,
as a second part of this study, a preliminary screening for therapeutic peptides has been
carries out using the gene chip microarray and real-time PCR on the Chinese scorpion
venom. Several interesting peptides have been found to induce changes in expression of
genes that are involved in neurogenesis in the brain, a finding that is previously unknown.
viii
Publications
Journal article
Joyce Siew Phui Yee, Gong Nanling, Fatemah Afifiyan, Ma Donghui, Poh Siew Lay,
Arunmozhiarasi Armugam, Kandiah Jeyaseelan (2004) Snake postsynaptic neurotoxins:
gene structure, phylogeny and applications in research and therapy. Biochimie 86, 137149.
Joyce Siew Phui Yee, Asif M. Khan, Paul TJ Tan, Judice LY Koh, Seah Seng Hong,
Chuay Yeng Koo, Siaw Ching Chai, Arunmozhiarasi Armugam, Vladimir Brusic and
Kandiah Jeyaseelan (2004). Systematic analysis of snake neurotoxins functional
classification using a data warehousing approach. Bioinformatics July 22.
Conference abstract
Siew Phui Yee Joyce; Asif Khan; Tan Thiam Joo Paul; Judice Koh, Arunmozhiarasi
Armugam; Kandiah Jeyaseelan; and Vladimir Brusic. (2003) Systemization of snake
venom neurotoxins based on structure and function. 14-19th September, Adelaide,
Australia.
ix
List of Tables
Table 1.1
Systematic classification of different species of snakes
around the world.
4
Table 1.2
Some of the common components in snake venoms of different species.
5
Table 1.3
Envenomation effects of neurotoxins across three different species.
8
Table 2.1
Primers sequences used for real-time PCR.
58
Table 3.1
Summary of the 272 neurotoxin entries extracted from various sources.
68
Table 3.2
Sequence identity for the four unique toxins that are not found
in public databases but available in published journals.
69
Table 3.3
Number of the complete, incomplete and missing sequences in
the snake venom neurotoxins final data set.
69
Table 3.4
The distinct disulfide pairing pattern of mature svNTXs.
78
Table 3.5
Summary of the individual entries based on GenBank Accession Number
of each member in each group as depicted in Fig 3.6
84
Table 4.1
Summary of the 875 genes function that was altered by treatment
of BMKII in human neuroblastoma cell.
98
Table 4.2
List of the ten selected genes after microarray gene chip analysis
99
Table 4.3
Changes in gene expression level as detected by microarray and real-time P 108
Table 4.4
Relative level of gene expression of the six genes using nine subfractions
from BmKII.
113
Table 4.5
Relative level of gene expression on the six genes using
subfractions of fractions RP5, 6 and 7.
117
Table 4.6
Relative level of gene expression on the six genes using 10 ng/ml
subfractions of fractions RP5 and 6.
125
Table 4.7
Relative level of gene expression on six genes using 50 ng/ml
subfractions of fractions RP5 and 6.
126
Table 4.8
Results of the N-terminal sequencing of five fractions.
127
Table 4.9
Blast search of the five peptides.
128
x
List of Figures
Figure 1.1
Venom apparatus of Naja sputatrix, a Malayan spitting cobra .
3
Figure 1.2
Mechanism of acetylcholine transmission at neuromuscular junction.
7
Figure 1.3
Clinical manesfestations of a snake bite.
8
Figure 1.4
The structure of nicotinic acetylcholine receptor and its binding protein .
15
Figure 1.5
The 3D-structures of three-finger neurotoxins from snake venoms that interact with
nicotinic acetylcholine receptors.
16
Figure 1.6
A model showing homology of rabies glycoprotein with the "toxic" loop of the neurotoxins.
21
Figure 1.7
Comparison of the lynx1 model and the αBgt experimental structure.
24
Figure 1.8
Anatomical structure of Buthus martensi Karsch .
30
Figure 2.1
A schematic representation of CysAlign methodology.
41
Figure 3.1
The user interface of the svNTXs database.
70
Figure 3.2
An output feature of the search (partial) using the keyword “Bungarotoxin”.
71
Figure 3.3
An example of an outlay of record available in the database.
72
Figure 3.4
Summary of the types of errors encountered in the public databases.
74
Figure 3.5
Unique disulfide pairing patterns of all the neurotoxins.
77
Figure 3.6
Classification of snake NTX based on structure, function and phylogenetic information.
81
Figure 3.7
Phylogenetic trees performed using parsimony analysis of all the mature svNTX.
90
Figure 3.8
Competitive binding studies of 125I-α-Bgt and native Bc-ntx4 to nAChR of Torpedo receptor.
91
Figure 3.9
Output of the annotation tool in svNTXs.
92
Figure 4.1
Separation of BmK crude venom using gel filtration.
95
xi
Figure 4.2
Cell survival test of human neuroblastoma cells treated with various concentration of BmkII.
96
Figure 4.3
Total RNA isolation from human neuroblastoma cells treated with 35 µg/ml BmKII.
97
Figure 4.4
Cluster 1 genes identified using Genesis® software.
100
Figure 4.5
Cluster 2 genes identified using Genesis® software.
101
Figure 4.6
Cluster 3 genes identified using Genesis® software.
102
Figure 4.7
Cluster 4 genes identified using Genesis® software.
103
Figure 4.8
Cluster 5 genes identified using Genesis® software.
104
Figure 4.9
Cluster 6 genes identified using Genesis® software.
105
Figure 4.10
Cluster 7 genes identified using Genesis® software.
106
Figure 4.11
Schematic representation of real-time PCR with the SYBR Green I dye.
107
Figure 4.12
Gene expression profile of ten genes studies using microarray and real-time PCR.
109
Figure 4.13
RP-HPLC of BmKII from scorpion venom.
111
Figure 4.14
Real-time PCR analysis using fractions RP1-9.
112
Figure 4.15
RP-HPLC of fraction RP5.
113
Figure 4.16
RP-HPLC of fraction RP6.
114
Figure 4.17
RP-HPLC of fraction RP7.
115
Figure 4.18
Real-time PCR assay of six genes using subfractions of fractions of RP5, 6 and 7 BmKII.
116
Figure 4.19
Fraction RP5.2 separated in Jupiter C4 µBore RP-HPLC column.
118
Figure 4.20
Fraction RP6.2 separated in Jupiter C8 µBore RP-HPLC column.
119
Figure 4.21
Fraction RP6.3 separated in Jupiter C8 µBore RP-HPLC column.
120
xii
Figure 4.22
Fraction RP6.4 separated in Jupiter C4 µBore RP-HPLC column.
121
Figure 4.23
MALDI-TOF of the five fractions.
124
Figure 4.24
Real-time PCR assay of six genes using 10 ng/ml of subfractions RP5 and 6.
125
Figure 4.25
Real-time PCR assay of six genes using 50 ng/ml of subfractions RP5 and 6.
126
Figure 5.1
Galanin receptord and their transduction mechanisms.
136
Figure 5.2
The ERBB signalling network.
138
Figure 5.3
Schematic diagram of FGF-2 signalling pathways which has been shown to activate a
number of intracellular routes.
143
xiii
Abbreviations
α-Bgt
Alpha bungarotoxin
AR
Amphiregulin
BLAST
Basic local alignment search tool
BmK
Buthus martensi Karsch
BSA
Bovine serum albumin
db
database
dNTP
deoxyribonuclease
ESI-MS
electron spray ionization-mass spectrometry
FGF2
fibroblast growth factor
hr
hour
IC50
concentration for 50% inhibition
kD
kilodalton
krit1
Krev interaction trapped 1
mg
milligram
min
minute
mM
millimolar
MW
molecular weight
nAChR
nicotinic acetylcholine receptor
nM
nanomolar
NTX
neurotoxin
PCR
polymerase chain reaction
xiv
PLA2
phospholipase A2
RP-HPLC
reverse phase-high performance liquid chromatography
RT
reverse transcription
SMDF
sensory motor derived neuron
SR
sacroplasmic reticulum
svNTXs
snake venom neurotoxins
µg
microgram
µl
microlitre
xv
Introduction
INTRODUCTION
Introduction
CHAPTER ONE
INTRODUCTION
1.1 General introduction
The animal kingdom contains many venomous species, including the coelenterates,
flatworms, annelids, echinoderms, mollusks, arthropods and chordates. These venomous
animals produce a variety of toxins to defend themselves from predators, to subdue their prey
and for digestion. Therefore, animal toxins have immobilizing effects and killing functions
towards a wide variety of creatures.
Snake venoms have attracted much medical attention since ancient civilizations and had been
used in medical treatment for thousands of years. In the 12th century, doctors used snake
venom to treat leprosy. Snakes belong to the order Squamata and suborder Serpentes
(Ophidia) of the class Reptilia (Kochva, 1987). They appeared in the Lower Cretaceous
Period about 130 million years ago, and have been considered to have evolved from lizards,
Varanidae, probably 30 million years ago (Clifford, 1955). There are about 3200 species of
snakes found worldwide, and 1300 of which are venomous (Hider, 1991). Venomous snakes
belong to the infraorder Caenophidia under Ophidia. They are more advanced and
widespread throughout the world (Phelps, 1989). Venomous snakes usually are defined as
those possessing a pair of venom glands and specialized fangs connected to venom glands by
ducts. The venom apparatus enables them to inflict serious bites in their victims. The venom
apparatus of a snake typically consists of a venom gland, venom duct and one or more fangs
located on each side of the head (Figure 1.1A). Venom is produced in paired modified
salivary glands which in most venomous snakes are located superficially beneath the scales in
the posterior part of the head and eyes. The gland is linked to the fang by a duct. Contraction
of muscles around the gland compresses the gland, forcing the flow of venom along the duct
to the fang where the size of these structures depends on the size and species of the snakes.
1
Introduction
Generally, four families of venomous snakes are known: Elapidae (cobra, mamba);
Hydrophiidae (sea snakes); Viperidae (true vipers and pit vipers and rattlesnakes) and
Colubridae (boomslang and mangrove; Fenton, 2002).
The elapids are a large group of venomous snakes, which are distributed over Africa, Asia,
the Southern parts of North America, Central and South America and Australia (Phelps,
1989).
There are about two hundred and twenty species of elapid snakes which are
represented by sixty-two genera. Generally, the elapids possess fixed front fangs that are
situated in the front of the upper jaw (Figure 1.1B). Most elapids are either terrestrial or
aquatic; only two genera are arboreal, the mambas and the tree cobras, Pseudohaje (Phelps,
1989).
Hydrophids or sea snakes are widely distributed throughout the equatorial and tropical
regions of the Indian and Pacific oceans, from the coast of Africa and America. They are
closely related to the elapids, having a similar venom and fang apparatus. Medically
important genera include Enhydrina, Hydrophis, Pelamis and Laticauda.
Viperids are amongst the best known and probably the most medically important venomous
snakes. They are divided into two subfamilies, the viperin and crotalid. The crotalid
encompasses all the pit vipers and are so called because they possess heat sensitive pits
situated on each side of the head between the nostril and the eye. The heat-sensing pits enable
the animals to detect a temperature change of 1 – 2°C above ambient from a distance of 1 – 2
feet. This facility is used for hunting warm-blooded preys at night. All members of the
viperid family have a set of very well-developed anteriorly-placed proteroglyphous fang
structure. The fangs are on a modified maxilla which is capable of considerable rotation thus
allowing the fangs to be folded against the roof of the mouth when not in use. This has
enabled the development of larger fangs than in other venomous snakes of equivalent size.
The crotalids are typically New World snakes found in America and parts of Southeast Asia,
but are absent from Australia and Europe. Medically-important genera include Crotalus,
Trimeresurus, Agkistrodon, and Sistrurus. The subfamily of crotalids, viperin is found in
Africa, Asia and Europe.
2
Introduction
The colubrid family is the largest single snake family with over 2500 species. It is found
throughout the world on all continents except Antarctica (where no snakes exist). There are
some species within this family which have developed toxic salivary secretions which may
enter a snake bite wound during the envenomation. However, current evidence suggests that
the majority of colubrids do not produce toxic secretions. All colubrids are back-fanged
(Minton, 1990). The position of the fangs limits fang length and makes it less easy for the
snake to bite its victim. Nevertheless, several colubrids have extremely potent venoms and
can cause serious envenomation and death in Man. The best known and most important of
these are some South African colubrids. The distribution of venomous snakes in the world
and their characteristics are summarized in Table 1.1.
Figure 1.1: Venom apparatus of Naja sputatrix, a Malayan spitting cobra. A) Illustration
showing fangs, venom duct and the main venom gland. B) Scanning electron micrograph of
the fang (Gopalakrishnakone, 1990).
3
Introduction
Family
Species
Distribution
Special characterictics
Elapidae
Kraits, Cobra, Mambas, Coral snakes
America, Asia, Africa, and
Australia
Small head, short and fixed fangs
Viperidae
Viper snakes
Europe, Africa, Asia and America
Large, flattened triangular head;
large grooved fangs on the maxillary bone
Colubridae
Tree snakes
In all parts of the world except Australia
Similar to Viperidae, but they possess
heat-snesitive pits on head
Hydrophiidae
Sea snakes
Asia and Australia
Nostrils dorsally on head; flattened tail
Table 1.1: Systematic classification of different species of snakes around the world
(Harris et al. 1991).
1.2 Snake venom components
Snake venoms are a complex mixture of various components with wide-ranging activities.
The composition is variable within a single subspecies or even an individual snake,
depending on age and season. The function of the venom is to subdue the prey. It also serves
as a defensive adaptation and to facilitate in the digestion of they prey (Zeller, 1977). Snake
venoms are usually colorless to dark amber viscous fluids with a viscosity of 1.5 to 2.5 and
specific gravity of 1.03 to 1.12 (Devi, 1968). Most of the snake venoms consist of a complex
mixture of proteins and non-protein components. The non-polypeptide components are the
least biologically active venom fraction. They are mainly amines, carbohydrates, nucleosides,
lipids, small peptides and metal ions (Devi, 1968). The lipids, carbohydrates and nucleosides
are present in low amount (1 - 2% of dry weight).
Enzymatic and non-enzymatic components constitute the polypeptide protein and together
they are made up over 90% of dry weight of snake venom (Hider et al., 1991). Most enzymes
and toxins are very stable. It was found that dried snake venom retains lethality and some
enzymatic activities even after storage for 25 – 50 years (Minton, 1990). The enzymatic
components range between 13 – 150kD and more than twenty enzymes have been detected in
snake venoms. Twelve of them are found in all venoms, although their levels differ markedly
(Table 1.2).
4
Introduction
Many of these enzymes are hydrolases such as exo- and endopeptidases and proteinases that
play a role in digestion. Others include neurotoxic phospholipase A2 (PLA2) that blocks
neuromuscular transmission, and hyaluronidase which facilitates the distribution of other
venom components throughout the prey (Harris, 1991). In addition, proteases that are
involved in blood coagulation can also be found in snake venom (Matsui et al., 2000).
The non-enzymatic polypeptides (5 – 10kD) found in snake venoms include cardiotoxins
(CTX, also called cytotoxins), neurotoxins, proteinase inhibitors, dendrotoxins, myotoxins
and acetylcholinesterase inhibitors (Hider, 1991).
Snakes
All snakes
Enzymes
L- Amino acid oxidase, phosphodiesterase, 5'-nucleotidase, phosphomonoesterase, deoxyribonuclease,
hyaluronidase, NAD-nucleosidase, peptidase and phospholipase A2,ribonuclease, adenosine triphosphatase,
Crotalids and viperids Endopeptidase, arginine ester hydrolase, kininogenase, thrombin-like enzyme, factor X- activator, prothrombin activator
Elapids
Acetylcholinesterase, phospholipase B amnd glycerophosphatase
Some snakes
Glutamic-pyruvic transaminase, catalase, amylase and B-glucosaminidase
Table 1.2: Some of the common components in snake venoms of different species.
1.3 Neurotoxins
The first amino acid sequence of a neurotoxin, Tx α from Naja nigricollis was reported in 1967
(Eaker and Porath, 1967). Since then, a large number of neurotoxins have been isolated and
characterized. Reports on the amino acid sequences of a number of homologous neurotoxins
from snakes such as cobras, kraits, mambas and sea snakes have been added to this growing
list of neurotoxins. At present, more than 100 neurotoxin amino acid sequences are known and
they form one of the largest families of protein with known primary structures (Endo and
Tamiya, 1991).
5
Introduction
Neurotoxins are capable of blocking nerve transmission by binding specifically to nicotinic
acetylcholine receptor (nAChR) located on the postsynaptic membranes of skeletal muscles
and neurons resulting in prevention of neuromuscular transmission leading to death by
asphyxiation (Tu, 1973).
1.3.1 Neurotransmission
Neurotransmission takes place between neuron and neuron as well as between nerve and
muscle cells. When a normal nerve impulse (depolarization wave) passes through the axon and
reaches the end of that axon, the calcium ion concentration in axon is increased and the
neurotransmitter, acetylcholine, is released from the vesicle at the end of the nerve. This
neurotransmitter moves across the synaptic crevice and reaches its receptor (acetylcholine
receptor, AChR) in the next neuron (as in nerve-nerve synapse) or in muscle (as in nervemuscle synapse; Figure 1.2). The neuronal AChR is composed of five subunits, α2β3. When
two molecules of acetylcholine attach to the two α-subunits, the AChR changes its
configuration and becomes an open ion channel, permitting certain ions to pass through so that
the cell’s plasma membrane gets depolarized relatively rapidly, yet transiently, from its resting
value (-50 mV to –100 mV) to a potential more positive than 0 mV. The depolarization wave
reaches the next neuron and can be transmitted to muscle similarly. The depolarization wave
transmitted to muscle is further propagated through the muscle plasma membranes, T-tubules
and sarcoplasmic reticulum (SR). The SR has a very high concentration of calcium ions. When
the depolarization wave reaches the SR, the calcium ions begin to leak out of SR and into the
myoplasm, causing the myofilaments to contract. As soon as the muscle is relaxed, the calcium
ions move back into the SR.
6
Introduction
Figure 1.2: Mechanism of acetylcholine transmission at neuromuscular junction.
Acetylcholine is formed by choline and acetylcholine-CoA by choline acetyltransferase in the
cytosol and stored in synaptic vesicle. The arrival of action potential at the terminus of
presynaptic membrane causes release of acetylcholine which leads to resulting influx of Na+
into the postsynaptic membrane. After release from synaptic cleft, acetylcholine will be
degraded by acetylcholinesterase into choline and acetate. Image obtained from Changeux
(1993).
1.3.2 Neurotoxins and envenomation
Snake envenomation is a major clinical problem worldwide, particularly in parts of the Asian
region where the annual mortality is estimated to be 100, 000 (Ingels, 2001). Neurotoxins have
been found most frequently in venoms of snakes of the family Elapidae and their closest
relatives, Hydrophiidae. Only a few neurotoxins have been identified in venoms of vipers
(Viperidae). The clinical manifestations of snakebite depend on two important factors: the
intrinsic toxicity of the venom on man and the amount injected. A general observation of
neurotoxic snake envenomation is the development of cranial nerve palsies which is
characterized by ptosis, blurred vision, difficulty in swallowing, slurred speech, weakness of
facial muscle and occasionally loss of the sense of taste and smell (Figure 1.3). This syndrome
is often accompanied by drowsiness, sometimes with mental confusion and euphoria. The
onset of this observation may be as soon as three minutes after a bite or as late as 24 hours.
7
Introduction
Both groups of pre- and postsynaptic apparently involved in most of the clinical
manifestations. Evidence suggests that presynaptic neurotoxins are associated with delayed
onset, prolonged symptoms and poor response to antivenoms, whereas the postsynaptic toxins
account for the early paralytic symptoms such as ptosis (Minton, 1990). A brief summary of
neurotoxin envenoming in Elapidae, Hydrophiidae and Viperinae families of snake is
presented in Table 1.3.
Elapidae
Hydrophiidae
Viperinae
√
√
Renal failure
√
√
√
√
√
√
Hypovolemic shock
x
x
√
√
√
√
√
√
√
Cranial nerve palsies
Local pain and swelling
Local necrosis
Respiratory paralysis
Rhabdomyonecrosis
x
√
√
√
Table 1.3: Envenomation effects of neurotoxins across three different species. Data
adapted from Minton, (1990). √ and x indicate the presence and absence of the observed
clinical conditions in human respectively.
B
A
Figure 1.3: Clinical manisfestations of a snake bite. A. Ptosis and moderate facial weakness
following the bite of an Australian tiger snake. B. Muscular weakness with the inability to
raise the head seen following a cobra bite in Malaysia. This is typical of a “broken-neck”
syndrome reported after bites of snake with neurotoxic venoms (Minton, 1990).
8
Introduction
1.4 Classification of neurotoxins
Snake neurotoxins can be divided into two groups: presynaptic or postsynaptic neurotoxins
depending on their mode of action. Presynaptic neurotoxins are either phospholipase A2
enzymes or contain these enzymes as an integral part of the neurotoxin complex and,
essentially, mediate their neurotoxicity by inhibiting the release of acetylcholine (Yang, 1994).
The postsynaptic neurotoxins bind to postsynaptic nAChRs at the skeletal muscle
neuromuscular junction to produce blockade of neuromuscular transmission (Lee, 1972). These
neurotoxins are also referred to as curaremimetic or α-neurotoxins.
1.4.1 Presynaptic neurotoxins
A toxin is considered presynaptically active if it can affect transmitter synthesis, storage,
release or turnover, no matter whether the toxin concerned is active in other regards or not
(Harris, 1991). Two kinds of snake venom presynaptic toxins have been recognized so far: 1)
β-neurotoxins characterized by a phospholipase A2 (PLA2) activity; 2) neurotoxins devoid of
enzymatic activity of PLA2, acting either by blocking a voltage-sensitive K+-channel, like
dendrotoxin (Hawgood and Bon, 1991) or by inhibiting the activity of acetylcholinesterase,
with similar overall effect to the increase of acetylcholine, like acetylcholinesterase inhibitor
(Dajas et al., 1987; Lin et al., 1987).
1.4.2 β-neurotoxins
β-neurotoxins are defined as presynaptically active PLA2 toxins which have a dominant
inhibitory action on the indirectly elicited twitch in in vitro neuromuscular preparations by
blocking the acetylcholine release (Fletcher and Rosenberg, 1997). The first β-neurotoxins
characterized is β-bungarotoxin from Bungarus multicinctus (Chang and Lee, 1963). It is now
clear that many of the snake venom PLA2 from kraits, elapids, crotalids and virepids are
presynaptically active (Harris, 1991; Hawgood and Bon, 1991).
9
Introduction
β-neurotoxins may be single chain polypeptides as in notexin (Halpert and Eaker, 1975), multisubunit complexes as in crotoxin (A and B chains; Bouchier et al., 1991) and taipoxin (α, β, γ
chains; Fohlman et al., 1977), or covalently linked assemblies of two or more subunits as in βbungarotoxin (Kondo et al., 1978), and textilotoxin consisting of five subunits (A, B, C and
two D) with only two identical D subunits covalently linked (Tyler et al., 1987). The lethal
activity of these proteins largely depends on the complexity of their quarternary structures.
Multimeric β-neurotoxins have been found to be more potent than monomeric toxins. LD50
(median lethal dose, µg/ kg mouse) varies, from 50 in case of crotoxin, to 15 for βbungarotoxin, 2 for taipoxin and 1 for textilotoxin (Lee and Ho, 1982). In all cases, systematic
intoxication resulted in respiratory paralysis due to cessation of neurotransmission.
β-bungarotoxin being the first presynaptic neurotoxins to be discovered, is very well studied.
The toxin is made up of two structurally different subunits linked by a disulfide bridge. Chain
A (120 amino acid, MW 13.5 kDa) is a PLA2 and chain B resembles proteinase inhibitors and
dendrotoxin or toxin I from Dendroaspis venoms. The overall fold in β-bungarotoxin is
strikingly similar to that of the mammalian class I and class II PLA2 enzymes (Kondo et al.,
1982a, b; Kwong et al., 1995). It was postulated that neuronal acceptors of β-neurotoxins
existed to direct β-neurotoxins almost exclusively towards the neuronal presynaptic plasma
membrane. For example, the acceptors for β-bungarotoxin have been found to consist of
subunits of voltage-sensitive K+ channels (Benishin, 1990). Chain B of β-bungarotoxin, which
is structurally similar to dendrotoxin, has been found to be responsible for the interaction
between toxin and its receptor. This result has been further supported by the observation that βbungarotoxin can bind at high-affinity with dendrotoxin receptors. Crotoxin, another multisubunit β-neurotoxin, has been found to dissociate on reaching its target site, with the nonenzymatic subunit chaperoning the PLA2 subunit to high-affinity sites in synaptic membranes
(Bon et al., 1979).
It seems that all β-neurotoxins selectively decrease the fast K+ current flowing out of the nerve
terminal and hence, they interact with a voltage-sensitive K+ channel, as in the case of βbungarotoxin and dendrotoxin. On the other hand, the absence of competition for their highaffinity binding on synaptosomal membranes between the various β-neurotoxins indicates that
10
Introduction
the various β-neurotoxins bind to different acceptor proteins. Hence, one can conclude that
various β-neurotoxins recognize various subtypes of voltage-sensitive K+ channel.
Presynaptic neurotoxins induce blockage of neurotransmitter release in three phases. The initial
transient inhibitory phase is followed by a facilitatory second phase and a late inhibitory phase
which is irreversible. In the first two phases, the enzymatic activity of the toxins apparently
plays no role, but is undoubtly needed in the third phase, in which a substantial decrease in the
number of the synaptic vesicles leads to final disappearance of the amount of neurotransmitter
stock in the nerve endings. A synaptic vesicle cycle is found to consist of 9 steps: docking,
priming,
fusion/exocytosis,
endocytosis,
translocation,
endosome
fusion,
budding,
neurotransmitter uptake and translocation back to the active zone at the plasma membrane in
the synaptic cleft (Sudhof, 1995; Van der Kloot and Kloot, 1994). The disappearance of the
neurotransmitter may be due to the interference of the last five steps of the synaptic vesicle
recycling.
Clues related to the neurotoxicity of β-neurotoxins have been available, even though further
studies are still required. First, the enzymatic activity is known to be required for the
expression of neurotoxicity, as shown in the third phase of β-neurotoxin intoxication cascade.
In addition, residues 59-89 (which encompasses the β-structure) seemed to be important for
neurotoxicity as drawn from studies on structural comparison of notexin from Notechis
scutatus scutatus, with other β-neurotoxins, and non-neurotoxic enzymes (Dufton and Hider,
1983a; Kini and Iwanaga, 1986; Tsai et al., 1987; Kondo et al., 1989). More recently, Hains et
al. (1999) developed a testable two-step proposal of neurotoxic PLA2 activity; involving the
favourable binding to acceptor molecules followed by enzymatic intrusion upon the target
membrane.
1.4.3 Acetylcholinesterase inhibitors
This group of neurotoxins cause continuous excitement of the muscle by binding to
acetylcholinesterase and thus rendering acetylcholine unhydrolysed (Rodriguez-Ithurralde et
al., 1981, 1983). Since they are capable of affecting transmitter turnover, they can be grouped
as presynaptic neurotoxins (Harris, 1991). Acetylcholinesterase inhibitors have so far been
11
Introduction
isolated only from African mambas (Dendroaspis), including fasciculin (Dajas et al., 1987)
and F7 (Lin et al., 1987) from D. augusticeps and toxin C and D from D. polylepis polylepis
(Joubert and Strydom, 1978; Karlsson et al., 1984). Structurally, they are related to cardiotoxin
and α-neurotoxins. They consist of 57-60 amino acid residues in a single polypeptide chain,
cross-linked by four disulfide bonds, showing similar primary structure to postsynaptic
neurotoxins and cardiotoxins. In addition, they show similar 3-D structure as shown by the
crystalline structure of fasciculin 2 (Le Du et al., 1989). However, they act by different mode
from α-neurotoxins and cardiotoxins. Fasciculin 2 has no presynaptic action on transmitter
release or on postsynaptic receptor-blocking action. Its main action is on acetylcholinesterase
(Harvey et al., 1984; Anderson et al., 1985). By inhibiting acetylcholinesterase, fasciculin
increased the amplitude and time course of the endplate potential (Lee et al., 1985) as well as
the amplitude of the miniature endplate potential (Cervenansky et al., 1991).
1.4.4 Potassium channel inhibitors
Dendrotoxins are small proteins that were isolated 20 years ago from mamba snake venoms.
Dendrotoxin is the first snake toxin found to bind voltage-sensitive K+ channel which is known
to play an important role in the repolarization process in nerve transmission. In addition, three
dendrotoxins (α-DaTX, β-DaTX, δ-DaTX) have also been isolated from the venom of D.
augusticeps. Furthermore, toxin I (DTX14) from venom of D. polylepis polylepis and βbungarotoxin from venom of B. multicinctus have also been isolated.
The functional effects of dendrotoxins have been well described in a review by Harvey (2001).
The first noticeable effect of dendrotoxin is to facilitate transmitter release at peripheral
synapses. Subsequently, the facilitatory effects of the dendrotoxins have been explained by
their blockage of some neuronal K+ channels. This toxin has been suggested to block rapidly
activating K+ current that is important for the control of the excitability of motor nerve
terminals. When injected into the central nervous system, dendrotoxin induces epileptiform
activity (Velluti et al., 1987; Coleman et al., 1992) and in high doses, can lead to neuronal
damage (Bagetta et al., 1992; Mourre et al., 1997). In addition, dendrotoxin increases
inhibitory postsynaptic currents in Purkinje regions of the mouse cerebellar slice preparations
12
Introduction
(Harvey, 2001). Dendrotoxin also increases acetylcholine release in rat striatal slices, an effect
attributed to the block of channels containing Kv1.2 subunits (Fischer and Saria, 1999).
1.5 Postsynaptic neurotoxins
Postsynaptic neurotoxins are those capable of blocking nerve transmission by binding
specifically to nicotinic acetylcholine receptors located on the postsynaptic membranes of
skeletal muscles and/ or of neurons. These toxins, which mimic the neuromuscular blocking
effects of plant alkaloid, tubocurarine, but with approximately 15 – 20-fold greater affinity and
poor reversibility of action, are also known as curaremimetic neurotoxins or simply as αneurotoxins, a suffix of historical significance (Chang, 1979; Chang, 1999). Most αneurotoxins are derived from Elapidae or Hydrophiidae snake venom and belong to the threefinger toxin family. It must be emphasized that snake venoms are not the only exclusive source
of α-neurotoxins. The venoms of marine cone snails also represent a rich combinatorial-like
library of evolutionarily selected, neuropharmacologically active peptides called conotoxins
that target a wide variety of receptors and ion channels. α-neurotoxins bear the imprint of a
region of the nAChR that is likely to be in proximity to, and perhaps even overlap, the binding
site for the natural neurotransmitter acetylcholine (Servent and Menez, 2001).
1.5.1 Nicotinic acetylcholine receptor
The discussion of α-neurotoxins will not be completed without a brief introduction to this well
characterized receptor. The nAChR is perhaps one of the best characterized ion-channel to
date, due in part to the discovery of α-bungarotoxin and a rich and accessible source of
receptor from the electric ray and eel. Consequently, the mammalian neuromuscular junction is
also the most studied and best understood synaptic region (Naguib et al., 2002). The nAChRs
are transmembrane allosteric proteins of MW approximately 290 kDa that are involved in fast
ionic responses to acetylcholine (Karlin et al., 2002). They are pentamers formed by the
association of five subunits arranged symmetrically around the ionic pore in a plane
perpendicular to the membrane (Miyazawa et al., 2003). Each subunit is composed of a large
amino-terminal domain that contributes to the formation of the ligand binding pocket; four
13
Introduction
membrane-spanning domains (MI, MII, MIII, MIV); a large and variable cytoplasmic loop
between MIII and MIV; and a small extracellular carboxyl terminal.
The MII domains of all five subunits contribute to the formation of the cation channel pore
(Figures 1.4 A & B). In vertebrates, the combinatorial assembly of various nicotinic receptor
subunits (α1- α10, β1- β4, δ, γ, and ε) generates a wide diversity of receptors, with various
electrical and binding properties. Generally, nicotinic receptors can be divided into two main
families: the muscle and neuronal nAChRs (Corringer et al., 2000). The well-characterized
muscle receptor consists of a combination of α1, β1, δ and γ or ε subunits in the stoichiometry
of (α1)2 β1 γ δ or (α1)2 β1 ε δ in the embryonic or adult receptor, respectively. These are
densely distributed on the postsynaptic membrane of the neuromuscular junction and mediate
intercellular communication between the nerve ending and skeletal muscle. The muscle-type
receptor (α1)2 β1 γ δ is also found in abundance in the electric organ of the Torpedo ray.
Neuronal nicotinic receptors are composed of α2 – α10 and β2 – β4 subunits.
An excellent insights into the structure of nAChRs and ligand-gated ion channels in general,
was made possible by the discovery and characterization of an acetylcholine-binding protein
(AChBP) from the snail Lymnaea stagnalis (Figures 1.4C & D; Smit et al., 2001; Brejc et al.,
2001) which is a remarkable homologue of the amino-terminal extracellular domain of nAChR.
The crystal structure of the AChBP revealed that each ligand-binding site is located in a cleft at
the subunit interface, conforming to the existing biochemical and mutational data on nAChRs
(Brejc et al., 2001). Based on the structure of AChBP, nearly all the residues of the agonistbinding site of nAChR that were previously identified by photoaffinity labeling and
mutagenesis experiments are located in the small cavity of about 10 – 12 Å diameter that is
primarily formed by aromatic residues contributed by participating subunits (Grutter et al.,
2001; Karlin, 2002; Sixma and Smit, 2003).
14
Introduction
A
B
C
Extracellular domain
N-terminus
D
Transmembrane
domains MI - MIV
Ligand-binding sites
Cell membrane
Channel pore
Intracellular domain
Figure 1.4: The structure of nicotinic acetylcholine receptor and its binding protein. A:
The nAChR is a pentamer of five homologous subunits. The muscle receptor of the
stoichiometry (α1)2β1γδ is represented in this model. The receptor is depicted perpendicular to
the axis of the ion channel pore. For clarity, the γ subunit is not shown. Each subunit is
composed of four helical transmembrane domains (MI, MII, MIII, MIV). The MII domain of
all five subunits lines the channel pore. B: Top view of the pentameric receptor, viewed along
the five-fold axis, showing the association of the five subunits. The extracellular aminoterminal domain of the α1-subunit and the adjacent subunit (γ or δ) cooperate to form two
distinct binding pockets for acetylcholine (or other agonists or competitive agonists) at the
interface between the subunits. C, D: The acetylcholine from binding protein from snail brain,
a structural homologue of the nAChR ligand-binding domain. C: Each subunit is a single
protein domain. The cavity and pocket at each interface likely constitutes the ligand binding
site. D: As viewed perpendicular to the five-fold axis (Nirthanan and Gwee, 2004).
15
Introduction
1.5.2 α-Neurotoxins
α-Neurotoxins from Elapid and Hydrophiid snake venoms belong to the three-finger toxin
superfamily of non-enzymatic polypeptides containing 60 – 74 amino acid residues. The
characteristic feature of all three-finger toxins is their distinctive structure formed by three
adjacent loops that emerge from a small, globular, hydrophobic core that is cross-linked by
four conserved disulfide bridges (Endo and Tamiya, 1991; Menez, 1998). The three loops that
project from the core region resemble three outstrectched fingers of the hand (Figure 1.5). The
toxin is essentially a flat “leaf-like” molecule. In addition to the structural plasticity of the three
fingers, the three-finger fold is also amenable to a variety of overt and subtle deviations, such
as the number of the β-strands present, size of the loops and C-terminal tail as well as twists
and turns of various loops, all of which may have great significance with respect to functional
diversity and selectivity of molecular targets (Servent and Menez, 2001; Kini, 2002). Hence,
despite the similar overall fold, three-finger toxins demonstrate an assorted range of
pharmacological activities including, but not limited to, peripheral and central neurotoxicity,
cytotoxicity, cardiotoxicity, inhibition of enzymes such as acetylcholinesterase and proteinases,
hypotensive effect, and platelet aggregation (Tsetlin, 1999; Kini, 2002; Hodgson and
Wickramaratna, 2002). It has been proposed that three finger scaffold is used by snake to target
different combinations of functional groups, generating a panoply of target specificities.
A
Short-chain toxin
C
B
D
Weak toxin
Kappa toxin
Long-chain
i
Figure 1.5: The 3D-structures of three-finger neurotoxins from snake venoms that interact with
nicotinic acetylcholine receptors. The three-dimensional structures are shown in similar orientation
and in line ribbon representation. Disulfide bridges are shown in black. The species names and the
Protein Data Bank accession codes for structures are as follows: A: Erabutoxin-a (Laticauda
semifasciata,5EBX, Corfield et al., 1989) B: Candoxin (Bungarus candidus, 1JGK, Paaventhan et al.,
16
Introduction
2001) C: α-Cobratoxin (Naja kaouthia, 2CTX, Betzel et al., 1991) D: α-Bungarotoxin (B. multicintus,
2NBT, Sutcliffe et al., 1992). All are averaged NMR structures except erabutoxin-a, 2.0-Å crystal
structure and α-cobratoxin, 2.4-Å-crystal structure.
To date, more than 100 three-finger α-neurotoxins have been isolated and sequenced from
Elapidae and Hydrophiidae snakes. Depending on their amino acid sequence and tertiary
structures, α-neurotoxins can be classified into short-chain, long-chain, kappa and weak
neurotoxins. Although the primary target of all these categories of the three-finger neurotoxins
appears to be the muscle-type nAChR, some toxins are known to interact with other subtypes
of nAChRs such as neuronal nAChR.
1.5.3 Short- and long-chain neurotoxins
Based on the length of their polypeptide chains, α-neurotoxins were initially classified as shortchain α-neurotoxins that have 60 – 62 residues and four conserved disulfide bonds in common
positions, between Cys-3 and Cys-24, Cys-17 and Cys-45, Cys-40 and Cys-61, Cys-62 and
Cys-68 (Figure 1.5A). These neurotoxins bind with high affinity to muscular-type nAChRs.
The second group constitutes the long-chain α-neurotoxins consisting of 66 – 70 amino acid
residues with disulfide bonds, the fifth being between Cys-30 and Cys-34, in addition to the
four disulfide binds that are common in short-chain α-neurotoxins (Figure 1.5C).
Notwithstanding their classification a short-chain and long-chain neurotoxins, both types of αneurotoxins bind with high affinity to the Torpedo or muscle (α1)2β1γδ nAChRs (Servent and
Menez, 2001). Nonetheless, it has been reported that short-chain α-neurotoxins tend to
associate with the nAChR 6 – 7 fold faster and dissociate 5 – 9 fold faster than long-chain αneurotoxins (Chicheportiche et al., 1975). Apart from differences in structure, long-chain but
not short-chain α-neurotoxins are also able to bind with high affinity (Kd approximately 10-8 –
10-9 M) to neuronal homopentameric α7, α8 and α9 nicotinic receptors (Servent et al., 2000;
Antil-Delbeke et al., 2000).
17
Introduction
1.5.4 Kappa neurotoxins
κ-Neurotoxins form a new family of snake venom neurotoxins that are structurally related to
long neurotoxins. With the exception of κ-cobrotoxin, all the other κ-neurotoxins identified
consist of 66 amino acid residues and on the basis of amino acid alignment, contain five
disulphide bonds as in long neurotoxins (Figure 1.5D). The first κ-neurotoxin, κ-bungarotoxin
was purified from B. multicinctus venom and it is a minor component representing only 0.1%
of total venom (Grant and Chiapinelli, 1985). κ-bungarotoxin exists in physiological solution
as a dimer of identical subunits, each subunit having a molecular weight about 7.3 kDa. The
dimer is not covalently linked and is dissociated into monomers in the presence of sodium
dodecyl sulfate, urea, or high-ionic strength and high-pH buffers (Chiappinelli et al., 1985), in
contrast to monomeric α-neurotoxins either in solution or in crystal.
All κ-neurotoxins also contain the amino acid deletions in positions 16 – 20 and amino acid
additions between positions 36 and 40, which are characteristic of long neurotoxins (Fiordalisi,
1994). Moreover, the κ-neurotoxins display a unique single-residue insertion in position 52
(Fiordalisi, 1994). These toxins have shorter carboxyl-terminal tail than any other long chain αneurotoxins. κ-Neurotoxins bind with high affinity to the α3-containing neuronal nAChRs and
with low affinity to the α4-subunit-containing neuronal nAChR (Chiappinelli, 1992). Contrary
to short and long neurotoxins, κ-neurotoxins are weak antagonists to muscle-type nAChR
(Grant et al., 1985; Dewan et al., 1994; Sutcliffe et al., 1992) The absence of Trp-32 in all the
κ-neurotoxins could be the reason for the lack of binding to muscle-type nAChR (Grant, 1988).
To date, this group contains nine members isolated from Bungarus (Grant et al., 1985) and one
from Naja (Chang et al., 1998). The exact basis of the distinct physiological selectivity of these
structurally-similar α- and κ-neurotoxins is not well understood.
1.5.5 Weak neurotoxins
Weak neurotoxins, also known as atypical or miscellaneous toxins, constitute another class of
three-finger toxins that consist of 62 – 68 amino acid residues and five disulfide bridges.
However, unlike long-chain α-neurotoxins and κ-neurotoxins, the fifth disulfide bridge in weak
18
Introduction
neurotoxins is located in loop I (N-terminus loop, Figure 1.4B). Weak toxins are typically
characterized by a lower order of toxicity with lethal dose (LD50) varying from 5 – 80 mg/ kg
as opposed to prototypical α-neurotoxins with LD50 approximately 0.04 – 0.3 mg/ kg. Weak
neurotoxins from cobra venoms such as WTX (Naja kaouthia) and Wntx-5 (N. sputatrix)
produced a weak inhibition of muscle nAChRs in micromolar inhibitory concentrations (Utkin
et al., 2001; Poh et al., 2002). Candoxin from B. candidus was a potent inhibitor of muscle
nAChRs in low nanomolar range (IC50 approximately 10 nM). This toxin was also found to
inhibit neuronal α7 nAChRs at low nanomolar (IC50 approximately 50 nM) inhibitory
concentration (Nirthanan et al., 2002). It is conceivable that weak neurotoxins that bind weakly
to muscle nAChRs may yet have other unidentified molecular targets.
1.6 Applications of snake venom neurotoxins in research and therapy
Although early venom research was motivated by the desire for satisfactory cures for snake
envenomation, the general perspectives on animal toxins have changed dramatically due to the
accumulating data that has revealed a far wider scope for these natural biomolecules. It has
been 40 years since it was first realized that the physiologically active components of snake
venoms might have therapeutic potentials (Senior, 1999). These biomolecules have assumed
great importance as molecular probes and pharmacological tools to investigate the functional
biology of receptors and ion channels as well as providing lead compounds for the design of
clinically useful drugs (Harvey et al., 1998; Harvey, 2002a, b).
There is perhaps no better example to highlight the significant contributions made by venom
peptides to science and medicine than the discovery of α-bungarotoxin from Taiwan banded
krait, which has spawned the field molecular pharmacology by enabling the isolation and
characterization of nAChR from electric eel and other sources (Chang and Lee, 1963; Lee,
1972).
1.6.1 Viral infection
The possibility that nAChR is a host cell receptor for the neurotropic rabies virus was first
discussed by Lentz et al. (1982). These authors suggest that the virus binds at neuromuscular
junction and the binding is drastically reduced by the specific AChR antagonist α19
Introduction
bungarotoxin (α-Bgt). A stricking homology between a region of rabies virus glycoprotein and
the putative functional sites of snake venom neurotoxin has been demonstrated by Lentz et al.
(1984). This would imply a functional convergence between these distantly related proteins.
Purified rabies virus glycoprotein was able to compete with the potent neurotoxin of snake B.
multicinctus α-bungarotoxin for binding to acetylcholine receptor. Moreover, anti-peptide
monoclonal antibodies against glycoprotein 190-203 had been found to efficiently inhibit the
binding of both rabies virus glycoprotein and α-bgt to acetylcholine receptor. In general, the
regions of virus molecules involved in binding to the cellular components acting as viral
receptors might be structurally similar to the binding domains of cellular proteins that normally
bind to the cell components (Figure 1.6). Such similarity between parts of viruses and ligands
or normal cell constituents may provide a basis for the pathogenesis of some autoimmune
diseases because viral infection could be followed by production of antibodies to the idiotypes,
which would react with the cellular structures (Sege et al., 1983). Identification of the viral
domains that bind to cells should be important in the treatment of viral diseases.
Neri et al. (1990) also reported the homology of the neurotoxin toxic loop with the sequence of
164-174 of HIV-1 gp120. Human rhabdomyosarcoma cell line, TE671, is known to express a
muscle-like nicotinic receptor (Schoepfer et al., 1988) to which α-bungarotoxin binds with high
affinity. The binding of α-bungarotoxin to TE671 cells was found to be inhibited by gp120
from HIV-1 strain IIIB. Therefore, HIV infection of muscular and neuronal cells in vitro might
be mediated by gp120 binding to nicotinic acetylcholine receptor present in these cells; which
raises the same possibility that the same mechanism may be observed for HIV neurotropism.
The results suggest that nAChR may also bind HIV-1 gp120 in neuronal cells since α-bgt
binds to some population of nicotinic receptor expressed in the nervous system.
20
Introduction
Figure 1.6: A model showing homology of rabies glycoprotein with the “toxic” loop of the
neurotoxins. The segment of the glycoprotein (residues 174 to 202) corresponding to loop 2 of
the long neurotoxins (position 25 to 44) as determined by computer modeling is positioned
relative to a schematic representation of loop 2. Within circles, residues or gaps in the
glycoprotein are shown on the left and those in the neurotoxin on the right. One letter in the
circle implied the same residue in glycoprotein and toxin. Bold circles are residues highly
conserved or invariant among all the neurotoxins. A ten-residue insertion in the glycoprotein is
enclosed in the box. The rabies virus sequence is from CVS strain. Neurotoxin sequence is
from Ophiophagus hannah, toxin b (inset). Schematic representation of neurotoxin structure
showing positions of loops 1, 2 and 3 (Lentz et al., 1984).
1.6.2 Myasthenia gravis
α-Bgt played an important role in understanding the pathogenesis of myasthenia gravis (MG), a
disease which causes weakness of skeletal muscles. Muscular tissue of patients exhibited a
considerably reduced number of nAChR sites available for toxin binding (Mebs, 1989). The
autoimmune nature of the disease became evident when animals immunized with isolated
nAChR developed symptoms essentially identical to MG. It became clear that an autoimmune
attack directed to the body’s own nAChR causes accelerated degradation of these receptors in
skeletal muscles (Drachman, 1981). The challenge now is to find selective and efficient
21
Introduction
markers and blockers of those neuronal AChRs and perhaps weak neurotoxins can be used as
potential tools to answer this question.
1.6.3 Alzheimer’s disease
Recently, Nirthanan et al. (2002) identified candoxin from B. candidus as a reversible
antagonist of muscle nicotinic acetylcholine receptors and an irreversible ligand to neuronal
nicotinic acetylcholine receptor. This compound may be useful as a probe to label, identify or
isolate the α7-subtype of neuronal nicotinic receptor, in which this receptor is of great interest
in several disease conditions such as Alzheimer’s disease. The rapid onset and reversible
properties of candoxin, like d-tubocurarine which is used as a muscle relaxant, can be exploited
for therapeutic use.
1.6.4 Thrombotic disease
A potent glycoprotein from Dendroaspis jamesonii which is structurally related to short
neurotoxins was discovered by McDowell et al. (1982) to be platelet aggregation inhibitor. The
finding that an Elapidae snake has a protein with the general structure of a neurotoxin but with
the function of platelet inhibitor, is unique and have important implications for evolutionary
biology. This unique inhibitor named mambin is non-neurotoxic and may have therapeutic
potential for treatment of thrombotic disease (McDowell et al., 1982).
1.6.5 Research tools
Muscle nAChRs are pseudosymmetric pentameric complexes formed by four different subunits
(α, β, γ or ε and δ) whereas neuronal nAChRs identified so far are pentamers formed by one
(αx), two (αx, βγ) or three (αx, αy, βz) types of subunits (McLane et al., 1993). Neuronal
nAChRs, in addition to having alternative subunit stiochiometries, are composed of several
different subunit subtypes, with at least eight different α subunits and five different β subunits
being identified (reviewed in Deneris, 1991).
22
Introduction
By far, the most valuable probes to characterize muscle nAChR are long and short neurotoxins
from snakes which bind with extremely high affinity (KD = 10-12 – 10-9 M) to skeletal muscle
nAChRs (Chiappinelli, 1991). Occupation of the toxin of either one or both of the ACh
recognition sites on the α subunits of a muscle receptor prevents the ion channel associated
with the receptor from opening in response to cholinergic agonists (Neubig and Cohen, 1980).
In addition, long neurotoxins have been used to characterize neuronal nAChRs containing α7
subunits (Seguela et al., 1993). These neuronal receptors have a high affinity for the long
neurotoxins at KD = 0.7-1.7nM (Tindall et al., 1978; Wolf et al., 1988). For example, αbungarotoxin has been an important antagonist in establishing the functional properties of these
neuronal AChRs (Zhang et al., 1994). The α-bungarotoxin-sensitive neuronal receptors have
certain properties unlike other AChRs such as the ability to form functional homomeric
channels in oocyte expression studies and high permeability to Ca2+ ions (Seguela et al., 1993;
Alkondon et al, 1993).
κ-Neurotoxins are selective and potent antagonists used to characterize neuronal nAChRs in
which short and long neurotoxins have no effect (Chiapianelli, 1991). For example, κbungarotoxin had been used as a potent inhibitor of α3β2 neuronal nAChRs (McLane et al.,
1993).
One of the many applications of toxin as a probe was exploited by Sekhon et al. (1999). They
tested their hypothesis that maternal smoking during pregnancy is associated with alterations in
pulmonary function at birth and increase respiratory illnesses after birth. They found that the
direct results of these observations are due to nicotine binding with nAChRs. Labeled αbungarotoxin was used to label α7 nAChRs in developing monkey lung and the results
demonstrated that nicotine significantly increased α7 nAChRs in airway epithelial cells,
alveolar type II cells, alveolar macrophages and pulmonary neuroendocrine cells (PNEC).
Hence the use of labeled neurotoxin in this study can contribute to the understanding of
mechanistic changes that occur in the fetus of smoking mothers.
The role(s) of weak neurotoxin in the venom is still unknown and the challenge now is to fully
unravel its physiological importance in the snake.
23
Introduction
1.6.6 Novel functional homologs
Miwa et al., (1999) discovered a novel, endogenous homolog of snake neurotoxins, lynx1 in
mouse cerebellum, hippocampus, and cortex. This new relative of the family of elapid
neurotoxins was identified in the course of a search for developmentally regulated genes in the
cerebellum. Based on overall sequence similarity, conservation of cysteine-rich consensus
motif, and common gene structure with neurotoxin gene, lynx1 was classified as a small
(11kD) toxin-like (prototoxin) central nervous system protein (Figures 1.7A and B).
In
contrast to secreted snake neurotoxin, lynx1 is normally present at the cell surface as a
glycophosphatidylinositol (GPI)-anchored protein (Miwa et al., 1999). Further studies found
that this protein could associate and modulate nAChR, exhibiting preferential binding to α7
and α4 subunits (Ibanez-Tallon et al., 2002). Studies on mice carrying lynx1 null mutations
could shed light on the role for this protein in the CNS of mammals.
Figure 1.7: Comparison of the lynx1 model and the αBgt experimental structure. (A) The
figure shows the three-dimensional model of lynx1. Strands are shown as green arrows and
disulfide bridges are coloured yellow. (B) Experimental NMR structure of α-Bgt (PDB 1abt).
The orientation and colouring is the same as for lynx1. N- and C-terminal ends of both
molecules are labeled (Miwa et al., 1999).
24
Introduction
1.6.7 Chimeric toxins
It was observed that when a long neurotoxin whose characteristic fifth disulphide bond was
selectively reduced, it behaved like a short neurotoxin in terms of specificity of receptor
recognition (Servent et al., 1998). Addition of a fifth disulphide bond at the tip of the central
loop of a short neurotoxin increases its affinity for neuronal receptors. Ricciardi et al. (2000)
tested the idea that structural deviations between toxins adapting the same fold reflect
functional diversity. They also demonstrated that when a recombinant fasciculin/ toxin α
chimera containing loop 1 and tip of loop 2 of fasciculin was transferred into toxin α, the
chimera exhibited nearly all acetylcholinesterase-blocking activites of fasciculins (Ricciardi et
al., 2000). This would imply that toxin usually adopt a limited structural deviations which can
result in a multiple functional diversity.
1.7 Scorpion
Scorpions belong to arthropods in the animal kingdom and they are the largest as well as the
oldest arachids. They are representated by 1, 500 distinct species. To date, scorpions have a
wide geographical distribution and live on all major land masses except Antarctica. Their
habitats include desert, savanna, grasslands, temperate forests, tropical forests, rain forests and
the intertidal zones. Some remarkable species are found even at snow-covered mountain over
5, 500 m in altitude and in caves at depth of more than 800 m (Francke, 1982).
Scorpions can be divided into six families: Bothriuridae, Scorpionidae, Buthidae, Vejovidae,
Chlaerilidae and Chactidae. Only Buthidae scorpions produce neurotoxic secretions.
Scorpions which are dangerous to humans are found in desert or semiarid regions throughout
the world. Among the approximately 1500 scorpion species, only about 25 species are of
medical importance. Morbidity and mortality rates due to scorpion stings are especially high in
children have been reported in various countries. In Mexico, Brazil, North Africa, India and in
the Middle East (Mazzotti, 1963; Bucherl, 1971; Balozet, 1971; Bawaskar, 1989), scorpions
still possess a medical problem and life hazard to humans.
25
Introduction
1.7.1 Buthus martensi Karsch
The Chinese scorpion Buthus martensi Karsch (BmK) belonging to the Buthidae family is
widely distributed from northwestern China to Mongolia and Korea. The sting by this species
can cause excruciating local pain that might last for several hours and induce a series of
inflammation and pain responses (Balozet, 1971; Bai et al., 2003). They can grow up to 4.8 –
5.2 cm in length. As early as one thousand years ago, the pharmaceutical role of scorpions was
widely recognized by the Chinese. Li Shi-Zhen in 1578 described the properties and curative
effects of scorpions in detail. In Traditional Chinese Medicine at this present time, the whole
scorpion body especially the tail, is being used as a medicine to treat neuromuscular diseases
such as paralysis and epilepsy. In addition, scorpions have been used for treatment in apoplexy,
hemiplegic and facial paralysis.
Detailed knowledge about the biological properties of the venom of BmK has been generally
limited and only little is known about the neurotoxic components present in the venom of this
species. In the past years, BmK venoms has been studied extensively and this lead to the
discovery of 77 different peptides (Goudet et al., 2002).
1.7.2 Scorpion venom
Scorpions produce powerful venom in glands located in the distal end of its tail, known the
telson. The venom is injected into a victim through a sharp stinger. Scorpion venoms are lethal
to a broad spectrum of animals such as rodents (rats, mice and guinea pigs), rabbits, chicks,
fish, arthropods (fly larvae, crickets, moths and mealworm) and many others. There are
however, a few animals which are resistant to scorpions venoms such as the tarantula spider,
locust and of course, scorpions themselves.
Although Chinese scorpion is generally not dangerous to human, the venom is considered to be
a mixture of ‘rather active toxins’, suggesting that it had probably several toxic components
(Balozet, 1971). The toxicity (LD50, i.p) of this venom in mice was found to be 3.38 mg/ kg
weight (Gong et al., 1992), which is moderate in comparison with the LD50 of the venoms of
26
Introduction
other species. BmK venoms induce convulsions and paralysis in mammals, crusteacea and
insects.
Scorpion venoms, like the snake venom contain a complex protein composition. They consist
of mucopolysaccharides, small amounts of hyaluronidase and phospholipases, low molecular
weight compounds such as noraderenaline, serotonin,
histamine, protease inhibitors and
histamine releasers and multiple basic, predominantly neurotoxic proteins (Simard and Watt,
1990; Martin-Eauclaire and Courad, 1995;). The scorpion venom contains an array of several
structurally distinct families of peptides that modulate ion channels. These toxins can be
classified into four groups according to their effects on specific ion channels, such as Na+, K+,
Ca2+ and Cl- ion channels (Garcia et al., 1997; Gordon et al., 1998; Tytgat et al., 1999;
Valdivia and Possani, 1998; Possani et al., 1999).
Ion channels are crucial elements for the activity of living cells, targeting these membranebound proteins is thus an efficient means for venomous animals to capture their preys or to
defend themselves against enemies. Natural toxins also provide powerful tools for
understanding the physiological contribution of ion channels to cell and organ behaviour and
for probing and correlating ion channel structure and function. Moreover, elucidation of the
mechanisms of toxin action especially on their three-dimensional structures provide wider
perspectives in designing of drugs (Menez, 1998).
The best studied peptides are long-chain toxins that contain 60 – 70 amino acids residues with
four disulfide bridges (Possani, 1984). The targets of these toxins are voltage-sensitive Na+
channels of excitable muscle and nerve cells. These ion channel toxins can be divided into αtoxins and β-toxins, based on their physiological effect on the opening and closing kinetics of
Na+ channels. The α-toxins prolong the action potential by blocking Na+ channel inactivation
and β-toxins shift the voltage activation towards a more negative potential thereby affecting
Na+ channel activation and promoting spontaneous and repetitive firing (Couraud et al., 1982;
Catterall et al., 1986).
27
Introduction
The difference in the effects of the α- and β-toxins is attributed to their ability to bind to two
distinct sites on the Na+ channel receptor. α- and β-toxins bind to sites 3 and 4 on the Na+
channel respectively (Jover et al, 1980). Alternatively, Na+ channel toxins classified based on
their species specificity as insect-, mammal-, and crustacean-specific toxins (Gordon et al.,
1996; Meves et al., 1984; Garcia et al., 1997). An exception to this rule is toxin AaH IT4
which is cross-species specific affecting both insects and mammals (Loret et al., 1991).
Hundreds of distinct peptides specific for Na+ channels have been purified from 20 – 30
species of scorpions; at least 120 complete primary structures have been identified.
On the other hand, a subset of short-chain scorpion toxins has also been found to interact with
the voltage-dependent or Ca2+-activated K+ channels or with Cl- channels. Toxins that affect
voltage-gated K+ toxins typically contain 31 – 38 amino acids with three or four disulfide
bridges (Garcia et al., 1997). A recently purified 23-residue toxin from the scorpion Tityus
cambridgei was reported to block K+ channel (Batista et al., 2000). Two scorpion peptides
have been reported to modify the binding of ryanodine to Ca2+ - channels. Imperatoxin A,
which is 33 amino acids long, increases the binding of ryanodine resulting in continuous Ca2+
channel stimulation (Zamudio et al., 1997a). Imperatoxin I is a heterodimer which blocks Ca2+channel by inhibition of ryanodine binding (Zamudio et al., 1997b).
Scorpion toxins that interact with Cl- channels and their homolog are approximately 36 amino
acids long with four disulfide bridges (Lippens et al;, 1995; DeBin et al., 1993). Chlorotoxin is
the first high-affinity peptide ligand for Cl- channels. In recent years, Cl- channel have been a
significant focus of interest as defect in this channel is the cause of cystic fibrosis.
1.8 Microarray
A microarray is an ordered array of microscopic elements that allows specific binding of genes
or gene products. Microarray is a new scientific word derived from the Greek word mikro
(small) and the French word arayer (arranged). Microarrays, also known as biochips, DNA
chips and gene chips contain minute quantities of DNA, protein or small organic compounds
which can be probed with possible binding ligands. The first use of microarray was in
28
Introduction
immunological assays but progress in genome sequencing has created a demand for
simultaneous multi-gene analysis (Ekins, 1989; Ekins and Chu, 1992).
Microarray technology developed in the early 1990s by Mark Schena and collegues at Stanford
University rely heavily on six major disciplines: biology, chemistry, physics, engineering,
mathematics and computer science (Schena et al., 1995). The advent of this miniaturized
technology for the study of molecule-molecule interactions has changed the way that new
challenges are approached in cell biology. In addition, major advances in detecting
fluorescence-based experiment had enabled simultaneous analysis of thousands of variables in
a single experiment (Howbrook et al., 2003).
1.8.1 DNA microarray
Array-based gene expression analysis in which immobilized DNA probes hybridizing to RNA
and cDNA targets has become an important primary tool in many research projects. Expression
analysis for the quantitative gene expression of many genes can be performed using one- or
two-color fluorescent schemes. One colour analysis is primarily used for arrays prepared using
photolithography method. In 1991, Fodor described an innovative application of
photolithography, which has been used in the industrial production of electronic semiconductor
devices. Photolithography uses ultraviolet light and solid-phase synthesis to manufacture
microarray (Fodor et al., 1991). This light-based synthesis approached became one of the
widely used methods in the manufacture of microarray. Briefly, we attached synthetic linker
modified with photochemically removable protecting groups to a glass substrate and direct
light through a photolithographic mask to specific areas on the surface to produce localized
photodeprotection. The first series of chemical building block is incubated with the surface and
chemical coupling occurs at those sites that have been illuminated in the preceding step. Next,
light is beamed to different regions of the substrate by a new mask and chemical cycle is
repeated (Pirrung et al., 1998). Photolithography allows the construction of arrays with
extremely high information content. Since the arrays are constructed on a rigid material (glass),
they can be mounted and inverted in a temperature-controlled hybridization
29
Introduction
Experimental design
Biological experiment
Sample B: eg.
treated sample
Sample A: eg.
untreated sample
RNA extraction
RNA sample A
RNA sample B
Fluorescent labeling
Microarray
fabrication
Labeled sample A
Labeled sample B
Microarray
Microarray
hybridization
Image
acquisition
Image
analysis
Data preprocessing
and normalization
Identification of
differentially expressed
Figure 1.8:
Exploratory data
analysis
Classification
Other analyses
(e.g. pathway
Flowchart showing a typical microarray experimental procedure. After
carrying out the biological experiment, the samples from tissues or cells, are collected. For
example, sample A is the control experiment and sample B is the tested experiment. Total
RNA is isolated and labeled with fluorescent dyes and co-hybridized with the microarray. The
hybridized microarray is scanned to acquire the fluorescent images. Statistical analysis can be
used to infer the microarray data, depending on one’s aim of study. Image adapted from Leung
and Cavalieri, (2003).
30
Introduction
chamber (Lipshutz et al., 1999). A fluorescently tagged nucleic acid sample injected into the
chamber is allowed to hybridize to the complementary oligonucleotides on the array. Laser
excitation enters through the back of the glass support and focused at the interface of the array
surface and target solution. The fluorescence emission caused is collected by a lens and passed
through a series of optical filters to a sensitive detector which converts it to a quantitative twodimensional fluorescence image (Ekins, 1989; Ekins and Chu, 1992). A summary of a typical
microarray experiment is depicted in Figure 1.8.
1.8.2 Applications of DNA microarray
Gene expression studies using DNA microarray accounts for 81% of the scientific
publications to date, but microarrays are being used for many other purposes including
genotyping, tissue analysis, and protein studies. In the field of development, microarrays can
be employed to build a database of gene expression levels as a function of cell and tissue type.
These databases are extremely valuable as they provide deeper understanding of the basic
mechanisms that control multicellular development and might shed some insights into the
pathological cellular events, including the onset and progression of human disease.
The onset and progression of human disease are determined by a complex set of factors,
including genetics, diet, the environment, and the presence of infectious agents. Microarray
makes a unique tool in studying each of these contributing factors. For example, gene
expression patterns in brain tissue from normal individuals and Alzheimer’s patients can be
studied to elucidate the genetic basis of this illness. Almost all human diseases can be studied
using microarray analysis and the ultimate goal of this work is to develop treatments or cures
for every human disease by 2050 (Schena, 2003).
Drug discovery is one of the hottest areas in research and development. Many known drugs
impart their therapeutic activity by binding to specific cellular targets, inhibiting protein
function, and altering the expression of cellular genes. In principle, it is possible to use
microarray for drug discovery and clinical trials by generating gene expression profiles in
patients undergoing disease progression or drug treatment. Many illnesses lead to specific
31
Introduction
changes in gene expression and drugs that reverse these changes might be beneficial.
Expression profiling may be useful in identifying drugs that alter gene expression of nontarget
genes, as a means of identifying drugs with possible side effects. This might lead to safer
medicines with minimal side effects. In addition, this technology can be used for patient
genotyping, and the capacity to partition the population into drug responders and
nonresponders based on genotype may enable more personalized medicine.
Microarray has also been successfully used for providing the biological and medical
community with a greater understanding of venom – induced pathologies and the subsequent
treatment of envenoming (Gallagher et al., 2003). This microarray – based toxicological
approach was employed in vivo to investigate the effects of phospholipase A2 from Naja
sputatrix to study the change in global gene expression especially on aquaporins and Na+/ K+ATPase genes (Cher et al., 2003). Another group used an in vitro system to study venominduced apoptosis in human umbilical vein endothelial cells by use of subpathological levels of
Crotalus atrox and Bothrops jararaca (Gallagher et al., 2003). These studies collectively
demonstrate that snake venom components can be used as biologically active proteins in
pharmaceutical lead compounds.
1.9 Neurogenesis
The pioneering neuroscientist Ramon y Cajal (1928) observed that:
“In adult centers the nerve paths are something fixed, ended, immutable. Everything may die,
nothing may be regenerated. It is for the science of the future to change, if possible, this harsh
decree.”
The mature central nervous system (CNS) was distinguished from the developing nervous
system by the lack of growth and cellular regeneration. The fixed neuronal population of the
adult brain was understood for necessary maintenance of functional brain circuitry. This might
account for the lack of endogenous CNS repair following injury or disease.
32
Introduction
However, in the last several decades, mounting evidence has led to the view of the CNS as a
dynamic, plastic organ, endowed with some potential for self-repair and regeneration. Great
progress have been made in the understanding of continued neurogenesis in adult brain and this
has raised hopes that self-renewal leading to structural repair by neurons may be possible in
new neurons (Hallbergson et al., 2003).
Neurogenesis in the adult brain can be divided into three phases in accordance with the
sequence of neurogenesis during CNS development: (a) proliferation, where new cells are
generated; (b) migration towards the target areas; and (c) terminal differentiation into distinct
phenotypes (Hallbergson et al., 2003).
Neurogenesis is a term which implies progression through differentiation and not only where
cases of proliferation is studied. Studies have found that new functional neurons are generated
in two adult germinal centres, adding further complexity to the definition of adult plasticity to
include circuitry (van Praag et al., 2002).
An important observation showed that adult neurogenesis is not static but indeed responds to
environmental factors such as physical activity, environmental enrichment, caloric restriction
and modulation of neural activity (Peterson, 2002; Ray and Peterson, 2002). The
responsiveness of adult neurogenesis to environmental influences may suggest that its
regulation is under the control of expressed factors whose level of availability dictates the rate
of neurogenesis (Hallbergson et al., 2003).
The spatial and temporal expression of various trophic or growth factors play a role in guiding
cell-fate choices and determine the size of the neuronal population. In addition to protecting
neurons, trophic factors have been shown to stimulate proliferation of adult-derived stem cells
and to instruct their differentiation. Examples of trophic factors include amphiregulin and
fibroblast growth factor (FGF) (Kuhn et al., 1997, Zigova et al., 1998; Pencea et al., 2001;
Benraiss et al., 2001; Falk et al., 2002). Amphiregulin is part of the family of endothelial
growth factor (EGF) protein and it has have been suggested to play a role in stem cell
proliferation and neurogenesis in the adult brain (Falk et al., 2002). FGF-2 on the other hand,
33
Introduction
is another potent mitogen for a variety of cells and modulates embryonic development and
differentiation, adult angiogenesis, wound healing and tissue repair. In the brain, FGF-2 is
important as a survival factor and is neuroprotective against a variety of insults (Peterson and
Gage, 1999).
So far, about 70 toxins from BmK have been identified. They are mostly α and β-toxins as well
as toxins blocking K+ and Cl- channels. These toxins have been identified mostly using
conventional method such as proteomics. It would be interesting to see if BmK might have any
pharmacologically important factors that can affect neurogenesis in human brain cells.
1.10 Database creation
The first characterization of α-bungarotoxin 40 years ago led to a boom in the discovery of
other similar snake neurotoxins. In 1990, there are about 100 known neurotoxin protein
sequences in GenBank. However, to date, there are approximately 555 neurotoxin entries in
public databases, from snake venom alone. Therefore, the accumulation of snake venom
neurotoxins (svNTXs) had greatly increased over the years and this makes it an attractive
bioinformatics study for further functional and structural characterizations of svNTXs.
Established methods of determining functions of svNTXs are based on study of chemical
mutagenesis (reviewed by Karlsson, 1979), structural studies (reviewed by Dufton and Hider,
1983b) and pharmacological and biochemical characterization experiments (Endo and Tamiya,
1991). Many research groups have focused on isolating, purifying and characterizing
individual toxins or small groups of toxins. The number of characterized svNTXs reported in
the literature and molecular databases is increasing rapidly. In addition, functional properties of
individual svNTXs are poorly annotated in molecular databases. Insufficient cross-linking
between databases leads to redundancy and often conflicting information, indicating potential
errors in data.
The growing number of well-studied svNTXs and the high complexity of related information,
such as their structural and functional properties have created a need for improved data
34
Introduction
management. The analysis of svNTXs data is growing increasingly complex and there is a need
for a single, well annotated, and cleaned database, which will facilitate svNTXs analysis.
Data for the study of svNTXs are scattered across multiple sources. Hence, there is a need for
specialized database in svNTXs to be used as a tool for the study of svNTXs. The database
contains entries collected from different sources, organized, analysed and classified according
to their structure-function relationship. The svNTXs were annotated for their pharmacological
properties and enzymatic activity from literature sources. From the data, pharmacological
groups were formed to classify svNTXs based on structure and function relationship. This
general classification of svNTXs can be used to identify putative residues for each
pharmacological property by sequence comparison between the groups. Well organized and
rich data allow the use of sophisticated tools for improved structure-function analysis.
1.10.1 Current problems with analysis of snake toxins using bioinformatic resources
Current data on svNTXs are deposited in public repositories GenBank, EMBL (Stoesser et al.,
2001), DDBJ (Tateno et al., 2000), SwissProt, and PIR (Barker et al., 2001). The svNTXs
three dimensional structures are deposited in the PDB database. Functional properties of
svNTXs can be found mainly in published articles while functional annotation of sequences in
public sequence databases is very limited (Brusic et al., 2000). Structural properties of some
svNTXs can be found in the PDB database and published articles. Of 272 svNTXs reported in
February 2004, 1.5% have been published in journals articles only, and the remaining 98.5%
have been found in public databases.
A recent analysis on scorpion toxin data (Srinivasan et al., 2002) from public databases
revealed the presence of numerous errors in the sequences, incomplete data, poor annotation
and discrepancies of information for the same entry from different sources. For example, some
of the errors encountered were wrong link between databases, different names for the same
sequence, different sequences for the same toxin and absence of link between databases. In
order to minimize these errors, certain measures such as cross-checking with original
publications were taken. In the context of all known data, a unified classification of svNTXs
35
Introduction
does not exist. Detailed classification based on structure-function relationship was also
constructed. Several bioinformatics tools for analysis of biological data include: 1) pairwise
sequence alignment, ii) multiple sequence alignment (local and global), ii) phylogenetic
analysis, iii) 3-D structure analysis.
i)
Pairwise alignment is carried out to locate the conserved regions between two
sequences. In global alignment, entire sequences are aligned at the same time using
extension of dynamic programming, while in local alignment, conserved regions are
found by statistical methods, derived by removing stretches of global alignment.
BLAST 2.0 (Altschul et al., 1997) and CLUSTAL X 1.82 are used for local and
global alignment respectively.
ii)
Multiple sequences alignment is the hierarchical extension of pairwise alignment.
They are widely used to identify conserved motifs and to predict functional role. It
is also a basis for phylogenetic analysis.
iii)
Phylogenetic analysis helps to determine which sequences are more closely related
evolutionarily.
36
Introduction
1.11 Aims of the project
The aim of this project has been divided into two parts. The initial part of the project has been
to build a snake venom neurotoxin database.
The specific goals are:
1) To build a clean information enriched database for svNTXs.
2) To classify toxins based on their structure and function relationships.
3) To provide an in-house platform to predict function of new toxins.
The second aim of the project has been to screen for potential therapeutic agent(s) in the
venom of Chinese scorpion, Buthus martensi Karsch (BmK) using DNA microarray and realtime PCR. The motivation is to elucidate some of the biological basis of using scorpion venom
in traditional medicine. This scorpion venom has been found to have effects on the
cardiomuscular system (Gong et al, 1992) in addition to having analgesic effect and antitumor
activity (Liu and Pei, 1989; Zhang et al., 1987). It will be interesting to discover novel drug
leads in this well studied scorpion.
The specific goals are:
1) To examine the subtoxic effects of venoms on gene expression on human
neuroblastoma cells and to highlight its potential use in the drug discovery.
2) To screen for active fraction(s) in the venom affecting the expression of genes involved
in neurogenesis and angiogenesis using real-time PCR.
37
Materials and Methods
MATERIALS AND METHODS
Materials and Methods
CHAPTER TWO
MATERIALS AND METHODS
Section A:
2.1 Bioinformatics
The flowchart below demonstrates the overall picture of the entire bioinformatics project.
Public sequence databases
Data
Keyword search retrieval
BioWare
Raw data set
Filtering
Initial data set
Further cleaning
Final preliminary data set
Data integrated to BioWare format
Redundant entries removed
Final data set
PDB
svNTX, 3-D structures
svNTX
database
Functional
annotation
Published
journal reports
Analysis
Applications of database
CysAlign
Disulfide bridge patterns
Pharmacological properties
Classification tree
38
Phylogenetic analysis
Materials and Methods
2.2 Data collection and cleaning
Structural and functional data in svNTX database where obtained from various sources. Protein
sequences information was gathered from public databases such as GenBank, SwissProt and
EMBL. The three dimensional (3-D) structures of svNTXs were extracted from PDB. The
BioWare system available on-line at http://sdmc.a-star.i2r.org.sg/Templar from Institute for
Infocomm Research (I2R) was used to facilitate data retrieval from public databases.
Protein sequence entries were gathered from the public databases by query ‘Serpentes and
neurotoxin’ using keyword search via BioWare Retrieve module. The return result, a raw data
set, underwent a preliminary filtering to remove irrelevant records, such as neurotoxins from
other species and neurotoxin fragments. Records not selected at this step were recorded into a
rejected list by BioWare to avoid future retrieval of the same entry during database updates. The
raw data set after preliminary filtering will be referred to as the initial data set.
Data from various databases vary in format. There was a need to integrate these data into one
single format. Therefore the data were merged and formulated for the BioWare integrated format.
The final preliminary data set was next checked for duplicate entries. This was facilitated by the
BioWare preparation module, which generates a sequence comparison report summary, based on
pairwise alignment of etries in the data set. Entries having the same sequence (100%) identity
were treated as duplicates. Duplicates of the same name and taxon (organism source) were
compared; the entry judged to contain the most complete information was kept and the rest were
deleted. Duplicates from different species were retained in the data set. Sequences having shared
fragment or partial identity, where one sequence was an identical fragment of another, were
checked for their uniqueness by referring to journals and cross-references field from the public
39
Materials and Methods
databases. Most of the entries that formed earlier versions of entries in the data set were also
removed. All the deleted entries were added to the rejected file. The resultant data set was
considered as the final data set.
The
cleaned,
final
data
set
is
available
on-line
at
http://sdmc.a-
star.i2r.org.sg/Templar/DB/snake_neurotoxin which was created using the BioWare-Templar
module. In addition, protein 3-D structures were searched in the PDB databases. PDB structures
were extracted and added to the svNTX database. Sequence entries and their corresponding 3-D
structures were then manually linked.
2.3 Data annotation
Nearly 300 published journal articles and papers listed in the database were analysed for data
enrichment with functional and structural enrichment. Six new fields containing information
normally absent in public database entries were created for each entry. These fields are
Action_site, Physiological_function, Critical_residues, Toxin_activity, Binding_affinity, and
Miscellaneous. Action_site denotes the site of action of svNTXs. Physiological_function and
Toxin_activity refer to published svNTXs function and LD50 values, respectively.
Critical_residues reveal the critical amino acid residues that are involved in toxin-receptor
interaction. Information from binding studies performed on svNTXs-receptor is represented by
the Binding_affinity field. Miscellaneous field refers to other additional information relevant to
functional or structural description of a svNTX. The description of data annotation is further
discussed under results section.
40
Materials and Methods
2.4 Data analysis
The svNTXs database was analysed to form a classification chart for the svNTXs based on
structure and function. The disulfide bridge patterns, evolutionary relationships and
pharmacological properties were used for the analysis. Sequence comparison of svNTX was
carried out using CLUSTALW with the default parameters. From the multiple alignment
analysis, it is evident that svNTXs contain highly conserved cysteine residues. Cysteine residues
in a protein usually form a disulphide bridge with another specific cysteine residue. Classification
of protein family based on cysteine pairing pattern has been used to classify scorpion toxins (Tan
et al., 2001). It might be interesting to test this pairing pattern in svNTXs. The use of CysAlign
tool in BioWare is diagrammed below.
Database in flat file format
Plot disulphide bridge pattern for each entry
that has disulphide bond information
Search through each entry for information on disulphide bond
Group entries with similar disulphide bridge pattern
Figure 2.1: A schematic representation of CysAlign methodology.
41
Materials and Methods
2.5 Phylogenetic study
Signal and leader sequences were removed and only mature, full length amino acid was used for
phylogenetic studies. Phylogenetic studies were undertaken only for groups having more than
four or more members for better statistical realiability. CLUSTAL-X (Thompson et al., 1997)
was used for multiple alignment of amino acid sequences. The pairwise alignment parameters
used were a gap opening penalty of 35 and a gap extension penalty of 0.75 whereas the multiple
alignment parameters were a gap opening penalty of 15 and a gap extension penalty of 0.30.
Each phylogenetic subgroup was generated by using the SEQBOOT program of PHYLIP 3.6a2
(Felsenstein, 1985; 2001). Statistical reliability of the trees was assessed using 100 bootstrap
replications. The bootstrap values of 90 and above are considered to be significant. The most
parsimonious trees were calculated using the PROTPARS program using the maximum
parsimony (MP) method (Saitou and Nei, 1987). MP method was chosen because this program
takes into account the number of changes required at the nucleotide level to substitute one amino
acid for another. In this scheme, the most accurate phylogenetic tree is one that is based in the
fewest changes in the genetic code. The strict consensus trees were obtained using the
CONSENSE program (Felsenstein, 2001). The rooted tree diagrams were generated with the
TREEVIEW program (Page, 1996). Bee venom PLA2 (Swiss-Prot A59055) and Lynx1 (SwissProt AAF16899) were utilised as outgroups for analysis of presynaptic and postsynaptic NTXs
subgroups, respectively.
2.6 Bioinformatics tools used
BioWare (Biological Data Warehousing) is a convenient, user-friendly tool for rapid building of
data warehouses containing biological information. It contains different programs to retrieve raw
data from diverse sources on the Internet, formulate and integrate this information into a central
42
Materials and Methods
repository and for rapid building of personal databases using a well-defined framework of the
resultant database. This software is divided into three main modules which are used in the
creation of svNTX database.
1.
BioWare-Retrieve obtains the required data from GenBank, Swiss-Prot and PDB sources.
2.
BioWare-preparation allows manual annotation of the data prior to creation of the
database.
3.
Templar creates a personal searchable database as according to the specifications of the
users on the uploaded data file.
CysAlign facilitates in the grouping of entries in the database which share similar disulphide
pattern, provided the pairing information is present in the entry. These groupings can be used to
classify similar protein structurally.
Clustal X, is a general purpose multiple alignment program for protein or DNA. BLAST (Basic
Local Alignment Search Tool) is a local alignment program that uses a heuristic algorithm to
detect relationships among sequences which share only isolated regions of similarity (Altschul et
al., 1990). BLAST output assigns a statistical score to sequences similar to query sequence,
making it easier to differentiate real and false hits.
PHYLIP (the PHYLogeny Inference Package) is a set of modular programs used for inferring
phylogenies based on evolutionary trees. It contains a broad variety of analytical methods such as
parsimony, distance matrix, and likelihood methods. This program was used to build evolutionary
trees in this project.
43
Materials and Methods
Section B: Experiment protocols
2.7 Scorpion venom and venom glands
The venom and venom glands of the Chinese scorpion, Buthus martensi Karsch were
obtained from Wuhan University, China.
2.8 Purification of scorpion venom
Lyophilized crude venom (20 mg) was reconstituted in 2 ml water and separated on a gel
flitration chromatography using Sephadex G50 column. Peak II of eluant was subjected to RPHPLC (Amersham) using a Jupiter C18 or C4 µBore column. The buffer systems used were 0.1%
of trifluoroacetic acid (Buffer A) and 80% acetonitrile in 0.1% trifluroacetic acid (Buffer B).
Flow rate was maintained at 1 ml/min. Individual fractions were collected, dried by speed vac,
and reconstituted in 100 µl of water and subjected to gene expression studies.
2.9 Purification of snake Bcntx-4
Bungarus candidus crude venom (4 mg) was purified on RP-HPLC (SMART system,
Pharmacia) using a Jupiter C4 column. The buffer systems used were 0.1% of trifluoroacetic
acid (Buffer A) and 80% acetonitrile in 0.1% trifluroacetic acid (Buffer B). Flow rate was
maintained at 1 ml/min. Individual fractions were collected, dried by speed vac, and reconstituted
in 100 µl of water and subjected to receptor binding studies.
44
Materials and Methods
2.10 Mass spectrometry determination and N-terminal amino acid sequencing
The RP-HPLC purified proteins were subjected to N-terminal amino acid sequencing using a
liquid-phase sequencer equipped with an on-line amino acid analyzer after analysis by
matrix-assisted light desorption ionization mass spectrometry-time of flight (MALDI-TOF).
2.11 Protein estimation
Protein concentrations were determined by Bradford microassay procedure (Bradford, 1976)
using Bio-Rad reagents. The samples were diluted 5 times and 1 ml of 5 times diluted dye
concentrate reagent was added into the sample. After 15 minutes of incubation, the
absorbance was read at 595 nm. A standard curve was drawn using bovine serum albumin
(BSA) at concentrations ranging from 2 –10 µg/ml.
2.12 Cell culture and medium
Human neuroblastoma cells (ATCC number: CRL2270) were grown in DMEM medium
supplemented with 10% fetal bovine serum, 50 units of penicillin, 50 µg/ml streptomycin.
Cells were cultured in incubator using 5% CO2 and 95% air at 37°C.
2.13 MTT cell viability assay
MTT test is a colorimetric assay system which measures the reduction of 3-(4, 5dimetylthiazole-2-yl)-2, 5-diphenyl tetrazolium bromide; MTT) into an insoluble formazan
product by activity of the mitochondrial enzymes in viable cells. 25 mg of MTT was
dissolved in 50 ml of DMEM to a concentration of 0.5 mg/ml, filtered and stored in dark until
use. A 96 well plate was seeded with 5000 cells per well and allowed to grow for 24 hrs to
80% confluency at 37°C with 5% CO2. The cells were then treated with 600, 400, 200, 100,
45
Materials and Methods
50 µg/ml of BmKII fractions and incubated for 24 hrs. After incubation, the media was
removed, cells washed twice with 1x saline and 60 µl MTT was added. MTT incubation was
carried out for 1 hr. The medium was removed and the undissolved purple formazan crystals
were solubilized in 250 µl of dimethylsulfoxide (DMSO). Complete dissolution of crystals
was achieved by shaking plates gently for 10 min at room temperature. Blanks were set up in
wells that did not contain cells. Absorbance at 550 nm wavelength was measured
spectrophotometrically. Viability of exposed cells was expressed as a percentage of untreated
cells (100%).
2.14 Microarray
Neuroblastoma cells were treated with 35 µg/ml of BmKII fraction for 18 hrs. Total RNA
was isolated according to protocol in section 2.25. RNA isolated was processed and
hydridized to each array of the U133 GeneChip™ Array Set according to the protocols
described in the GeneChip™ expression analysis technical manual (Affymetrix, Santa Clara,
CA). Untreated sample was used as a negative control. Data from each treatment were scaled
to an average intensity of 800. Relative mRNA expression levels were expressed as plus or
minus fold changes compared with untreated control using Microarray Suite software 5.0
(Affymetrix, Santa Clara, USA). All genes showing 4-fold or more were included in the
subsequent analyses. Cluster members were classified according to their biological function
as described in the NetAffy database (Affymetrix, Santa Clara, USA).
2.15 Total RNA cleanup
Total RNA (20 –50 µg) was cleaned up using RNeasy spin columns (QIAGEN) before
microarray analysis. The final volume of total RNA is made up to 100 µl with sterile water.
46
Materials and Methods
RLT buffer (350 µl) was then added into the RNA and transferred into a spin column. The
tube was spun for 2 min. Eluant was collected and spun again for 2 min. Eluant was
discarded and 500 µl of RPE buffer was added in this washing step. Tube was spun again for
2 min and waste discarded. The column was air dried until β-mercaptoethanol was not
apparent. RNase-free water (15-30 µl) was added into column, waited for 1 min, followed by
centrifugation for 2-3 min. The eluted RNA was quantitated spectrophotometrically at 260
and 280 nm.
2.16 cDNA synthesis
A total of 5 µg of cleaned RNA was used for first strand synthesis.
2.16.1 Primer hybridization
Reagents used in the first strand synthesis:
Total RNA
5 µg
T7-d(T) 24 primer (100 pmol/µl )
1 µl
DEPC-H2O
for final reaction volume of 20 µl
The above reaction mix was incubated at 70°C for 10 min. Then, the tube was briefly pulsed
and put on ice.
2.16.2 Temperature adjustment
The above reaction was then added into the following reagents and incubated at 37°C for 2
min.
5x first strand cDNA buffer
4 µl
100 mM DTT
2 µl
10 mM dNTP mix
1 µl
47
Materials and Methods
2.16.3 First strand synthesis
SuperScript II RT (1 µl) was added to the above tube and incubated at 37°C for 1 hr. The
reaction was terminated on ice. The tube was centrifuged briefly to bring down condensation
of water on the sides of the tube.
2.17 Second strand cDNA synthesis
The second strand synthesis was immediately carried out after the first strand synthesis. The
following reagents were added to the first strand synthesis tube.
DEPC-treated water
91 µl
5x second strand reaction buffer
30 µl
10 mM dNTP mix
3 µl
10 U/µl E.coli DNA ligase
1 µl
10 U/µl E.coli DNA polymerase I
4 µl
2 U/µl E.coli RNase H
1 µl
The contents in the tube were gently tapped and briefly spun to remove condensation. Then,
the reaction mixture was incubated at 16°C for 2 hrs in a cooling waterbath. Following 2 hrs
incubation, 2 µl [10 U] of T4 DNA polymerase was added and incubated again at 16°C for 5
min. The reaction was terminated by addition of 10 µl 0.5 M EDTA.
48
Materials and Methods
2.18 Cleanup of double stranded cDNA
The clean-up was carried out in a two-step reaction. The first step involved the phase lock
gels (PLG)-phenol/chloroform extraction and the final step involved ethanol precipitation.
PLG form an inert, sealed barrier between the aqueous and organic phases of phenolchloroform extractions. The solid barrier allows more complete recovery of the sample
(aqueous phase) and minimizes interface contamination of the sample. A standard
phenol/chloroform extraction can be performed as an alternative to the PLG procedure.
The PLG tube was briefly spun for 12, 000 x g for 20-30 sec. 162 µl
of
phenol:chloroform:isoamyl alcohol in a 25:24:1 ratio saturated with 10 mM Tris-HCl pH 8.0,
1 mM EDTA) was added to the final cDNA synthesis preparation. The tube was vortexed
briefly. The entire 324 µl cDNA-phenol/chloroform mixture was added to the PLG tube. This
suspension was centrifuged at 12,000 x g for 2 min. The upper aqueous phase was transferred
to a fresh 1.5 ml tube. In the preceding step, 0.5 volumes of 7.5 N ammonium acetate and 2.5
volumes of absolute ethanol were added to the sample and vortexed. The mixture was
centrifuged at 12, 000 x g for 20 min. The supernatant was removed and the pellet was
washed with 0.5 ml of 80% ethanol (stored at -20°C). The tube was then centrifuged at 12,
000 x g at room temperature for 5 min. The 80% ethanol was carefully removed and the 80%
ethanol step was repeated once again. The pellet was air-dried. The dried pellet was
resuspended in a small volume of RNase-free water (12 – 15 µl).
49
Materials and Methods
2.19 Synthesis of biotin-labeled cRNA
The RNA labeling transcript step is important for the production of large amounts of
hybridizable biotin-labeled RNA targets by in vitro transcription from bacteriophage T7 RNA
polymerase promoter. The use of T7 RNA polymerase and biotin-labeled nucleotides, large
amounts of single-stranded nonradioactive RNA molecules can be produced during short
incubation period. RNA-DNA hybrids have a higher melting temperature than corresponding
DNA-DNA hybrids, therefore, single stranded RNA targets offer higher target avidity and
greater sensitivity than DNA probes. The RNA targets offer selectivity unavailable with
DNA targets – being single stranded, they are strand specific and hybridize more effectively
to probes because the target population does not self-hybridize. Microchip array assays
effectively used RNA transcripts that are labeled with biotin-modified ribonucleotides. The
biotin labeled RNA targets that are hybridized to arrays of DNA probes can be detected by a
reporter molecule linked to streptavidin, avidin or anti-biotin antibody. This type of complex
can be detected directly by excitation of a fluorophore conjugated to streptavidin or indirectly
by using an enzyme conjugate that can produce an insoluble colored precipitate.
The following components were added to an RNase-free microfuge tube.
Template DNA
1 µg of cDNA
Distilled or deionised water
to final reaction volume of 40 µl
10x HY reaction buffer
4 µl
10x Biotin labeled ribonucleotides
4 µl
10x DTT
4 µl
10x RNase inhibitor mix
4 µl
20x T7 RNA polymerase
2 µl
50
Materials and Methods
The mixture was carefully mixed and collected at the bottom of the tube by a brief
centrifugation. The tube was immediately placed in a 37°C waterbath. The tube was tapped
gently every 30 – 40 min during the 4 – 5 hrs incubation. The end-product was stored at 20°C.
2.20 Cleaning up and quantifying in vitro transcription (IVT) products.
The IVT cleanup reaction is essential to remove unincorporated NTPs. Only one-half of the
IVT reaction was used for the cleanup. We employed RNeasy spin columns from QIAGEN
as preferred method for IVT cleanup. The pure, eluted IVT product was quantitated using
spectrophotometric analysis. For quantification of cRNA using total RNA as starting
material, an adjusted cRNA yield was calculated to reflect carryover of unlabeled total RNA.
Using an estimated 100% carryover, the following formula was used to determine adjusted
cRNA yield.
Adjusted cRNA yield = RNAm – (total RNAi) (y)
RNAm = amount of cRNA measured after IVT (µg)
Total RNAi = starting amount of total RNA (µg)
y = fraction of cDNA reaction used in IVT
The minimum concentration of purified cRNA must be 0.6 µg/µl before the cRNA can be
fragmented for target preparation.
2.21 Fragmentation of cRNA for target preparation
The adjusted cRNA concentration was used in this reaction. The following components were
added to initiate the fragmentation reaction.
51
Materials and Methods
20 µg cRNA
1 – 32 µl
5x fragmentation buffer
8 µl
RNase-free water
to 40 µl
The final concentration of RNA in the fragmentation mixture can range from 0.5 µg/µl to 2
µg/µl. The fragmentation buffer has been optimized to breakdown full length cRNA to 35 –
200 bases fragments by metal-induced hydrolysis. The fragmentation mixture was incubated
at 94°C for 35 min. Reaction was terminated on ice. The undiluted, fragmented sample RNA
was kept at -20°C.
2.22 Eukaryotic target hybridization
In a hybridization cocktail for a single probe array, the following components were added:
Fragmented cRNA
15 µg
Control oligonucleotide B2 (3 nM)
5 µl
20X Eukaryotic Hybridization Controls
15 µl
Herring Sperm DNA (10 mg/ml)
3 µl
Acetylated BSA (50 mg/ml)
3 µl
2X hybridization buffer
150 µl
H2O
to final volume of 300 µl
The frozen stock of 20X GeneChip® Eukaryotic Hybridization Control cocktail was heated at
65°C for 5 min to completely resuspend the cRNA before aliquoting. It contains bioB, bioC,
bioD and cre prokaryotic enzymes. BioB, BioC and BioD are genes of the biotin synthesis
pathway from the bacteria E.coli. cre is the recombinase gene from P1 bacteriophage. These
genes can be labeled and could served as hybridization controls when mixed with labeled
52
Materials and Methods
eukaryotic cRNA samples. The antisense strand of B. subtilis RNA controls are used for
BioB, BioC and BioD genes. These controls can be spiked into samples during mRNA
preparation to monitor the efficiency of target preparation, hybridization, washing and
staining. The probe array was equilibrated to room temperature immediately before use. The
hybridization cocktail was heated to 99°C for 5 min in a heat block.
The probe array and a test array that were filled with 200 µl 1X hybridization buffer were
incubated at 45°C for 10 min with rotation. The hybridization cocktail that has been heated at
99°C was transferred to a 45°C heat block or 5 min. The cocktail was spun at a maximum
speed in a microcentrifuge for 5 min to remove any insoluble material from the hybridization
mixture. The buffer solution was removed from the probe array cartridge and filled with 250
µl of the hybridization cocktail avoiding any insoluble matter in the solution at the bottom of
the tube. Probe array was placed in a rotisserie box in a 45°C oven. The
probe array was rotated at 60 rpm for 16hrs.
2.23 Washing, staining and scanning
After 16 hrs of hybridization, the hybridization cocktail was removed from the probe array
and kept in a centrifuge tube. The probe array was filled with 200 µl of non-stringent wash
buffer. We employed an antibody amplification wash and stain procedure. Streptavidin
phycoerythrin (SAPE) staining reagent was prepared. SAPE was stored in the dark at 4°C. It
should not be frozen. The tube was tapped to mix well before preparation of any stain
solution. The SAPE stain solution should be prepared immediately before use.
53
Materials and Methods
For each probe array to be stained, the following components were combined in a
microcentrifuge tube:
2X MES stain buffer
600 µl
50 mg/ml acetylated BSA
48 µl
1 mg/ml SAPE
12 µl
distilled water
540 µl
The SAPE solution is mixed well and divided into two aliquots of 600 µl each to be used for
first and third staining respectively. An antibody mixture was prepared with the following
reagents.
2X MES Stain buffer
300.0 µl
50 mg/ml acetylated BSA
24.0 µl
10 mg/ml normal goat IgG
6.0 µl
0.5 mg/ml biotinylated antibody
3.6 µl
distilled water
266.4 µl
A fluidics protocol was used to test the array chip and test chip. 80 µl of hybridization buffer
was added into the test chip and the following steps were used to test the quality of reagents.
Post Hyb Wash #1
10 cycles of 2 mixes/cycle with wash buffer A at 25°C
Post Hyb Wash #2
8 cycles of 15 mixes/cycle with wash buffer B at 50°C
Stain
The test probe was stained for 10 minutes in SAPE solution at 25°C
Post stain wash
10 cycles of 4 mixes/cycle with wash buffer A at 30°C
2nd stain
The probe array was stained for 10 min in the antibody solution at 25°C
3rd stain
The probe array was stained for 10 min in SAPE solution at 25°C
Final Wash
15 cycles of 4 mixes/ cycle with wash buffer A at 35°C.
54
Materials and Methods
Temperature was held at 25°C.
At the end of this wash, 80 µl of hybridization buffer was removed from the test chip and
mixed back into the rest of 220 µl hybridization buffer. The 300 µl of hybridization buffer
was injected into the probe array chip and the following steps were performed in the fluidics
station.
Post Hyb Wash #1
10 cycles of 2 mixes/cycle with wash buffer A at 25°C
Post Hyb Wash #2
4 cycles of 15 mixes/cycle with wash buffer B at 50°C
Stain
The probe array was stained for 10 minutes in SAPE solution at 25°C
Post stain wash
10 cycles of 4 mixes/cycle with wash buffer A at 25°C
2nd stain
The probe array was stained for 10 min in antibody solution at 25°C
3rd stain
The probe array was stained for 10 min in SAPE solution at 25°C
Final wash
15 cycles of 4 mixes/cycle with wash buffer A at 30°C. The final
holding temperature is 25°C.
The above fluidics protocol was selected and ran using the GeneChip® Fluidics Station 400.
At the end of the wash protocol, the probe array was scanned by Affymetrix® Microarray
Suite. The laser was warmed up prior to scanning by turning the laser at least 15 min before
use. Probe array that was stored at 4°C had to be warmed up to room temperature before
scanning. After scanning the probe array, the resulting image data created was stored on the
hard drive of the GeneChip® Analysis Suite/ Microarray Suite workstation as a .dat file with
the name of the scanned experiment.
55
Materials and Methods
2.24 Microarray data analysis
In the first step of the analysis, a grid was automatically placed over the .dat file demarcating
each probe cell. One of the probe array library files, the .cif file was used by Microarray Suite
to determine the appropriate grid size used. The alignment of the grid was confirmed by
zooming in on each of the four corners and on the center of the image. If the grid was not
aligned properly, the alignment was adjusted by placing the cursor on an outside edge or
corner of the grid. The cursor image will be changed to a small double-headed arrow. The
grid will then be adjusted using the arrow keys on the keyboard or by clicking and dragging
the borders with a mouse.
Affymetrix® Microarray Suite was employed to analyze hybridization intensity data from
GeneChip® expression probe arrays. This software calculates a set of metrics that describe
probe set performance. The gene expression levels from treated and untreated samples were
compared using a comparison expression analysis that is available in the software to calculate
a set of comparison metrics. Some of the metrics which were used by a decision matrix to
determine a Difference Call for each transcript: Increased (I), Decreased (D), Marginally
Increased (MI), Marginally Decreased (MD) or No change (NC). In our microarray
experiment, we only took into consideration I and D transcript level call for further analysis.
A “fold change” calculation was also computed to indicate the relative change of each
transcript represented on the probe array. Only those set of genes having 4 fold change in
their expression level were included for further functional annotation. The detailed steps for
running the software is available on the Microarray Suite User’s Guide (Affymetrix, Santa
Clara, USA).
56
Materials and Methods
2.25 Isolation of total RNA
Total RNA was isolated by a single-step method using TRIZOL® (Invitrogen). The total
RNA was treated with RNase-free DNase I at 37°C for 20 min and kept at -70°C until further
use. The concentration and purity of RNA was then estimated spectrophotometrically at 260
nm and 280 nm. The intregrity of RNA was analysed using 1% denaturing formaldehyde gel
elelectrophoresis.
2.26 RNA gel electrophoresis
Denaturing agarose-formaldehyde gel electrophoresis was used for the size fractionation of
RNA (Lehrach et al., 1977). Container or gel tank in contact with RNA samples were swiped
in 70% ethanol. A 1% (w/v) agarose-formaldehyde was prepared by melting 0.5 g of agarose
in 43.5 ml of autoclaved water and cooled to 60°C. 5 ml of 10x MOPS, 1 ml of 37%
formaldehyde and 1 µl of 10 mg/ml of ethidium bromide were added and casting of the gel
was carried out in fume hood. RNA sample was prepared as follows:
RNA (0.5 µg – 10 µg)
4.5 µl
10x MOPS buffer
2.0 µl
Formaldehyde (pH>4.0, 37%)
3.5 µl
Deionized formamide
10.0 µl
RNA loading buffer
3.5 µl
The sample was mixed and heated to 60°C for 10 min prior to loading and electrophoresis
was carried out in 1x MOPS buffer. RNA bands were visualized under UV-light after
electrophoresis. The image was recorded, captured and saved using a gel documentation
system (Bio-Rad, USA).
57
Materials and Methods
2.27 Real-time quantitative PCR
Gene specific PCR primers were designed using the PrimerExpress 2.0 (Applied Biosystems,
USA) according to their corresponding mRNA sequences. The primers were designed to
yield amplicons of less than 100 base pairs and synthesized by Alpha DNA USA). 18S
ribosomal RNA was used as an internal calibrator. The primer sequences for the ten genes
examined were listed in Table 2.1.
Gene
Tm
Amplicon Size
58
60
70 bp
Rev: 5' CCCTCTCGGAAGCATCCATA 3'
Amphiregulin
Fwd: 5' ACTCGGCTCAGGCCATTATG 3'
Rev: 5' ATGGTTCACGCTTCCCAGAGTA 3'
62
66
68 bp
Annexin A2
Fwd: 5' GTGCATATGGGTCTGTCAAAGC 3'
Rev: 5' TGGCTGTTTCAATGTTCAAAGC 3'
66
62
72 bp
Calponin 3
Fwd: 5' ACATTACAGCCGGTGGACAAC 3'
Rev: 5' TCCTTTCTGGGAAGCAACTTTG 3'
64
64
72 bp
Cysteine rich protein 1
Fwd: 5' CCAAGTGCAACAAGGAGGTGTA 3'
Rev: 5' CCCACATTTCTCGCACTTCA 3'
66
60
92bp
Fibroblast growth factor 2
Fwd: 5' AGCGACCCTCACATCAAGCT 3'
Rev: 5' CGGTTAGCACACACTCCTTTGATA 3
62
70
77 bp
GABA(A) alpha 5 receptor
Fwd: 5' CCCCAGACACGTTCTTCCA 3'
Rev: 5' TTGTTGGGCGTGGTCATGT 3'
60
58
64 bp
Galanin
Fwd: 5' GAACAGCGCGGGCTACCT 3'
Rev: 5' GGCCATTCTTGTCGCTGAA 3'
60
58
71 bp
KRIT2
Fwd: 5' GGAATGCGTCTCTCTCAAGAAAC 3'
Rev: 5' TGGCCAGTCACGAACATGTT 3'
68
60
117 bp
Neuregulin 1
Fwd: 5' TGTGCAAGTGCCCAAATGAG 3'
Rev: 5' ACTGTAGAAGCTGGCCATTACGT 3'
60
68
68 bp
Aldehyde dehydrogenase
Primers sequences
Fwd : 5' GCAGTGAAGGCCGCAAGA 3'
Table 2.1: Primer sequences used for real-time PCR. Primers were designed using
PrimerExpress 2.0 software (Applied Biosystems, USA).
A two-step reaction was used in this study. For each reverse transcription (RT) reaction, the
following reagents composition was used.
10x PCR Buffer
1.0 µl
MgCl2 (25 mM)
2.2 µl
58
dNTPs (25 mM each of dATP, dCTP, dGTP and dUTP)
Materials and Methods
2.0 µl
Random hexamers (224 µM)
0.6 µl
RNase inhibitor (20 U/λ)
0.2 µl
Reverse transcriptase (50 U/λ)
0.3 µl
DEPC H2O
2.76 µl
A total of 12 µl of RT reaction mixture and 3 µl of total RNA (100 ng/µl) was used to carried out
RT reaction. RT reaction was carried out as: 25°C/ 10”, 37°C/ 60” and 95°C/ 5”.
PCR was then performed on the RT products. For each PCR reaction, the following reagents
were used:
SYBR® green mix
25 µl
Sense primer (100 nM)
x µl
Anti-sense primer (100 nM)
y µl
Sterile water
14.8 µl
The PCR amplification was then performed at 50°C for 2 min and 95°C for 10 min per cycle
for 40 cycles with each cycle at 94°C for 15 sec and 60°C for 1 min. All reactions were
carried out in triplicate using the ABI Prism 7000 SDS (Applied Biosystems, USA).
2.28 Purification of Torpedo californica acetylcholine receptors
Preparation of nicotinic acetylcholine receptor from Torpedo californica was carried out
according to the procedure described by Ishikawa et al. (1977) with some modifications. 10 g
of frozen tissue was defrosted and cut into small pieces. The tissue was homogenized with 20
ml distilled water for 4 min with 3 interval time in a warring blender in ice-cold vessel. The
59
Materials and Methods
suspension was then again homogenized in IKA-Werk homogenizer (200 rpm) followed by
low speed centrifugation at 3000 x g for 10 min at 4°C. The supernatant was centrifuged at
10, 000 x g for 20 min at 4°C. The pellet was resuspended in 8 ml of Tris buffer and
homogenized with natural homogenizer. The homogenate was spun as above and the pellet
was resuspended in 5 ml of Tris buffer and centrifuged as above. The pellet then was
resuspended in Tris buffer to the concentration of 1 mg/ml, aliquoted in eppendorf tubes and
kept at -70°C. The whole procedure was done at 4°C.
2.29 Competitive binding assay
Competitive binding assay were performed using nAChR. Increasing concentrations of
unlabelled Bc-ntx4 was added to a single concentration of 5 nM
125
I-α-Bgt to study their
inhibitory effect on high affinity toxin binding. 2.5 µg of Torpedo membrane was incubated with
125
I-α-Bgt at room temperature for 1 hr in presence of increasing concentrations of competitors in
the total volume of 200 µl. Reaction was terminated on ice and the membranes were recovered by
centrifugation at 10, 000 x g for 5 min. Membranes were was washed with 1 ml of Tris buffer
supplemented with 0.1% bovine serum albumin (BSA) and dried before subjecting to radioactive
monitoring in a COBRA Auto Gamma Counter (Parkard Instrument Company, USA).
2.30 Statistical analysis
Statistical analyses were performed using unpaired Student’s t test and results were expressed
as mean ± S.E. p value of < 0.05 was considered statistically significant.
60
Materials and Methods
Section C List of chemicals used:
Chemical and solvents are all of analytical reagent grade obtained mainly through National
University Medical Institutes (NUMI) laboratory from the following suppliers:
Sigma Chemicals Co., St. Louis, MO, USA
Bovine
serum
albumin
(BSA),
formamide,
L-glutamine,
MOPS
(3,-(N-Morpholino)
propanesulfonic acid), DMEM basal medium, MES free acid monohydrate, MES sodium salt,
EDTA disodium salt (0.5 M solution), Tween-20.
Sigma-Aldrich
Goat IgG, anti-streptavidin antibody (goat), MTT
Gibco-BRL Gaitherssburg, MD, USA
Agarose, fetal bovine serum
Pharmacia
Bromophenol blue, RNase-free DNase I.
Merck
37% Formaldehdye
Boehringer Mannheim
Deoxyribonucleotides (dATP, dCTP, dGTP, dTTP, dUTP)
BDH
Ethidium bromide
Roche
SYBR® Green PCR Master Mix, TaqMan® RT Master Mix.
Invitrogen Life Technologies
Trizol® Reagant, Acetylated bovine serum albumin (BSA) solution (50 mg/ml)
61
Materials and Methods
Promega Corporation
Herring sperm DNA
Affymetrix, Santa Clara, CA, USA
Human genome gene chip U133 Plus 2.0 Array, GeneChip® Eukaryotic Hybridization Control
Kit (contains Control cRNA and Control oligo B2)
Molecular Probes
R-Phycoerythrin Streptavidin
Solutions and buffers
2.32 Reagents for PR-HPLC
2.32.1 Buffer A:
Trifluoroacetic acid (TFA, HPLC grade)
0.1% (v/v)
2.32.2 Buffer B:
Acetonitrile (HPLC grade)
80% (v/v)
Trifluoroacetic acid (TFA, HPLC grade)
0.1% (v/v)
2.33 Buffers for RNA clean-up
2.33.1 RLT buffer
Commercially available from QIAGEN. Contains guanidine thiocyanate. 10 µl of 14.3 M of
β-mercaptoethanol was added to 1 ml of RLT buffer. This mixture was found to be stable for
a month.
2.33.2 RPE buffer
Commercially available from QIAGEN.
2.34 Buffers for cRNA target hybridization
2.34.1 12X MES stock
62
Materials and Methods
MES free acid monohydrate
1.2 M
MES sodium salt
0.89 M
The reagents was mixed throughly and adjusted to final volume of 1000 ml of autoclaved
water. The solution (pH between 6.5 – 6.7) was filtered through a 0.2 µm filter. Solution was
stored at 4°C and shielded from light. The solution was discarded if colour turned yellowish.
2.34.2 2X hybridization buffer
12X MES Stock
0.1 M
5 M NaCl
1M
0.5 M EDTA
0.02 M
10% Tween 20
0.1% (v/v)
The final volume of buffer was adjusted to 50 ml of autoclaved water. It was stored at 4°C
and shielded from light.
2.35 Buffers for array processing, washing and staining
2.35.1 Non-stringent Wash Buffer (Wash Buffer A)
NaCl
0.9 M
NaH2PO4
0.06 M
EDTA
0.006 M
10% Tween-20
0.01 % (v/v)
The final buffer volume was adjusted to 1000 ml of water and was filtered through a 0.2 µm
filter and stored at 4°C.
2.35.2 Stringent Wash Buffer (Wash Buffer B)
12X MES Stock Buffer
0.1 M
NaCl
0.1 M
63
Materials and Methods
0.01 % (v/v)
Tween 20
The final buffer volume was adjusted to 1000 ml of water and filtered through a 0.2 µm filter
and stored at 4°C.
2.35.3 2X stain buffer
12X MES stock buffer
0.1 M
NaCl
1M
Tween 20
0.05 % (v/v)
The final volume of buffer was adjusted to 250 ml of water and filtered through a 0.2 µm
filter. Solution was stored at 4°C and shielded from light.
2.35.4 10 mg/ml Goat IgG Stock
The antibody stock was resuspended in 5 ml PBS and stored at 4°C.
2.35.5 20X SSPE buffer
NaCl
3M
NaH2PO4
0.2 M
EDTA
0.02 M
2.36 10X Phosphate-buffered saline (PBS)
NaCl
0.2 M
KCl
0.0027 M
Na2HPO4
0.0081 M
KH2PO4
0.00176 M
The above reagents were weighted and added into 800 ml distilled water. The pH was
adjusted to 7.4 with HCl and topped up with H2O until a final volume of 1000 ml. Stock PBS
solution was autoclaved and stored at room temperature.
64
Materials and Methods
2.37 Reagents for RNA gel electrophoresis
2.37.1 Ethidium bromide
Ethidium bromide was dissolved in water at 10 mg/ml concentration and stored in a light-tight
bottle. The final working concentration was 0.1 µg/ml.
2.37.2 10x MOPS running buffer
Morpholinopropanesulphonic acid (MOPS), pH 7
0.4 M
Sodium acetate
0.1 M
EDTA (pH 8)
0.01 M
The above solution was adjusted pH7 and topped up to a final volume of 1000 ml.
2.73.3 DEPC-treated water
Diethyl pyrocarbonate (DEPC)
0.2 % (v/v)
DEPC was added to 100 ml of deionised water and shaken vigorously to mix. It was incubated at
37°C for 1 hr and autoclaved to inactivate the DEPC. The solution was stored at room
temperature.
2.37.4 Deionized formamide
Formamide (550 ml) was added to 500 mg Dowex XG8 mixed bed resin for 1h, followed by
filtration through Whatman No.1 paper. Deionized formamide was stored at -20°C.
2.37.5 RNA loading buffer
Glycerol
50% (v/v)
EDTA (pH 8)
1 mM
65
Materials and Methods
Bromophenol blue
0.4% (w/v)
The above reagents were made up to 10 ml of water and stored at room temperature.
2.37.6 Formaldehdye
Formaldehdye (37% v/v) was prepared and stored in a light-tight container at room temperature.
2.38 Buffer for real-time PCR
2.38.1 10x PCR buffer
Tris-HCl (pH 8.3)
0.1 M
KCl
0.5 M
Gelatin
0.01% (v/v)
2.39 Buffer for receptor binding studies
2.39.1 Krebs-Ringer HEPES (KRH) buffer, pH 7.4
NaCl
0.128 M
KCl
0.005 M
CaCl2
0.0027 M
MgSO4
0.0012 M
Na2HPO4
0.001 M
Glucose
0.01 mM
HEPES
0.02 mM
66
Materials and Methods
RESULTS
Results
CHAPTER THREE
CONSTRUCTION OF PROTEIN DATABASE OF SNAKE VENOM NEUROTOXINS
3.1 Introduction
The most serious effects of snake envenomation are due to neurotoxins (NTXs). For example,
the most toxic component in cobra venom is the postsynaptic neurotoxins with an LD50 (i.v.)
of 0.05 – 0.2 µg/g of mouse (Tan, 1983). Toxin entries in public DNA and protein databases
represent only a tiny fraction of less than 1% of the estimated natural library (Tan et al., 2003).
Because of growing number of identified svNTX sequences, it is increasingly difficult to study
them by experimentation alone. Detailed bioinformatics analysis offers a convenient
methodology for efficient in silico preliminary analysis of possible function of new toxins. The
in silico approach can assist in designing experiments for functional characterization of the
newly identified svNTX sequences, particularly those identified as novel cDNAs.
A bioinformatics approach was utilized in the construction of a svNTXs database. This chapter
focuses on the compilation and consolidation of svNTXs data, which were stored in a newly
created, specialized database. The entries in svNTXdB have been classified and annotated to
enhance new knowledge discovery. It also had a set of tools including structure-function
prediction module. Our primary objectives were to facilitate information retrieval and data
analysis of enriched and unique compilation of svNTXs extracted from public databases and
published reports. Each database entry is then hyperlinked to an external database such as
NCBI, SwissProt or EMBL.
3.2 Data acquisition
As of April 2003, the svNTXs database contains 272 unique entries of neurotoxin. A
preliminary search using BioWare retrieval software with the keyword “Serpentes and
Neurotoxin” returned 821 entries on svNTXs. Serpentes keyword was included in the search as
it much more representative of all snake species and together with neurotoxin word in the
67
Results
search, entries that had both terms were extracted. These 821 entries were the initial data set.
Depending on their function, various databases carry a wide range of format. We merged the initial
dataset into a BioWare integrated format. Preliminary filtering was carried out to remove nonsnake neurotoxins and replicate entries. Ultimately, 272 entries remained in the dataset, which are
unique. This entry was referred to as the final data set. The breakdown of the each entry was listed
in Table 3.1. Four entries were found only in the published journal. These were listed in Table 3.2.
The majority of the entries in the final data set were mainly complete mature peptides with the
signal peptide. Out of 272 entries, 260 neurotoxins had the complete mature peptide and 12 were
incomplete. Only 99 neurotoxins out of 272 entries had the signal peptide sequence. The summary
of the protein identify was listed in Table 3.3.
Table 3.1: Summary of the 272 neurotoxin entries extracted from various sources.
68
Results
T o x in
F15
Sequence
H L L Q F N K M IK F E T R K N A V P F Y A F Y G C Y C G W G G Q R R P K
D A T D R C C F V H D C C Y G K L T K C N T K W D IY R Y S L K S G Y IT C
G K G T W C K E Q IC E C D R V A A E C L R R S L S T Y K N E Y M F Y P K
SRCRRPSETC
R e fe re n c e s
T o y a m a e t a l., 2 0 0 3
W - I II
FVCHNQ Q SSQ PPTTTNCSG G ENKCYKKQ W SDHRG K
I IE R G C G C P T V K K G IK L H C C T T E K C N N
S a m e jim a e t a l., 1 9 9 7
W - IV
L L C H N Q Q S S T S P T T T C C S G G E S K C Y K K R W P T H R G T IT E
R G C G C P T V K K G IE L H C C T T D Q C N L
S a m e jim a e t a l., 1 9 9 7
D IC L S T P D V K S K T C P P G
N ir th a n a n e t a l., 2 0 0 2
M ik a t o x in
Table 3.2: Sequence identity for the four unique toxins that are not found in public databases
but available in published journals.
Sequence
Complete
Incomplete
Missing
Total
Signal peptide
99
11
162
272
Mature
260
12
272
Table 3.3: Number of the complete, incomplete and missing sequences in the snake
venom neurotoxins in the final data set.
3.3 Database features
Snake NTX had five major search or extraction tools: BLAST search, Query svNTX, Structure
viewer, Download FASTA and Annotate svNTX. BLAST search allows user to perform a
sequence similarity search against snake NTX database (Altschul et al., 1997). Users can
specify output alignment result in either standard BLAST output or color-coded multiple
sequence alignment generated using Mview program (Brown et al., 1998). The Query svNTX
feature allows users to search entries in the database by simple keywords such as Naja,
bungarotoxin, or ion channel toxin. The search results are displayed in a tabular form as a list
containing accession numbers, species and toxin name. Each match in the result output is
hyperlinked to the full data record. Structure viewer feature (requires Internet Explorer 6.0 or
Netscape 4.75) consists of three-dimensional (3-D) structures of snake venom NTXs that are
extracted from PDB. This simple feature allows users to view available 3-D file using Chime
viewer or download PDB files of user-specified records. Annotate svNTX is a simple
prediction module tool which allows user to predict function of an unknown svNTX. Users can
69
Results
also choose to download FASTA formatted files of amino acid from the database under the
option Download FASTA.
Figure 3.1 shows the typical outlay of the svNTXs homepage. The five major extraction tools
were indicated on the left. Figure 3.2 shows the summary of the search result using keyword
search “Bungarotoxin”.
Figure 3.1: The user interface of the svNTXs database. Description of the five major tools
will be discussed in the text.
70
Results
Figure 3.2: An output feature of the search (partial) using the keyword “Bungarotoxin”.
The accession number labeled blue can be hyperlink to the database records.
71
Results
3.4 Descriptions of the svNTX individual record
Each NTX entry in the database is stored as an individual record. A representative record in
shown in Figure 3.3.
Figure 3.3: An example of an outlay of record available in the database. The individual
fields in each record will be explained in the preceding section. This entry shows fields that are
72
Results
found mostly in each entry. Complete entries have additional fields for structural and
functional information.
Each NTX entry in the database was given a unique accession number ‘DBACC’ in the form
of [D][six digit number], where the six digit number was the unique descriptor for each record.
‘Date’ field indicates the date when the entry was made.
‘Name’ field contains the name of the toxin used in published journal and in public databases.
The ‘Accession’ field provides hyperlinks to corresponding entries of the relevant databases
such as GenBank, Swiss-Prot or EMBL where the original sequence was deposited. The
organism source of each toxin and its taxonomical classification can be found in the ‘Source’
and ‘Species’ fields respectively.
Functional information based on experimental studies from various reports had been included
to enhance unique features of the database. These were represented by six fields known as
‘Action_site’,
‘Physiological_function’,
‘Critical_residues’,
‘Toxin_activity’,
‘Binding_affinity’ and ‘Miscellaneous’. The ‘Binding_affinity’ and ‘Critical_residues’
fields are unique in our database and not available in any public domain.
The ‘Action_site’ field indicates the site of action in which the NTX acts either
presynaptically or postsynaptically. A brief description of the NTX function is denoted by the
field ‘Physiological_function’. ‘Toxin_activity’ fields refer to the toxicity in terms of LD50
values. ‘Binding_affinity’ fields refer to the toxin-receptor interaction in terms of Kd or IC50
values. The critical amino acid residues involved in receptor binding and other miscellaneous
comments are provided in the ‘Critical Residues’ and ‘Miscellaneous’ fields respectively.
Each record has its corresponding reference(s) listed.
Structural features of the toxin, such as residues forming the disulfide bridges are described in
the field ‘Features’. Putative structural information derived by similarity to known structures
by amino acid alignment is indicated as ‘BY SIMILARITY’. A ‘svNTXdB’ sign followed by
‘BY SIMILARITY’ field implies data added by the authors which was not available in the
public databases. In addition any ‘Conflict’ field list down the amino acid discrepancies
between records of a particular NTX in different databases or published journal. The field
73
Results
‘Translation’ provides the amino acid sequence of the toxin. ‘Structure’ field contain
internal hyperlink to the PDB structure stored in the database, subjected to the availability of
the 3-D structures.
The ‘Reference’ field consists of a list of literature references, with author names and journal
titles. Some reference entries have the sign ‘svNTXdB’ which denotes publications that have
been added by us which are not listed in any public databases.
The svNTX database was largely compiled from public databases, which are unfortunately
prone to errors. Common errors that are encountered as summarized in Figure 3.4. For
example, entries Q9PSN6, Q91138 and Q91139 in the public database show the origin of these
toxins from Naja naja (Indian cobra) when the correct species should be Naja sputatrix
(Malayan cobra). Other common errors from these sources were different names for similar
toxin and different sequence for similar toxins. Although Swiss-Prot typically provides lethal
dose (LD50) values, one particular entry - long NTX OH-5 (P80965) - did not have this
information although its LD50 is available in the original paper. These errors were reduced by
checking database entries and cross-checking with original publications.
Figure 3.4: Summary of the types of errors encountered in the public databases.
74
Results
3.5 Classification of snake venom neurotoxins
svNTX data was analysed to form a classification system for prediction of their structure and
function. Properties such as cysteine pairing patterns, phylogenetic relationships, and detailed
functional and structural classification were considered in order to build a classification chart.
Initially, cysteine pairing pattern was analysed as this method has been shown to accurately
classify the members of scorpion toxin families (Tan et al., 2003). However, the result was
inconclusive as distinct structure-function relationship could not be observed for each
disulphide bridging pattern. Although 12 unique disulfide pairing patterns were observed, we
could not correlate any significant structure-function relationship because multiple functional
groups were observed in one individual disulphide group (Figure 3.5). For example,
fasciculins (acts presynaptically and inhibiting acetylcholinesterase) and short-chain
neurotoxins (acts postsynaptically and binds to nAChR) in cysteine pairing group 1 (Table 3.4)
share similar disulfide pairing pattern but exhibit very different pharmacological functions.
Hence, disulfide pairing pattern cannot be used as an indicator for structure-function prediction
in snake neurotoxins.
One of the more commonly used bioinformatics method for analyzing venom-toxin data is
phylogenetic analysis (see Tan et al., 2003). This method had been employed to determine
diversification of conotoxins and classification of scorpion and snake toxins (Tytgat et al.,
1999; Espiritu et al., 2001; Okuda et al., 2001; Tsai et al., 2002; Fry et al., 2003). However,
phylogenetic groups alone did not correlate well to functional groups of svNTXs. Therefore,
we decided to classify svNTXs by combining pharmacological function and phylogenetic
studies. Phylogeny was determined using maximum parsimony (MP) method to test whether a
direct structure-function prediction can be deduced. Members in a clade having a bootstrap
value of 90 are grouped together. Groups having 5 or more members will only be considered
for phylogenetic analysis. Detailed pharmacological functional data such as site of action and
molecular targets of the svNTXs was used based on annotations available in the database. We
combined the functional annotation with the MP method to define structure-function
classifications. The database currently contains data of 260 full length svNTXs that have welldefined pharmacological functions. 53 (20.4%) proteins belong to the presynaptic NTXs
group, 155 (59.6%) proteins belong to the postsynaptic NTXs, 3 (1.2%) have both pre and
75
Results
postsynaptic effect, and 49 (18.8%) have other functional properties. Those classified as
“other” may have either pre or postsynaptic effects but their exact function(s) are currently
unknown.
A
B
C1
C2
C3
C4
C5
C6
C7
C8
C
C1
C1 C2
C3 C4 C5
C6 C7 C8 C9 C10
D
C2 C3 C4 C5 C6 C7 C8 C9 C10
C1 C2 C3 C4 C5 C6 C7 C8 C9 C10 C11 C12
F
E
C1
C1 C2 C3 C4 C5 C6 C7 C8 C9 C10 C11 C12 C13 C14
C2
H
G
C1
C1 C2 C3 C4
C3
C5 C6 C7 C8 C9 C10 C11 C12 C13 C14
76
C2
C4
C5
C6
Results
I
C1
J
C2 C3 C4 C5 C6 C7 C8 C9 C10 C11 C12
C1 C2 C3
C4 C5
C6
L
K
C1 C2 C3 C4 C5
C6 C7 C8 C9 C10
C1 C2 C3 C4 C5 C6 C7 C8 C9 C10 C11C12C13C14C15C16
Figure 3.5: Unique disulfide pairing patterns of all the neurotoxins. The C represent the
cysteine residue in the mature peptide, followed by a number (n) indicating the nth cysteine in
the peptide.
77
Results
Gr oups
Ent r i es
A
O57327
BAC78199
P01403
P01432
P01415
Q9YGJ5
Q9W7J7
P25683
Q9W7K0
Q9DEQ3
0901189A
Q9W7J9
P25675
P25495
P10808
Q9PRJ7
P01428
P19959
P01424
Q9PRJ6
P83302
Q90VW1
P25494
P01427
P01431
P01420
Q9W717
Q8UW26
P01423
Q9PRJ3
AAB36087
A27580
CAB43097
P59276
P10458
S14003
Q9YGX0
P14613
P59275
P01430
P32879
O57326
Q9YHU9
AAM00185
Q9PRJ0
AAD40971
Q9YGW8
P82462
B
P01384
P25670
P15817
P80965
764177A
AAC83986
P01395
P01383
P01379
AAC83982
P34073
P25672
P14612
AAC83996
P07526
P01390
O12961
P25668
Q9W726
P01393
P01385
P01381
AAC83985
Q8UW29
C
CAA06886
P81783
P29182
P01401
Q9W7I3
Q9YGI1
Q9W796
P25680
Q9W7I4
P01400
P81782
Q9YGJ0
O12963
Q9YGI2
P29181
D
PSKFAU
AAB36096
Q8AXW2
Q8AXW0
Q8QFW3
P00619
P59018
Q9PTA7
E
P10116
P00608
Q8UUI0
Q9DF52
P00628
P20258
P00614
Q9PSN5
F
P00979
P00982
P00981
P00980
G
Q10755
P24027
I51381
P14420
H
P24335
I
P00626
J
AAC83988
K
Q9PTA1
L
P00616
P01421
P01425
P01414
P82464
P80495
Q8UW27
P10460
P01437
Q9W7K2
P19960
P01417
W-IV
P43445
P25493
N1NJ2P
P01433
P80548
P25492
P25676
Q9YGI0
P01438
P81030
Q9PUB7
Q9YGX1
S48648
P01435
P25681
Q91138
P19958
P01434
0901189B
P18328
Q9PRI1
P01412
P25497
P80494
P01429
P01426
Q9YGC4
Q9YGW9
Q9W7K1
Q9W7J6
Q9PWJ4
P34075
P83346
P01422
Q91139
AAB50805
Q9YGC7
Q9YHV0
P34076
Q9W727
671059A
Q9YGJ6
P14534
Q9DE57
P01416
Q9YGI8
P82849
Q9PRJ5
Q9PSN6
P01418
W-III
Q9YGC2
P80958
P25496
AAC83981
P01391
Q9W729
P25674
P01394
P15816
P82662
AAC83987
CAA72434
P01397
P01387
P25673
P01389
BAA32992
P01386
P25671
CAB51841
AAC83997
P15815
P01398
P01380
P01396
O42256
P58370
P25679
P15818
P29180
Q9YGH9
Q9YGI7
CAD18848
O42255
P01399
P29179
O93422
P82935
Q9PTA5
P17934
P00617
Q90251
Q9PU97
Q9PTA6
Q8QFW4
P00618
PSKF3U
P20259
Q8UUH8
P00610
P00609
P23026
P00629
Q8UUH9
Q8UUH7
Q02471
P11407
P14424
P14421
Q8JFG0
P13495
O12962
Q8UW28
O42257
Q9W7J5 AAC83990
P80156
P01382
P01388
P25667
AAC83995 Q9W797
Table 3.4: The distinct disulfide pairing pattern of mature svNTXs. Functional grouping
could not be observed using this method. For example, fasciculin (P01403) and short
neurotoxins in Group A share similar disulfide pairing pattern but different pharmacological
function. The entry in red is a partial sequence of a short neurotoxin.
78
Results
The classification trees of the full length svNTXs was depicted in Figure 3.6. This classification
system was broken into six layers. Layer 1 depicts the NTX site of action; layers 2 and 3 represent
functional and structural properties of NTXs respectively; layer 4 denotes the molecular target of
each svNTX, and layer 5 represents the subclassification of NTX based on phylogeny relationship.
The final layer represents group labels. Presynaptic NTXs exhibit at least eight different functions
(Figure 3.6, layer 2), and can be classified into five major groups: 1) NTXs that inhibit
acetylcholine release; 2) NTXs that facilitate acetylcholine release either by blocking voltage gated
potassium channel or some unknown mechanism; 3) NTXs that inhibit acetylcholinesterase and 4)
NTXs that block calcium channel; and 5) NTXs that exhibit multiple functions such as
myotoxicity, hemorraghic/hypotensive or bactericidal activity.
Group 1 neurotoxins had two distinct groups, those possessing inter disulfide (S-S) chains and
those without. The molecular target of these neurotoxins is the phospholipid membrane. The
group 1 of svNTXs formed six phylogenetic subgroups (Figure 3.6; layer 6). Toxins in groups
1-A1, 1-A2, 1B-3 and 1B-4 originate from the Elapidae snake family and groups 1B-1and 1B2 were from Viperidae family toxins.
Several interesting features can be observed from Figure 3.6. Phospholipase A2 KPA2
(Q9DF52) from Bungarus caeruleus and phospholipase A2 He (Q9PSN5) from Notechis
scutatus scutatus in group 1B-4 exhibit additional function such as anticoagulant or myotoxic
activities. However, three other members (P00628, P00629, P00616) from this group do not
show anticoagulant activity. This could suggest that, based on evidence from phylogenetic
analysis, these three svNTXs could possibly have anticoagulant/myotoxic activity that is yet to
be determined. One svNTX from group 1B-2 (F15; Toyama et al., 2003) and two svNTXs
from group 1B-3 (P00608 and P00610) also demonstrated additional functional properties
besides neurotoxicity (Figure 3.6; Table 3.5). Some of the functions were myotoxicity,
hemorrhagic and bactericidal activities. It may be possible that other neuoroxins members in
these groups possess multiple function(s) as well.
Groups 2A1-3 of presynaptic neurotoxins from Dendroaspis facilitate acetylcholine release
and block voltage-gated potassium channel (Harvey and Anderson, 1991). Group 2B was
found to be a unique group – it is the only three-finger neurotoxin reported to date to act
79
Results
presynaptically to facilitate acetylcholine release (Kuhn et al., 2000). Group 3 consisted of
presynaptic neurotoxins that inhibit acetylcholinesterase activity. Acetylcholinesterase-type
NTXs had so far been isolated from African mambas (Dendroaspis) and are cross-linked by
four disulfide bonds.
Group 4 consisted of only one member, lethal peptide I (P24335) which contained only 1
disulfide bond and found to block calcium channels at the presynaptic membrane (Aiken,
1992). Group 5 svNTXs had multiple functions in vivo such as myotoxicity, anticoagulant,
hemorraghic/ hypotensive or bactericidal effects.
In contrast, postsynaptic svNTXs fall into two major groups based on their interaction with
either nicotinic acetylcholine receptor (nAChR) or L-type calcium channels. The postsynaptic
neurotoxins that interact with nAChR were subdivided into five subgroups such as short, long,
weak, kappa, and the unknown group of svNTXs (Groups 6 – 10). Groups 6 – 9 NTXs formed
a similar folding pattern of three loops adopting the three-stranded antiparallel α-pleated sheet
but group 10 svNTXs had a non-3 finger fold with one disulfide bond (P24335).
We employed a phylogenetic analysis on the short-chain neurotoxins and found six subgroups.
Suprisingly, all short-chain neurotoxins originate from the Elapidae family (Figure 3.6). From
extensive literature search, ten short-chain neurotoxins have been reported to interact with
muscle-type nAChR (Marchot et al., 1988; Weber and Changeux, 1974; Pillet et al., 1993;
Gong et al., 1999, 2000). One interesting neurotoxin was fasciatoxin from group 7B-1
(P14534). It is an unusual short-chain neurotoxin from Bungarus fasciatus (Liu et al., 1989).
This protein is 63 amino long but has a unique C-terminal tail of PSTPST not found in any
other short-chain neurotoxins. So far, short-chain neurotoxins are found to interact specifically
with muscle-type nAChRs. Critical residues that are involved in the molecular recognition of
toxin-receptor were studied elsewhere by individual chemical modifications (review by Endo
and Tamiya, 1991). Lys-27 and Lys-47 are critical for receptor binding (Hori and Tamiya,
1976; Ishikawa et al., 1977; Faure et al., 1983). Pillet et al. (1993) suggested that three
ionogenic residues Lys-27, Asp-31 and Arg-33 in erabutoxin a from L. semifasciata are
involved in direct interaction with AChR.
80
Results
Layer 1
Layer 2
Layer 3
Layer 4
Inter S-S chain
Layer 5
Phospholipid
Inhibits Ach release
No inter
S-S chain
Blocks
voltage
gated K+
channel
Facilitates
Ach
release
Presynaptic
membrane
Inhibits
acetylcholinesterase
Blocks Ca2+ channel
Phospholipid
Intra S-S chain
GRP 1A-1
2
GRP 1A-2
3
GRP 1B-1
9
GRP 1B-2
12
GRP 1B-3
5
GRP 1B-4
Kv1.1
3
GRP 2A-1
Kv1.2
2
GRP 2A-2
Kv1.6
1
GRP 2A-3
Intra S-S chain
1
GRP 2B
Intra S-S chain
2
GRP 3
Intra S-S chain
1
GRP 4
1
Myotoxic, anticoagulant ,
hemorraghic/hypotensive
and bactericidal activity
No inter
S-S chain
Phospholipid
GRP 5A-1
1
GRP 5A-2
3
GRP 5A-3
1
Muscle-type nAChR
α-7 neuronal
nAChR
long
Blocks
neuromuscular
transmission
4
unknown
short
unknown
kappa
unknown
Binds L-type
calcium channel
unknown
Presynaptic &
postsynaptic
membrane
unknown
α-3 neuronal nAChR
2
α-7 neuronal nAChR
1
unknown
weak
Hemorraghic/
hypotensive activity
Intra S-S chain
GRP 6A-1
GRP 6A-2
GRP 6B
3
Muscle-type nAChR
Postsynaptic
membrane
Layer 6
14
1
GRP 6C-1
42
GRP 6C-2
4
GRP 7A-1
2
GRP 7A-2
1
GRP 7A-3
3
GRP 7A-4
1
GRP 7B-1
75
GRP 7B-2
GRP 8A
GRP 8B
3
GRP 8C-1
3
GRP 8C-2
Muscle-type nAChR
3
α-7 Neuronal
nAChR
3
unknown
3
GRP 9C
Muscle-type nAChR
1
GRP 10
GRP 9A
GRP 9B
GRP 11
2
GRP 12
2
Phospholipid
No inter S-S chain
No inter S-S chain
1
2
GRP 13
GRP 14
GRP 15
49
Figure 3.6: Classification of snake NTX based on structure, function and phylogenetic information. Only full length
neurotoxins are included in this classification. Boxed number refers to the total number of neurotoxins sharing the same
physiological function. Layer 1 denotes the neurotoxin site of action; layers 2 and 3 represent functional and structural
properties of neurotoxin respectively; layer 4 refers the molecular target of each svNTX layer 5 represents the
subclassification of neurotoxin based on phylogenetic relationship. Layer 6 represents group labels.
81
Results
Long-chain neurotoxins from Elapidae family were subclassified into five subgroups. Several
members of long-chain neurotoxins interact with alpha-7 neuronal nAChR (Group 6B), in
contrast to short-chain neurotoxins. Phylogenetic analysis could not be performed on this
group because it has only three members at present. To date, five and three long-chain
neurotoxins were demonstrated experimentally to interact with muscle-type nAChR (Group
6A-1 and 6A-2; Figure 3.6) and alpha-7 neuronal nAChR (Group 6B-1) respectively (Ishikawa
et al., 1977; Chang et al., 1993; Servent et al. 1998). The presence of the fifth disulfide bond
in long-chain neurotoxins had been implicated in the interaction with neuronal nAChR
(Servent et al., 2000). Arg-33 was especially important for this binding specificity (Antil et al.,
1999). It would be of great interest if the important residues that interact with these two
receptors can be delineated. Forty five long neurotoxins were not experimentally determined
whether they interact with either muscle-type or neuronal nAChR. Phylogenetic analysis
indicated two main groups within this orphan group: groups 6C-1 and 6C-2 (Figure 3.7E).
svNTXs in group 7A-1 and 7A-2 did not have the consensus Lys-27, Asp-31 and Arg-33
residues which were implicated in receptor binding (Pillet et al., 1993). However, svNTXs in
groups 7A-3 and 7A-4 (except P01426) possessed conserved residues of Lys-27, Asp-31 and
Arg-33 for receptor interaction. Known kappa neurotoxins have 65-66 amino acid residues.
Their disulphide bonding pattern had not yet been determined.
One distinguishing functional property of kappa svNTXs was their ability to interact with α3
neuronal nAChR (Figure 3.6, Group 8A). So far, kappa svNTXs were found only in Bungarus
species and it was believed that they do not interact with muscle nAChR. In addition, the
specific receptor of six kappa neurotoxins remain unknown (Group 8C-1 and C2) and
phylogenetic analysis showed two distinct groupings.
Phylogenetic analysis was not performed on weak neurotoxins because of the small number of
toxins (Groups 9A-C). This group of toxins was found to interact weakly with muscle- and α7neuronal nicotinic acetylcholine receptor (Poh et al., 2002; Utkin et al., 2001). To date, our
database contains 86, 50, 8 and 6 full length short, long, kappa and weak NTXs, respectively.
Certain postsynaptic neurotoxins (Figure 3.6, group 11) interact with L-type calcium channel
(Strydom, 1977; Yasuda et al., 1994). Crotoxin (P24027) and phospholipase A2 HTe
82
Results
(Q9PSN5) in group 12 exerted their effect postsynaptically (Bon et al., 1979; Francis et al.,
1995). However, their specific postsynaptic actions are yet to be determined. Three toxins in
groups 13 and 14 exerted neurotoxic effect both pre- and postsynaptically (Francis et al., 1995;
Bon et al., 1979). Specific functions of toxins in group 15 are still unknown. They are
designated as neurotoxin homologs, neurotoxin-like proteins, or neurotoxin isoforms that are
still not well studied. The individual records of neurotoxin in each group in Figure 3.6 are
listed in Table 3.5 according to their GenBank Accession Number.
83
Results
Table 3.5: Summary of the individual entries based on GenBank Accession Number of
each member in each group as depicted in Figure 3.6. Field in red bold and underlined
indicates the presence of entry in more than one group. Fields in italic (F15, W-III and W-IV)
represents entries found in published journal but not in any public database.
84
Results
A
B
85
Results
C
D
86
Results
E
outgroup
P01383
Q9W7J5
P13495
Group 6C-1
Q42257
P25671
P25672
P25673
764177A
P25668
P01388
P01389
P01390
P25674
P01386
P07526
P01387
P01393
P01396
P25667
P01397
P01395
P01394
P25670
RAA32922
0901189A
0901189B
Q8UW29
Q8UW28
P01381
P01380
P34073
P01384
P01385
ACC83990
ACC83998
ACC83992
ACC83985
ACC83986
ACC83987
CAB51841
ACC83995
ACC83997
ACC83996
87
Group 6C-2
Results
F
88
Results
G
89
Results
H
Figure 3.7: Phylogenetic trees performed using parsimony analysis of all the mature svNTX.
Bootstrap values are the result of 100 replicates. Only bootstrap values of greater than 90 in the each
internal node are grouped as one group. The outgroup Bee venom PLA2 (Swiss-Prot A59055) and
Lynx1 (Swiss-Prot AAF16899) were utilised as outgroups for analysis of presynaptic and
postsynaptic NTXs subgroups respectively. Outgroup was used to give a better representation of the
evolutionary relationship. A and B: Presynaptic neurotoxins; C: Presynaptic neurotoxins with
multiple functions; D: long neurotoxins with known receptor binding; E: orphan long neurotoxins; F:
short neurotoxins with known receptor binding; G: orphan short neurotoxins with unknown receptor
functions and H: orphan kappa neurotoxins with unknown receptor functions.
90
Results
3.6 Receptor binding studies to test the in silico prediction tool in the database
A native Bc-ntx4 protein (B. candidus toxin with an unknown function) was tested using
competitive binding studies with 125I-α-Bgt in muscle (Torpedo) nAChR. The binding activity
studies of Bc-ntx4 on AChR isolated from T. californica showed weak affinity even in the
micromolar range (Figure 3.8). Using the ‘Annotate svNTX’ function in the database, Bc-ntx4
was predicted to be a weak neurotoxin. This further validates the proof-of-concept of the in
silico
prediction
tool.
Amino
acid
sequence
of
Bc-ntx4
is
LTCLICPEKYCQKVHTCRDEENLCVKRFYEGKRFGKKYPRGCAATCPEAKPHEIVECC
STDKCNK (AY611643).
10-10
10-9
10-8
Figure 3.8: Competitive binding studies of
10-7
125
105
I-α-Bgt and native Bc-ntx4 to nAChR of
Torpedo receptor. Data are average values of duplicate determinations.
91
10-6
Results
Figure 3.9: Output of the annotation tool in svNTXs. Bc-ntx4, a protein of unknown
function was purified from RP-HPLC and annotate in silico using Annotate svNTX in the
database. The output result returns a putative function of weak neurotoxin – like activity. This
agrees with our experimental result in Figure 3.8 which found Bc-ntx4 to be a weak
neurotoxin.
92
Results
3.7 Conclusion
Snake NTXs are a large family of active peptide with considerable sequence homology, but
with different biological properties. Sequence, functional and structural information on snake
venom neurotoxins (svNTXs) are scattered across multiple sources such as journals, books,
and public databases, with very limited functional annotation. A svNTXs database was created
at http://sdmc.i2r.a-star.edu.sg/Templar/DB/snake_neurotoxin. This database can also be
found under Swiss-Prot Toxin Annotation Project website (http://www.expasy.org/sprot/). At
present, 272 NTXs sequences are available in the database and each sequence contains fully
annotated function and literature references. An annotation tool to aid functional prediction of
newly identified NTXs as an additional resource for toxinologists. In addition, a classification
system based on structure-function and phylogenetic relationship derived from the 272 NTXs
was also developed to allow better prediction tools.
93
Results
CHAPTER FOUR
SCREENING FOR POTENTIAL THERAPUETIC AGENTS USING
TOXICOGENOMIC APPROACH
4.1 Introduction
Scorpion venoms consist of biologically active proteins that contribute to the pathological
effects of the venom. Traditionally, venoms have been studied with the aim of understanding
the structure and function of individual toxins and their mechanism of action. The typical
method is to isolate the protein, followed by biochemical characterization and then
determination of its biological activity in vitro and in vivo.
Recently, proteomics studies on Buthidae venoms was employed to analyze toxic fractions
from the venom of Brazilian scorpion, Tityus serrulatus in order to shed light on the molecular
composition of this venom and to facilitate the search of pharmacologically active compounds
(Pimenta et al., 2001). Using this method, the researchers are able to detect 380 different
components present in the scorpion venom. These studies clearly indicate that scorpion venom
represents a wide variety of proteins. Thus, scorpion venoms may be considered to be a
naturally occurring ‘library’ of biologically active components that could be potentially
exploited for drug discovery. To date, Buthus martensi Karsch (BmK) contains more than 70
different peptides, toxins or homologues (Goudet et al., 2002). These toxins have been found
to act on Na+, K+ and Cl- channels.
Technological advances in microarray applications now allow for a rapid, broadband analysis
of the functional effects of substances on expression of genes in cells and tissues. This
investigation is to assess the use of scorpion venoms as pharmaco-active libraries. The
hypothesis of the study is that at LD50 concentrations (where no overt phenotypic effect of the
venom on cells in tissue culture is observed), the venom will produce effects on the gene
expression profiles of the cells that may lead to some of the scorpion venom components as
pharmaceutically useful compounds.
94
Results
4.2 Purification of scorpion venom using gel filtration chromatography
Gel filtration separates proteins based on their molecular sizes. Protein mixtures are applied to
a gel filtration column containing a matrix of defined pore size, eluted with an aqueous buffer
and collected as individual fractions and analysed. Sephadex G50 was used to isolate proteins
in the range of 1.5 – 30kD. 20mg of lyophilized BmK crude venom was reconstituted in 2 ml
water and separated using gel filtration. Two fractions were obtained (Figure 4.1). The first
peak, BmKI consisted of high molecular weight proteins (10-20kD). The second peak, BmKII
containing low molecular weight proteins (3-7 kD) was subjected to further studies.
BmKII
1.8
1.6
BmKI
1.4
OD, 280nm
1.2
1
0.8
0.6
0.4
0.2
0
0
10
20
30
40
-0.2
Fraction, 1.5ml
Fig 4.1 Separation of BmK crude venom using gel filtration.
95
50
60
70
Results
4.3 MTT test
The cell survival test (Figure 4.2) showed that BmKII is not toxic to human neuroblastoma
cells even at a concentration of 500 µg/ml. At 35 µg/ml, the cell survival was at 99%. Hence,
35 µg/ml of BmKII was subsequently used in all experiments treated with this venom.
110
100
90
% cell survival
80
70
60
50
40
30
20
10
[BMKII] ug/ ml
0
0
100
200
300
400
500
600
concentration of BMKII, ug/ ml
Figure 4.2: Cell survival test of human neuroblastoma cells treated with various
concentration of BmKII.
96
Results
4.4 Total RNA isolation
Total RNA was isolated from human neuroblastoma cells treated with BmKII at a
concentration of 35 µg/ml and incubated for 18hrs. The intregrity of RNA was found to be
acceptable for further studies as they showed two distinct bands for 28S and 18S ribosomal
RNAs (Figure 4.3).
1
2
28S
18S
Figure 4.3: Total RNA isolation from human neuroblastoma cells treated with 35µg/ml
BmKII for 18hrs. Lane 1: untreated neuroblastoma cells; lane 2: 35µg/ml BmKII-treated
neuroblastoma cells.
4.5 Microarray analysis
Neuroblastoma cells were treated with partially purified BmKII (35 µg/ml) for 18 hrs. The
global gene expression changes were monitored by microarray genechip procedure. Results
from microarray analysis showed 875 genes that were four-fold upregulated upon treatment
with BmKII fraction. The 875 genes were analyzed in terms of their function using molecular
information available at http://www.affymetrix.com and published journals in NCBI database.
The summary of the function of 875 genes is listed in Table 4.1. From the microaray analysis,
ten genes were finally selected. They were found to be involved in neurogenesis and
angiogenesis pathways as well as other genes involved in metabolic pathway,
neurotransmission, development and signaling pathway. These ten genes were listed in Table
4.2. Figures 4.4 – 4.10 show the results of cluster analysis (Clustals 1-7) which contain the ten
selected genes.
97
Results
Actin/myosin binding protein
Angiogenesis
Anti coagulant
Anti inflammatory
Anti proliferative/ Tumor suppre.
Anti-apoptosis
Anti-differentiation
Anti-oxidant
Apoptosis
Cell adhesion
Cell cycle
Cell motility
Detoxification
Development
Differentiation
DNA repair/damage
Endocytosis/Exocytosis
Homeostasis
Immune response
Inflammation
Inhibitor of growth suppressor
Metabolic pathway
Neurogenesis
Protein biosynthesis
Protein degradation/turnover
RNA binding protein
RNA synthesis/processing
Signalling pathway
Solute carrier/ channel
Transcription factor
Transport/ Trafficking
Change in of gene expression
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
unchanged
+
+
+
+
unchanged
+
+
+
+
Table 4.1: Summary of the 875 genes function that was altered by treatment of BmKII in
human neuroblastoma cell. Functions of the genes were annotated using published literature
in NCBI. + and – sign indicate up- and down-regulation of genes respectively.
98
Results
Gene name
Fold change
Neurogenesis
Amphiregulin
Fibroblast growth factor
13.9
5
Angiogenesis
Krit-1
0.21
Others
Calponin 3
GABA (A) receptor alpha 5
Annexin A2
Galanin
Cysteine rich protein 1
Aldehyde dehydrogenase 1
Neuregulin 1
(sensory and motor-neuron derived isoform)
0.2
103
4.3
9.2
803
81.6
7.8
Table 4.2: List of the ten selected genes after microarray gene chip analysis. The gene
expression profiles of these ten genes were further studied using real-time PCR.
99
Results
12
EST/DB_XREF=gi:A44
PRO
tudor domain containing 1
plectin
zinc finger protein 237
phospholamban (PLN)
selectin E (SELE)
cDNA FLJ12153 fis clone MAMMA 100458
lipoprotein lipase (LPL)
gamma aminobutyric acid A receptor
small inducible cytokine subfamily B
tumor necrosis factor
deleted in azoospermia (DAZ4)
FEA=EST/DB_XREF=gi:10743006
hypothetical protein VPS33A
hypothetical protein MGC10731
FEA=EST/DB_XREF=gi:2402080
mRNA for PQBP-1b
EVH1 domain regulating protein
cellular growth-regulating protein
caldesmon II
FEA=ES/DB_XREF=gi:796550
cDNA:FLJ22585
caspase 8, apoptosis-related cysteine protese (CASP 8)
myosin, heavy polypeptide 13, skeletal muscle (MYH13)
hypothetical protein FLJ13166
FEA=EST/DB_XREF=gi:5856236
regulated in glioma (RIG)
cysteine rich motor neuron (CRIM1)
matrilin rich precursor (MATN3)creatine kinase, mitochondrial 1
PAC296K21 on chromosome X
cysteine knot superfamily 1, BMP antagonist 1
FEA=EST/DB_XREF=gi:10837689
protein kinase DYRK4
hypothetical protein FLJ23119
retinal outer segment membrane protein 1 (ROM1)
FN5 protein (FN5)
integrin beta 4 (ITGB4)
neuregulin 1 (NRG)
solute carrier family 22
calcineurin A1
sequence from clone RP11-55H7
mitogen activated protein kinase kinase 6 (MAP2K6)
FEA=EST/DB_XREF=gi:6228495
DKFZP5640962 clone
hepatocyte transcription factor mRNA
FEA=EST/DB_XREF=gi:13295908
FEA/EST/DB_XREF=gi:10811389
FLJ23029 clone
RP5-843L14 on chromosome 20
rTS beta protein
dentin phosphoryn
Figure 4.4: Cluster 1 genes identified using Genesis® software. Two genes, GABA (A)
receptor and neuregulin 1 in this cluster were selected for further studied. Lane 1: untreated
neuroblastoma cell; lane 2: neuroblastoma cell treated with 35 µg/ml BmK II for 18 hrs. Green
bar and red bar represent gene downregulation and upregulation respectively.
100
Results
12
ubiquitous TRP motif, Y isoform (UTY)
cDNA DKFZp586E121
hypothetical protein FLJ22477
clone FLB4941 PRO1292
lipocalin 2
insulin-like growth factor 1 (IGFBP-1)
hypothetical protein FLJ14146
FEA=EST/DB_EST=gi:11954150
cathepsin X precursor
paired immunoglobulin-like receptor alpha
junction plakoglobin (JUP)
cDNA FLJ22868
hypothetical protein FLJ20272
fibroblast growth factor 2 (FGF2)
FEA=EST/DB_XREF=gi:10809262
FK506 binding protein 1B (FKBP1B)
vaccinia related kinase 2 (VRK2)
cDNA FLJ11303
hypothetcial protein (RIF)
lengsin (LGS)
Figure 4.5: Cluster 2 genes identified using Genesis® software. Fibroblast growth factor,
FGF2 in this cluster which is involved in neurogenesis was further investigated. Please refer to
Figure 4.4 for legends.
101
Results
12
protein tyrosine phosphatase
FEA=EST/DB_XREF=gi:2824690
CD2-associated protein
clone HQD131
KIAA0941
FEA=EST/DB_XREF=est7
putative DNA dependent ATPase
cDNA DKFZp584F059
est501657122R1
hypothetical protein FLJ12178
SUMO-1 specific protease
clone 4484560
KIAA0042
FEA=EST/DB_XREF=gi:5364202
tryptophan 2,3-dioxygenase
serologically defined colon cancer antigen 1
human autonomously replicating sequence (ARS)
FEA=EST/DB_XREF=gi:10284299
Ran-binding protein
FEA=EST/DB_XREF=gi: 119428450153
est 02485841F1
KIAA1109 protein
FEA=EST/DB_XREF=gi:500781
FEA=EST/DB_XREF=gi:12803262
vav2 oncogene
KIAA0395 gene
estbc44bc8
helicase II
FEA=EST/DB_XREF=gi:2574859
esttz22b10
FEA=EST/DB-XREF=gi:8159499
Krit1
KIAA0270 gene
KIAA0372 gene product
clone RP11-15H23
clone HQD105
structural maintenance of chromosome (SMC) family member
W D-2 protein
FEA=EST/DB_XREF=gi:10797095
nucleobindin 2
FEA=EST/DB_XREF=gi:13338044
FEA=EST/DB_XREF=gi:5803553
FEA=EST/DB-XREF=gi:11016007
Notch homolog 3
FEA=EST/DB_XREF=gi:5885103
hypothetical protein FLJ23321
glycoprotein nmb
heparan sulfate sulfotransferase
cadherin 11
granule associated RNA binding protein
KIAA0203
FEA=EST/DB_XREF=gi:5850810
FEA=EST/DB_XREF=gi:5837414
tetratricopeptide repeat domain 3
ribonuclear protein subantigen subunit
unknown protein clone MOC:3860
zinc finger protein 195
FEA=EST/DB_XREF=gi: 5382740
FEA=EST/DB_XREF=gi:5390527
FEA=EST/DB_XREF=gi:1158223
cDNA FLJ202742
forkhead box F1
decorin variant c
membrane-associated nucleic acid binding protein
KIAA0914 protein
FEA=EST/DB_XREF=gi:3948440
FEA=EST/DB_XREF=gi:1940853
insulin-like growth factor 2 receptor
FEA=EST/DB_XREF=gi:1128208
KIAA0995 protein
cDNA clone FL23455
sequence from clone RP11-108L7
homoe box c10
filamin c
FEA=EST/DB_XREF=gi:1337022
myosin, heavy polypeptide 3
titin
Figure 4.6: Cluster 3 genes identified using Genesis® software. Krit-1 is a gene involved in
angiogenesis and its gene expression was further studied. Please refer to Figure 4.4 for legends.
102
Results
12
FEA=EST/DB_XREF=gi:6704806
aspartyl tRNA synthetase
FEA=EST/DB_XREF=gi:10744829
plasminogen activator
FEA=EST/DB_XREF=gi:5003552
neuritin
dystrophin associated glycoprotein
tissue factor pathway inhibitor beta
aldolase c
prostaglandin-endoperoxide synthase
dikkopf homolog 3
hypothetical portein FLJ10493
FEA=EST/DB_XREF=gi:10828334
MHC class I antigen
alpha-2 type IV antigen
dihydropyrimidine dehydrogenase
selenium binding protein 1
FEA=EST/DB_XREF=gi:2348877
FEA=EST/DB_XREF=gi:2789059
FEA=EST/DB_XREF=gi:5894201
FEA=EST/DB-_XREF=gi:2797331
unknown protein
W W -domian containing oxidoreductase (W W OX)
HLA class I heavy chain
Cw1 antigen
FEA=EST/DB_XREF=gi:847688
HLA-B mRNA
cDNA DKFZp4348044
B-cell RAG associated protein
FEA=EST/DB_XREF=gi:3500885
cDNA DKFZp434N0813
collagen type IV
FEA=EST/DB_XREF=gi:5036103
FEA=EST/DB_XREF=gi:110D9578
cDNA FL21183
proalpha 1 procollagen
FEA=EST/DB_XREF=gi:384900
collagen alpha 2 chain precursor
FEA=EST/DB_XREF=gi:2321327
lumican
FEA=EST/DB_XREF=gi:5868436
FEA=EST/DB-XREF=gi:11006648
selenoprotein P
sal-1 like protein
lipopolysaccharide-binding protein (LBP)
matrix metalloproteinase 2 (MMP2)
FEA=EST/DB_XREF=gi:970050
calponin 3
KIAA0970 protein
plelenomorphicadenoma gene-like
e-syn protooncogene
cNDAFL22535
FEA=EST/DB_XREF=gi:2042123
protein kinase c
latrophilin
transcription factor c-maf
FEA=EST/DB_XREF=gi:3232123
HSP0040 protein
FEA=EST/DB_XREF-gi:1685105
protocadherin gamma subfamily A
platelet derived growth factor C
chondroitin sulfate proteoglycan 2
est602020842F1
fibulin
FEA=EST/DB_XREF=gi:5143449
proteoglycan PGM
OB-cadherin-1
FEA=EST/DB_XREF=gi:11112418
Figure 4.7: Cluster 4 genes identified using Genesis® software. Calponin 3 is a gene
involved in angiogenesis and its gene expression was further studied. Please refer to Figure 4.4
for legends.
103
Results
12
cDNA DKFZ434H1419
cystineglutamate exhanger
FEA=EST/DB_XREF=gi:5769332
nucleosome assembly protein 1-like 2
cell cycle progression 8 protein
KIAA0843 protein
immunoglobulin heavy chain gene
CYR61
PRO02259
clone MGC:2854
cDNA FLJ20660
RNA binding motif protein 3
FLJ23353
angiogenic inducer, 61 CYR61
hypothetical protein FLJ11151
peptidylprolyl isomerase D
FEA=EST/DB_XREF=gi:3228954
hypothetical protein HSPCO31
FEA=EST/DB_XREF=gi:5233843
hypotheticla protein FLJ22551
lipocortin (LIP) 2 gene
hypothetical protein FLJ13840
ADO21 protein
lipopolysaccharide specific-repsonse-68 protein
FEA=EST/DB_XREF=gi:2875596
Cdc6-related protein
FEA=EST/DB_XREF=gi:2321394
pyruvate dehydrogenase kinase
casein kinase 1
v-crk avian sarcoma virus oncogene homolog (CRK)
CDC6 homolog
DMC1
hypothetical protein FLJ20224
regulator for ribosome resistance homolog
clone MGC:12262
ATPase
hypothetical protein FLJ14054
tyhymidine kinase 1
RNA-binding protein
potassium channel (TW K-1)
gamma-aminobutyric acid (GABA) A receptor
uroplakin 1B
pseudouridine synthase 1
FEA=EST/DB_XREF=gi:4068966
insulin receptor substrate-2 (IRS2)
tetraspa NET-6 protein
pregancy associated plasma protein 1
FEA=EST/DB_XREF=gi:10302396
transcription factor CP2
fibronectin leucine rich transmembrane protein 3 (FLRT3)
FEA=EST/DB_XREF=gi:5177504
BCG induced integral membrane protein
clone RP-4-76112
testin
metastatic-suppresor gene CC3
15-hydroxyprostaglandin dehydrogenase
prostaglandin E synthase
gene encoding homeobox related protein
FEA=EST/DB_XREF=gi:AL578310
hypothetical protein DKFZp586H0623
cell death regulator aven
cDNA DKFZp5860051
estrogen-responsive B box protein
cDNA: FLJ23564
fatty acid coenzyme A ligase
hypothetical proteinFLJ12525
secreted frizzle-related protein 1
FEA=EST/DB_XREF=gi:11006713
epiregulin
gro-beta
fatty acid hydroxylase
signal transducer mRNA
DEADH polypeptide
nephropontin
clone 747H23
mitochondrial ribosomal protein S12
ATP-binding cassette
FEA=EST/DB_XREF=gi:4685039
solute carrier family 3
interleukin 18
cytokeratin 8
eukaryotic translation initiation factor 5A
prgrammed cell death 5 gene
tumor protein D52-like 1
trp-met fusion oncogene
FEA=EST/DB_XREF=gi:10373121
phosphogluconate dehydrogenase (PGD)
FEA=EST/DB_XREF=gi:12677244
Ets-related transcription factor
FEA=EST/DB_XREF=gi:6576051
peroxisome proliferative activated receptor gene
SIH003 gene
aldehdye dehydrogenase 1
S100 calcium-binding protein A10
hypothetical protein PP5395
clone GS1-99HB
CD24
Kunitz-type protease inhibitor
retinoic acid induced 3
aspartate beta-hydroxylase (ASPH)
FEA=EST/DB_XREF=gi:2910111
cDNA FLJ20161
clone MGC:10332
gamma glutamyltransferase 1
mRNA for clone 9112
receptor tyrosine kinase-like orphan receptor 1
Figure 4.8: Cluster 5 genes identified using Genesis® software. Aldehdye dehydrogenase
was upregulated 81.6 fold and its gene expression profile was further investigated. Please refer
to Figure 4.4 for legends.
104
Results
12
asparagine synthetase
fibroblast growth factor receptor 2
decay accelerating factor for complement
N-isoform-exon 11
hypothetical portein PR00758
FEA=EST/DB_XREF=gi:1093055
growth arrets and DNA-damage-inducible
cDNA DKFZp701H087
single Ig LI-1R related molecule
FEA=EST/DB_XREF=gi:12900950
MAD homolog 6
aspartyl beta-hydroxylase 2.8 kb transcript
Tat-interacting protein (30kD)
junctate
spindle pole body protein
FEA=EST/DB_XREF=gi:1015897
KIAA0064 gene product
polyadenylate binding protein-interacting protein 1
apolipoprotein C-1
transcription factor 2 hepatic nucler factor
hypothetical portein FLJ20607
CD24 signal transducer
S100-calcium binding protein A4
carnonate anhydrase VIII
lymphotoxin beta receptor
FEA=EST/DB_XREF=gi:11252643
FEA=EST/DB_XREF=gi:2263220
S100 calcium binding portein P
RAB4
hypothetical protein FLJ20273
hypothetcial protein FLJ23235
rhodanase
hypothetical protein FLJ20130
gamma aminobutyric acid A receptor
solute carrier family 21
mitogen-activated protein kinase kinase kinase 8
keratin 7
cytochrome P450 (CYP4F11)
hypothetical protein FLJ12591
cytochrome P450 (CYP24)
glucosaminyl transferase 3
clone 018603 gene
cysteine-rich protein 1 (CRIP1)
carbohydrate sulfotransferase 6
cytochrome b-245
GRO1 oncogene
hypothetical protein FL20452
serine protease inhibitor
ATPase, Na+/K+ transporting
KIAA0748
secreted apoptosis related protein 2
1, 2 cyclic-inositol-phosphate phosphodiesterase
ferrodoxin reductase
cDNA FLJ21431
FEA=EST/DB_XREF=gi:2525497
FEA=EST/DB_XREF=gi:6903427
FEA=EST/DB_XREF=gi:13134301
hypothetical protein FLJ20495
Figure 4.9: Cluster 6 genes identified using Genesis® software. Cysteine-rich protein 1
has the highest gene upregulation when studied using microarray. Please refer to Figure 4.4 for
legends.
105
Results
12
Keratin 19
Keratin 10
FEA=EST/ DB_XREF=gi: 11130198
FEA=EST/ DB_XREF=gi: 3740962
aldo-keto reductase family 1 (AKR1B1)
Clone MGC: 5333
UDP-glucose dehydrogenase (UGHD)
aldo-keto reductase family 1 isoform
dihydrodiol dehydrogenase
aldehyde dehydrogenase 3 family
cytochrome b-5 reductase
chlordecone reductase homolog
cytochrome P450
insulin-like growth factor-binding protein 4
pseudochlordecone reductase
tumor necrosis factor superfamily
FEA=EST/ DB_XREF=gi:12673408
UDP glycosyltransferase 1 family
Hypothetical protein MGC5395
FEA=EST/ DB_XREF=gi: 1040569
Tumor antigen (L6)
cDNA: FLJ 22893
Sjorgens syndromesoleroderma autoantigen 1 (SSSCA1)
Ribonucleotide reductase M2 polypetide
IAP homolog C (MIHC)
Periplakin (PPL)
Annexin A2 (ANXA2)
ATP binding cassette, sub-family C
Thioredoxin reductase 1, slone MGC:1920
Hypothetical protein FLJ10430
Brain acid-soluble protein 1
CCAAT enhancer binding protein (CEBP)
MHC class I polypeptide-related sequence B (MICB)
LPS-induced TNF-alpha factor (PIG7)
Potassium intermediate small conductance calcium-activated channel
NAD+-dependent 15 hydroxyprostaglandin dehydrogenase (PDGH)
LIM protein
Hypothetical protein MGC5338
Kynureninase
Nuclear receptor subfamily O (NROB1)
Gravin
Galanin
Aldehyde dehyrogenase 3 (ALDH3)
Heat-shock 70kD protein 1A
Id-1h
diaphorase
inhibitor of DNA binding 3
FEA=EST/BD_XREF=gi:11769659
Leucine zipper, down-regulated in cancer 1
Midline 1
Solute carrier family 21
Phosphotidic acid phosphorylase type-2c
Glutamate-cycteine ligase
FEA=EST/BD-XREF/gi:4188641
Proteoglycan 1
Epithelial-specific transcription factor
Glutamate cysteine ligase
Crk-associated substrate related protein
Clone MGC:4419
FEA=EST/DB_XREF=gi:1383407
Tropomodulin 3
Serum inducible kinase
Small inducible cytokine subfamily A
FEA=EST/DB_XREF=gi:13545549
J domain containing protein 1 (JDP1)
Dual specificity phosphatase 1 (DUSP1)
Beta 1,4 glycosyltransferase
Nonspecific crossreacting antigen
RAB4
Protein tyrosine phosphotase
Hypothetical protein FLJ11149
Neuregulin 1 (NRDG), transcript variant 1 SMDF
Annexin A4
Proliferating cell nuclear antigen
Pirin
Tissue inhibitor of metalloproteinase 3 (TIMP3)
Secretory granule proteoglycan peptide core
Desmoplakin
FEA=EST/DB_XREF=gi: 12800058
Aldehyde dehydrogenase 3
Transcription factor 8
Insulin-like growth binding protein 6
KIAA0307 gene product
Solute carrier family 16
FEA=EST/DB_XREF=gi: 10739541
KIAA0869 protein
NIT protein 2
Clone 640 unknown
Hematological and neurological expressed 1
mRNA for hepatocyte nuclear factor-3
FEA=EST/DB_XREF=gi: 11591542
Fatty acid binding protein 5
Tat-interacting protein (30kD)
Hypothetical protein FLJ10847
Clone MGC: 12282
FEA=EST/DB_XREF=gi: 12335213
Syndecan 1
Thyroid autoantigen
FEA=EST/DB_XREF=gi:12933969
FEA=EST/DB_XREF=gi:10732039
FEA=EST/DB_XREF=gi:4435781
Hypothetical protein FLJ11798
Hypothetical protein clone 1010
FEA=EST/DB_XREF=gi: 1727858
Latexin protein (LXN)
Glutaminyl peptide cyclotransferase
Hypothetical protein FLJ11264
Fucosyltransferase 8
Fatty acid co-enzyme A ligase
40S ribosomal protein S27 isoform
dickkopf homolog 1 (DKK1)
aldo-keto reductase family 1
amphiregulin
Figure 4.10: Cluster 7 genes identified using Genesis
®
software. Three genes in this cluster,
annexin A2, galanin and amphiregulin were chosen for further studied. Please refer to Figure 4.4 for
legends.
106
Results
4.6 Quantitative Real-time PCR analysis of genes affecting neurogenesis and angiogenesis
Quantitative real-time PCR is one of the most powerful and sensitive gene analysis techniques.
Its versatility has made it one of the fastest growing tools in molecular biology. The easier way
to monitor fluorescence in quantitative PCR is to use a dye that emits light upon intercalation
in double stranded DNA (Wittwer et al., 1997). The intensity of the fluorescent signal is
directly proportional to the amount of dsDNA species in the reaction.
Denature
Anneal
Extend
Figure 4.11: Schematic representation of real-time PCR with the SYBR Green I dye.
SYBR Green I dye (black diamonds) becomes fluorescent (green diamonds) upon binding to
double-stranded DNA, providing a direct method for quantitating PCR products in real time.
Gray arrow head represent the forward primer.
SYBR® Green assay was chosen in this studies as it is cheaper and relatively easy to use than
TaqMan® probes. Ribosomal RNA present in the total RNA sample is used as an internal
standard. Optimum concentration of total RNA for real-time PCR detection was found to be
100ng. A non-template control in which sterile buffer replaced template RNA served as the
negative control. The amount of target was normalized to ribosomal RNA. The following
formula was used to calculate the level of gene expression:
107
Results
∆CTS = ∆CT (sample) – ∆CT 18S RNA
∆∆CTS = ∆CTS – ∆CTS (of respective control)
Relative gene expression = 2-∆∆CTS
In order to confirm the level of gene expression in the microarray study, real-time PCR was
performed on these ten genes using SYBR® Green assay. Approximately 100 ng of total RNA
was used for real-time PCR experiment. Krit1 and calponin gene expression were found to be
downregulated in both microarray and real-time PCR (Table 4.3). The other eight genes were
found to be upregulated in both microarray and real-time PCR.
In summary, the gene
expression changes in microarray experiment were positively confirmed using real-time PCR
(Table 4.3 and Figure 4.12).
Amphiregulin
Krit1
FGF2
Calponin 3
GABA(A) receptor
Annexin A2
Galanin
Cysteine rich protein
ALDH
Neuregulin
Microarray
14.00
-5.00
5.00
-6.80
104.00
4.30
9.20
803.00
81.00
7.80
Real-time
2.90
-22.00
1.10
-60.00
3.10
5.20
3.20
2.50
433.00
1.20
Real-time SD
0.40
0.38
0.23
1.43
0.25
0.02
0.13
0.26
0.13
0.17
Table 4.3: Changes in gene expression level as detected by microarray and real-time
PCR. SD represents standard deviation.
108
Results
900.00
Microarray
800.00
Real-time
700.00
600.00
Fold change
500.00
400.00
300.00
200.00
100.00
0.00
Amphiregulin
Krit1
FGF2
Calponin 3
GABA(A)
receptor
Annexin A2
Galanin
Cysteine rich
protein
ALDH
Neuregulin
-100.00
-200.00
Gene
Figure 4.12: Gene expression profiles of ten genes studied using microarray and realtime PCR. Real-time PCR was repeated twice and carried out in triplicates.
109
Results
4.7 Further purification of BmKII using reverse phase-high performance liquid
chromatography
After confirming the expression of the ten genes, BmKII was then subjected to RP-HPLC. The
RP-HPLC separation of any peptide or protein mixture is dependent upon the strength of the
hydrophobic interaction of each component in the mixture with the hydrophobic surface of the
column matrix and the elution strength of the organic solvent in the mobile phase. As the
concentration of the organic solvent increases, the elution of the polar species occurs first
followed by the elution of non-polar species. Peptide or protein mixtures are applied to a RP
column containing a chromatographic matrix with a defined hydrophobic character. The
absorbed peptides or proteins are eluted in order of least to most strongly bound molecules by
increasing the organic solvent concentration in the elution buffer, collected as individual
chromatographic fractions, and analysed separately. The main difference between hydrophobic
interaction and reverse phase matrices is the relative hydrophobicity of the matrix, with the
reverse phase matrix having greater hydrophobicity.
In this method, the stationary phase is hydrophobic (non-polar), while the mobile phase is
polar. The functional groups normally attached to the silica gel are the alkyl chains such as
C18 (octyl). Venom polypeptides and toxins have compact spatial structure which allows their
charge and hydrophobic character to be expressed mainly on the surface of the molecule.
Partially purified BmKII venom was successfully separated into discernable peaks by RPHPLC using the Jupiter C18 column. The column was equilibrated with 0.1% trifluoroacetic
acid (TFA). The absorbed protein was eluted with 0.1% TFA at a flow rate of 1 ml/min. A
linear gradient (30 – 50%) of 0.1% TFA in water (Buffer A) and 0.1% TFA in 80%
acetonitrile (Buffer B) was applied for 20 min. Several proteins peaks were obtained during
the gradient elution. Once the protein fractions are collected, they are dried by speed-vac,
resuspended in sterile water and re-dried by speed vac again. In this way, the BmKII fraction
was divided into nine small subfractions (Figure 4.13). Each subfraction was pooled and its
protein concentration determined using Bradford assay. The individual fraction was examined
by real-time PCR (Table 4.4 and Figure 4.14) for its effects on expressions of genes in
neuroblastoma cells.
110
Results
RP1
RP2
RP3
RP4
RP5
RP6
RP7
RP8
RP9
70
5000
4500
60
4000
3500
mAU
3000
40
2500
2000
30
1500
20
1000
500
10
0
31
37
42
47
53
-500
58
63
69
0
Fraction, ml/min
Figure 4.13: RP-HPLC of BmKII from scorpion venom. This profile shows the BmKII
fractions containing small molecular weight protein of 7kD.
124
0
25001.0
Results
4.7.5 Effects of BmKII subfractions RP5 and 6 on six selected genes
Fractions RP5.2, RP6.2, RP6.3 and RP6.4 were subjected to another round of RP-HPLC and
found to contain single peaks (Figures 4.19- 4.22). Hence, they were considered a relatively
pure sample for further characterization. The MALDI-TOF analysis confirms that all fractions
contain a major peak of about 7kD (Figures 4.23). Two concentrations of 10 ng/ml and 50
ng/ml of fractions RP5.2G1, RP6.2F4, RP6.3F4, RP6.3F5 and RP6.4F7 were tested on
neuroblastoma cells and the gene expression changes measured using real-time PCR. Figure
4.24 and Table 4.6 show the gene expression profile of Krit1, galanin, FGF2, amphiregulin,
neuregulin and GABA (A) treated with 10 ng/ml of the five fractions.
Krit1
Galanin
7
FGF2
Amphiregulin
6
Neuregulin
GABA(A)
Fold change
5
4
3
2
1
F7
6.4
RP
F5
6.3
RP
F4
6.3
RP
-1
F4
6.2
RP
G1
5.2
RP
0
Fraction
Figure 4.24: Real-time PCR assay of six genes using 10 ng/ml of subfractions RP5 and 6.
RP5.2G1
RP6.2F4
RP6.3F4
RP6.3F5
RP6.4F7
Krit1
Fold
SD
Galanin
Fold
SD
FGF2
Fold
SD
Amphiregulin
Fold
SD
Neuregulin
Fold
SD
GABA(A)
Fold
SD
1.08
1.70
1.87
1.69
1.46
0.04
0.17
0.18
0.42
0.31
0.40
1.63
0.21
0.77
0.42
0.02
0.38
0.27
0.53
0.31
0.10
0.14
0.27
0.42
0.33
0.10
0.14
0.27
0.42
0.33
4.40
5.38
3.50
6.13
4.57
0.09
0.22
0.17
0.42
0.32
0.08
0.15
0.19
0.42
0.31
0.08
0.15
0.19
0.42
0.31
0.45
0.73
0.53
0.67
0.60
0.01
0.14
0.20
0.43
0.30
Table 4.6: Relative level of gene expression of the six genes using 10 ng/ml subfractions of
fractions RP5 and 6.
125
Results
20
Krit1
18
Galanin
16
FGF2
14
Neuregulin
Amphiregulin
GABA(A)
Fold change
12
10
8
6
4
2
0
RP5.2G1
RP6.2F4
RP6.3F4
RP6.3F5
RP6.4F7
-2
Fraction
Figure 4.25: Real-time PCR assay of six genes using 50 ng/ml of subfractions RP5 and 6.
RP5.2G1
RP6.2F4
RP6.3F4
RP6.3F5
RP6.4F7
Krit1
Fold
SD
Galanin
Fold
SD
FGF2
Fold
SD
Amphiregulin
Fold
SD
Neuregulin
Fold
SD
GABA(A)
Fold
SD
1.19
2.20
8.00
1.00
1.70
0.29
0.14
0.23
0.21
0.44
17.55
1.44
4.10
0.80
1.80
0.10
0.17
0.11
0.10
0.42
2.60
0.77
4.30
0.90
2.10
0.11
1.21
0.14
0.05
0.41
2.10
1.10
4.20
0.76
1.70
0.12
0.11
0.15
0.08
0.42
2.44
0.10
3.38
0.85
1.50
0.10
0.12
0.10
0.05
0.42
5.80
3.90
11.20
3.72
5.21
0.11
0.17
0.19
0.13
0.43
Table 4.7: Relative level of gene expression of six genes using 50 ng/ml subfractions of
fractions RP5 and 6.
At low concentration of 10 ng/ml treatment, amphiregulin and krit1 genes are consistently
upregulated. Galanin, FGF2, neuregulin and GABA (A) genes showed similar pattern of gene
downregulation. However a higher concentration of 50 ng/ml treatment showed very varied
level of gene expressions. Galanin was the most highly upregulated gene in RP5.2G1 (Table
4.7 and Figure 4.25). Interestingly, GABA (A) receptor gene was found to be consistently
126
Results
upregulated across the five treatments. In RP6.3F5, all the genes except for GABA (A)
receptor were downregulated (Table 4.7 and Figure 4.25).
4.8 N-terminal sequencing of the selected five subfractions RP5 and 6
N-terminal sequencing was performed on fractions RP5.2G1, RP6.2F4, RP6.3F4, RP6.3F5 and
RP6.4F7 purified on RP-HPLC column was determined by automatic Edman degradation
using a Procise-HT (Model 494) pulsed liquid-phase Protein Sequencer attached to a 140C
PTH amino acid analyzer (Applied Biosystems, Inc., USA). The sample containing 1-5 nmol
of protein was dissolved in 200 µl of 0.1% TFA and 10 µl was used for sequence
determination.
The N-terminal sequence of the 5 fractions was determined up to 27 amino acid residues. The
results of the N-terminal sequencing is listed in Table 4.8.
Residue
number
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
RP5.2G1
7202 Da
V
R
D
G
F
I
A
D
P
K
Protein fraction
RP6.2F4
7016 Da
D
R
D
A
Y
I
A
D
P
E
N
A
V
Y
A
G
N
E
RP6.3F4
7031 Da
G
R
D
A
Y
I
A
D
S
E
N
T
Y
T
A
L
N
P
Y
G
N
D
L
T
RP6.3F5
7024 Da
G
R
D
A
Y
I
A
D
S
E
N
T
Y
T
A
K
N
P
Y
N
D
L
T
RP6.4F7
7028 Da
G
R
Y
I
A
D
S
E
Table 4.8: Results of the N-terminal sequencing of five fractions. Amino acid residues are
indicated by the standard one-letter abbreviations.
127
Results
Blast search on the five fractions showed highest percentage similarity to known neurotoxins
from Chinese scorpions (Table 4.9). Protein RP5.2G1 showed similarity to bukatoxin and
toxin BmKT precursor, both which are alpha toxins from B. martensi. Both toxins bind to the
receptor at site 3 of the sodium channel and specifically prolong the action potential by
blocking the sodium channel inactivation. The interaction of alpha-toxin to the receptor is
voltage dependent. RP6.2F4 has similar identity to two toxins, BmK-IV and BmKalphaTx13
both alpha-type neurotoxins. These toxins bind to sodium channels and inhibit the inactivation
of the activated channels, thereby blocking neuronal transmission. It is interesting that
fractions RP6.3F4 and 6.3F5 displayed high protein sequence similarity with makatoxin I.
Both toxins shared similar sequence except at position 21 where leucine is replaced with lysine
in RP6.3F5. Makatoxin I is also an alpha toxin, where it binds to sodium channel and prolong
the action potential by blocking the sodium channel inactivation. In addition, fraction RP6.4F7
displayed highest protein similarity to makatoxin I and BmKVIII. Both toxins belong to the
alpha toxin family.
Fraction
Toxin identity
Percentage
similarity
87%
87%
GenBank
Accession Number
P82815
Q95P69
RP5.2G1
Bukatoxin
Toxin BmKT precursor (BmK T) (Antitumor-analgesic peptide)
RP6.2F4
Alpha-like neurotoxin BmK-IV (BmK4) (BmK-M4)
Alpha-neurotoxin TX13 precursor (BmKalphaTx13)
78%
78%
P58328
Q9NJC8
RP6.3F4
Makatoxin I (MkTx I)
88%
P56569
RP6.3F5
Makatoxin I (MkTx I)
84%
P56569
RP6.4F7
Makatoxin I (MkTx I)
BmK-VIII (BmKVIII) (BmK8)
80%
80%
P56569
P54135
Table 4.9: Blast search of the five peptides.
4.9 Conclusion
Scorpion venom BmKII was found to alter the gene expression pattern in human
neuroblastoma cell. 875 genes were upregulated by four-fold in our preliminary microarray
study. Six genes involved in neurogenesis and angiogenesis pathways were selected for further
gene expression analysis using real-time PCR. Crude venom BmKII was further separated into
128
Results
nine individual fractions and fractions 5, 6 and 7 were found to modulate gene expression of
six of the genes. Fraction RP5.2 consistently caused highest gene upregulation in the six
chosen genes with RP6.3 causing downregulation in GABA (A) receptor. The major toxin in
the five subfractions was found to be alpha-toxins from Buthus martensi. Alpha-toxins have
been shown to bind to sodium channel to cause prolonged channel inactivation. This study has
demonstrated that alpha-toxin may be exploited to bring about neurogenesis and angiogenesis.
129
Discussion
DISCUSSION
130
Discussion
CHAPTER FIVE
DISCUSSION
Section A
5.1 Neurotoxin database
A specialized database has been constructed using bioinformatics and for structure-function
studies on snake venom neurotoxins. The toxins data that are scattered across all databases are
finally consolidated into a single repository. This cleaned and annotated data set can give a
better insight into data representation, and also into the structure-function relationships of the
svNTXs.
Currently, the svNTXs database contains 272 entries of neurotoxins. This number is expected
to increase exponentially with time. BioWare provides a platform for easy update of new data
from any public databases.
The objectives of building a svNTX database for improved data management and
comprehensive analysis of the data have been fulfilled in this study. The svNTX database is
largely complied from public databases which can be prone to errors. Any errors or mistakes in
the database have been reduced by cross checking with published journal. This database is
intended to aid in the molecular analysis snake venom neurotoxins by facilitating the selection
of important experiments and design of experimental protocols. Collected dataset of svNTXs
have allowed for better structure and functional classification. This information on the
structural and functional prediction had lead to a building of a prediction tool in the database.
We tested native Bc-ntx4 protein, a B. candidus toxin with unknown function, using competitive
binding studies with 125I-α-Bgt in both muscle (Torpedo) nAChR . The binding activity studies
of Bc-ntx4 on AChR isolated from T. californica showed weak binding (Figure 3.8). Using
the ‘Annotate svNTX’ function in the database, Bc-ntx4 was predicted to be weak neurotoxins.
This further validated our proof-of-concept of the in silico prediction tool.
130
Discussion
The second aim of this bioinformatics study was to classify svNTXs according to structure and
function relationship based on several approaches. The first approach was to study cysteine
pairing pattern in the svNTXs dataset. It revealed 12 interesting disulphide pairing pattern.
Information for disulfide bridge patterns were only found in 251 entries but were absent in 21
entries. Some of the cysteine pairing groups had one or two members which formed
insufficient representatives for classification, as these patterns could be due to sequencing
errors. In addition, fasciculins (that act presynaptically by inhibiting acetylcholinesterase) and
short svNTXs (that act postsynaptically and bind to nAChR) share similar disulfide pairing pattern
but exhibited very different functions (Table 3.3). Hence, disulfide pairing pattern cannot be used
as an indicator for structure-function prediction.
The phylogenetic relationships of the three finger snake venom toxins have been extensively
reviewed (Dufton 1984; Tamiya and Yagi, 1985; Dufton and Hider 1991; Fry et al., 2003).
Dufton (1984) had systematically examined 139 homologous short and long svNTXs and
cytotoxins from elapid snake venoms and grouped the toxins according to similarity. In spite
of this, no clear picture has emerged to connect the structure-function relationship based on
phylogeny. Phylogenetic studies provide for better understanding of any evolutionary pattern in
svNTXs. Protein parsimony analysis available in PHYLIP statistical software was used to classify
the svNTXs. 15 specific evolutionary groups were obtained and all the members shared similar
function. With the exception of several members, some entries in each group have multiple
functions. It could be due to the fact that additional functions in other members are yet to be found.
Interestingly, some neurotoxins exhibit multiple functions unlike toxins from scorpion venoms.
Some of the toxins that exhibit multiple functions are listed in Table 3.6.
The svNTXs database revealed several interesting features. A thorough systematic
characterization of svNTXs, to our knowledge has not been carried out. We fill this gap with
the construction of clear and concise classification table based on structure-function and
phylogeny relationship (Figure 3.6). This table can also be used to predict function of newly
discovered snake neurotoxin.
The svNTXs database had several special features which are sets it apart from other sequence
databases: (i) 3-D structure feature viewer (ii) functional annotation of individual entries, and
(iii) functional properties prediction tool.
131
Discussion
The field of toxinology is growing rapidly especially in terms of information provided by
cDNA and protein data in the public database. The availability of a specialized database allows
easier systematic data analysis and also enables incremental data analysis upon updating with
new data. This database and analysis system will serve as an example for bioinformatics
application in the studies of snake neurotoxins.
The present work has laid the foundation for a more detailed and more systematic studies of
svNTXs.
The prediction module available in our database can be used for functional
prediction of any novel neurotoxins cloned or characterized in the future. We envisage that
increasing number of svNTXs in the future will make bioinformatics an attractive tool to speed
up experimental research.
5.2 Conclusion and future studies
This section focused on a systematic bioinformatics approach towards building a database for
svNTX and their structure-function study. The database contains functional and structural
information on svNTX gathered from various sources. Each record in the database, originally
from the public databases, was enriched by extracting information from the literature and
incorporating this into the svNTX database. The publically-available database may facilitate
bioinformatics study of structure-function of svNTX.
In addition to the tools available in svNTX, BLAST search, Query svNTX, Structure viewer,
Download FASTA and Annotate svNTX, this database can be further enriched with tools that
allow dynamic phylogeny. Dynamic phylogeny will allow users to construct phylogeny trees
on the spot in the database Then, any new evolutionary relationship of novel sequences can be
checked with the existing sequences for structure-function prediction. As a proof of concept,
an experimental procedure was carried for an unknown snake neurotoxin. This toxin was
found to be a weak neurotoxin based on in vitro and in silico approaches.
132
Discussion
Section B
5.3 Screening of scorpion venom for therapeutic peptides
Scorpion venoms are a rich resource of bioactive peptides. Accumulated data have
demonstrated that scorpion neurotoxins affect the ion permeability of excitable cells by
specific interaction with calcium, potassium, sodium, and chloride ion channels (Catterall,
1980). Detailed knowledge about the biological properties of the venom of the Chinese
scorpion, Buthus martensi Karsch (BmK), has been limited to neurotoxic components present
in the venom of this species. BmK has been used in traditional Chinese medicine to treat
epilepsy, antihyperalgesic and reversal effects on circulation failure (Zhou et al., 1989; Wang
et al., 2000; Peng et al., 2002). Although many peptides have been identified in this venom, no
modulators of genes affecting biological processes such as angiogenesis and neurogenesis
have been identified.
The effect of BmKII fractions on cell viability was first tested using MTT assay. As shown in
Figure 4.2, the venom is non-toxic to cells. This is further supported by the evidence that the
scorpion venom showed a LD50 value of 2.4 mg/kg following intraperitonial injection and a
minimum lethal dose (MLD) of 0.074 mg/kg after intracerebroventricular injection in mice
(Goudet et al., 2002). From the MTT assay, cells were found to show a 95% cell viability at
35 µg/ml of BmKII.
Thus gene expression studies were carried out at 35 µg/ml BmKII using a human
neuroblastoma cell line. Ten interesting genes that are involved in angiogenesis, neurogenesis
and ion channel modulation have been identified. In order to confirm the microarray results,
real-time PCR was used and the results supported each other.
One hypothesis was that BmKII partially purified fractions might possibly contain modulators
of these ten genes. The BmKII fraction was further purified into nine different subfractions,
termed RP1 – 9 using RP-HPLC. Each fraction was tested individually and the expression of
genes were monitored using using real-time PCR. Fractions RP5, 6, and 7 caused the greatest
133
Discussion
change in gene expression of those ten putative genes. Hence, these fractions were used for
further studies.
From extensive literature search six genes: galanin, krit1, neuregulin, GABA(A) receptor α5
subunit, amphiregulin and FGF2, are involved in many biological function in vivo such as
nerve regeneration, angiogenesis, cell proliferation and survival. The summary of the six
genes functions are described as below.
5.3.1 Galanin
Neuropeptides are a heterogenous class of biologically active substances that modulate the
function of neuronal circuits. These neuropeptides co-localized with the classical
neurotransmitters and determine a unique immunochemical profile of distinct neuronal
populations throughout the brain (Mazarati et al., 2001). These neuropeptides are released
from neurons in response to stimulus entering the spinal cord, binds to specific receptors on
target neurons and modulate the action of classical neurotransmitter in either negative or
positive fashion (Drake et al., 1994; Boehm and Betz 1997).
Galanin is a 29 or 30 amino acids long neuroendocrine peptide, originally isolated by
Tatemoto et al. 1983 from porcine upper intestine. Galanin has been identified in many
species, including rodents and primates (Langel and Bartfai, 1998). It is not a member of any
known family of neuropeptides, despite repeated efforts to discover related peptides. This
protein binds to family of receptors that belong to a superfamily of G-protein coupled
receptors (Amiranoff et al., 1989; Consolo et al., 1991). To date, three different receptor
subtypes of galanin receptor (GALR) have been cloned (GalR1, GalR2 and GalR3) from
rodents and humans (Mazarati et al., 2001; Figure 5.1). Ligand and receptor interactions
modulates release of neurotransmitters, affects learning and memory feeding, nociception and
nerve regeneration (Vrontakis et al., 2002).
Galanin mRNA and protein are found in specific neuronal systems in the brain, spinal cord, as
well as peripheral nervous system (Zhang et al., 1993a & b, 1995; Merchenthaler et al, 1993).
The protein exerts a strong neuroendocrine effect by modulating the release gonadotrophins,
134
Discussion
insulin, prolactin, growth hormone, and somastostatin (Vrontakis et al., 1991; Bartfai et al.,
1993). It is a potent inhibitor of the release of a number of neurotransmitters and hormones,
including acetylcholine, dopamine, insulin and gastrin (Rossowski and Coy, 1989; Pramanik
and Ogren, 1992).
A distinctive feature of galanin gene expression is its dramatic upregulation by estrogens, after
nerve injury, in the basal forebrain of Alzheimer’s patients and during development (Vrontakis
et al., 1989; Villar et al., 1989; Crawley, 1993; Xu et al., 1996). The galanin mRNA and
protein levels also change in some acute and chronic inflammation especially the neurons
where galanin is strongly upregulated following inflammation (Ji et al., 1995).
In the preliminary screening of all the BmKII fractions, galanin gene was upregulated the most
in RP5.2G1. This may be proved useful as a potential agonist. Galanin agonists have been
used in preventing anoxic damage and have additional advantage over Ca2+ channel blockers.
An application of galanin receptor agonists would be useful in preventing oxidative damage
during open heart surgery.
Galanin receptor agonist can also be used to enhance the release of growth hormone without
effecting the diurnal rhythm of this process and therefore appear as attractive agents to
increase growth hormone secretion in humans (Bauer et al., 1986). Galanin receptors seem to
control prolactin release from pituitary adenomas and are considered as targets for endocrine
manipulation of these tumors.
In addition, galanin agonist although not a potent analgesic agent on its own, appears to
prolong the morphine analgesia four-to eightfold. This may contribute to the reduction of
morphine doses to manage chronic pain (Wiesenfeld-Hallin et al., 1990).
135
Discussion
Figure 5.1: Galanin receptors and their transduction mechanisms. The first galanin
receptor was cloned from human Bowes melanoma cells by Habert-Ortoli et al., (1994).
Thereafter, two more galanin receptors, GAL2 and 3 have been cloned and the signal
transduction pathways have been determined for all the three receptors. GAL1 and GAL3
receptors can induce hyperpolarization and inhibit adenylyl cyclase (AC). AC can be inhibited
via GAL2 receptors. In addition, GAL2 receptors can also stimulate phospholipase C (PLC),
resulting in mobilization of Ca2+ and activation of protein kinase C (PKC) via diacylglycerol
(DAG). There is also evidence for the activation of the mitogen-activated protein kinase
(MAPK) pathway via GAL1 and GAL2 receptors. Image adapted from Liu and Hokfelt, 2002.
5.3.2 Amphiregulin
Cell growth and differentiation are regulated in part by the specific interaction of secreted
growth factors and their membrane-bound receptors. Receptor-ligand interaction results in
activation of intracellular signals leading to specific cellular responses.
Epidermal growth factor (EGF) consists of two families of structurally related ligands: the
EGF-like growth factors and the neuregulins. The former family includes transforming
growth-factor alpha (TGFα), heparin-binding EGF (HB-EGF), betacellulin (BTC) and
epiregulin (ERP) and amphiregulin (AR) (Lee et al., 2003; Troyer and Lee, 2001). EGF family
136
Discussion
members transmit their growth-modulating signals by binding to and activating receptors with
intrinsic tyrosine kinase activity (Plowman et al., 1990).
Tyrosine kinase receptors (TRK) belong to the ERBB family that are essential for the normal
development and are prominent targets for dysregulation in cancer (Lee et al., 2003). The
ERBB-signalling network comprises four homologous receptor tyrosine kinase named ERBB1
– 4 after the prototype EGF receptor (EGFR)/ERBB1 (Troyer and Lee, 2001). The binding of
the polypeptide ligand results in receptor homo or heterodimerization with activation of the
intrinsic tyrosine kinase, leading to the transphosphorylation of ERBB cytoplasmic tails
(Troyer and Lee, 2001). The phosphorylated tyrosines then serve as docking sites for
cytoplasmic signaling proteins, resulting in the activation of various downstream pathways,
including Ras-MAP kinase cascade, phosphatidylinositol-3 kinase and Stat transcription
factors (Troyer and Lee, 2001). The overall downstream signaling events may be involved in
regulation of cell proliferation, migration, differentiation or survival depending on cell type
and context.
AR protein was first discovered in MCF-7 cells and was termed amphiregulin to reflect it
bifunctional activities: it inhibits the growth of many human tumor cells and stimulate
proliferation of normal fibroblasts (Shoyab et al., 1988; 1989). This protein is a heparinbinding glycoprotein and exists as monomer as of either 78 or 84 amino acids long. AR
primarily composed of basic, hydrophilic residues as with other growth factors (Plowman et
al., 1990). AR mRNA had been found in ovary, testis, breast, placenta, pancreas, heart, colon,
lungs and brain (Plowman et al., 1990; Falk et al., 2002).
Recently, AR has been demonstrated as a potent EGF in adult neural stem cell (Falk et al.
2002). Its mRNA is highly expressed in choroid plexus as well as in hippocampal cells thus
suggesting that AR may regulate neural stem cell proliferation and neurogenesis in the adult
brain (Falk et al., 2002). It is also highly expressed in vascular smooth muscle cells (Kato et
al., 2003). This mitogen can also upregulate its own mRNA as well as α-thrombin. In addition,
AR can also caused an approximate 30-fold increase in DNA synthesis (Kato et al., 2003).
137
Discussion
Figure 5.2: The ERBB signaling network. The four ERBB receptors are shown along with
their activating EGF family and tyrosine kinase activity. Amphiregulin and ERBB1/ EGFR are
required for ductal morphogenesis, but is unclear whether ERBB2 is involved in this process.
Abbreviations of the ligands are described in the text. Image obtained from Troyer and Lee,
2001.
5.3.2.1 Applications
As mentioned earlier, neural stem cell cultures could be initiated with AR as the only growth
factor, demonstrating that at last some neural stem cells are responsive to AR in vivo. Fraction
RP6.3F4 BmKII upregulated the expression of AR gene 4-fold in vitro. This could mean that
AR could be used as a potential therapy to replace lost neurons in neurological diseases, either
by expanding the stem cells in vitro to transplant cells or by stimulating the neurogenesis in
vivo (Falk et al., 2002).
138
Discussion
5.3.3 GABA (A) receptor α-5 subunit
Although the brain is undoubtedly the most complex organ, it is possible to distil the way it
works into two opposing forces; excitation and inhibition. Neurons using neurotransmitters
receptor system, are excited by neurotransmitter glutamate and inhibited by the
neurotransmitter γ-aminobutyric acid (GABA; Whiting, 1999). GABA (A) receptors are
ligand-gated ion channels that are the major modulators of the inhibitory tone throughout the
central nervous system (CNS). They are the site of action of a number of clinically important
drugs, including benzodiazepines (BZs), barbiturates, and anesthetics (Whiting, 2001). It is
generally accepted that this receptor is a pentamer with an integral protein chloride ion channel
formed by the second transmembrane domain of each of the five subunits (Sur et al., 1998).
Over- or under-activity of excitatory or inhibitory force has been proposed to play a role in a
variety of neurodegenerative conditions such as stroke, Parkinson’s disease, epilepsy,
schizophrenia and anxiety (Kemp and McKernan, 2002; Meldrum and Whiting, 2001). In the
late 1980s and 1990s, with the application of molecular biology approaches, it becomes clear
that GABA (A) receptor subtypes are formed by co-assembly from 16 subunits (α 1 – 6, β1 –
3, γ1 – 3, δ, ε, π, θ; Whiting, 2001). Information of the physiological role of the various
subunits was contributed by gene knock-out studies (Collinson et al., 2002).
The primary role of the GABA (A) receptors α5 subunit had remained undefined until gene
knock-out studies were performed. α5-containing receptors have a particularly restricted
distribution in the hippocampus (Sur et al., 1998, 1999), cortex and olfactory bulb (Rosahl et
al., 2003). This localization suggests the receptor subtype may have a role to play in learning
and memory (Collinson et al., 2002).
Mice lacking α5-subunit have been found to have no major abnormalities, have normal
lifespan and no spontaneous seizures (Collinson et al., 2002). However, these mice
demonstrated an improved performance in working memory task (Collinson et al., 2002). In
addition, another group which studied partial knock-out α5 mouse found that trace fear
conditioning was enhanced in these mutant mice (Crestani et al., 2002). Altogether, this data
suggest that the α-5 subunit of GABA (A) receptor has an important role in controlling
139
Discussion
learning and memory, which makes the α5-containing GABA (A) receptor an interesting novel
drug target for the development of cognition enhancers (Rosahl, 2003).
In our screening experiments, BmKII crude venom upregulate GABA (A) receptor α5 subunit
gene expression in both microarray and real time experiment. Fractions RP5.2G1, RP6.4F7,
RP6.3F4, RP6.3F5 and RP6.2F4 at 50 ng/ml were found to upregulate the receptor gene.
Fraction RP6.3F4 was found to upregulate the gene expression by 11.2-fold (Table 4.7). At
low concentration of 10ng/ml, the receptor gene expression was downregulated in all the five
fractions. It is interesting to note that GABA (A) receptor is a chloride channel. However, the
major toxin in these five fractions were found to be alpha-toxins which bind to sodium
channels. Therefore, we can actually perform binding activity of toxin-receptor in vitro.
5.3.4 Sensory and motor-neuron derived factor (neuregulin)
Neuregulins belong to the family of ERBB ligands. The neuregulins include numerous
isoforms derived from four independent genes by alternative splicing. One of the neuregulin
isoforms, a sensory and motor-neuron derived factor (SMDF) directly binds ERBB3 and
ERBB4. It has been suggested that mammalian differentiation is regulated by neuregulin
receptors (Troyer and Lee, 2001). SMDF is a neural-specific gene and is found expressed in
human brain, spinal cord, and dorsal root ganglia (Ho et al., 1995). The high expression level
of mRNA is maintained in the adult rat motor neurons and dorsal root ganglia. SMDF is
biologically active and upon ligand – receptor interaction stimulates tyrosine phosphorylation
of a 185kD protein (Hayes et al., 1992).
The biological functions of SMDF are varied. Its high expression in spinal motor neurons and
dorsal root ganglia in the developing human and rodents might suggests a role at the
developing neuromuscular junction and possible role in the motor and sensory neuron
development (Ho et al., 1995). Consistent with this, SMDF promotes Schwann cell
proliferation as there is high level of gene expression in large neurons. Similar to other
neuronal peptides, SMDF may be produced in the motor neuron cell body but is transported
through the motor axons to the nerve terminal, where it exerts its effects on muscle
acetylcholine receptor synthesis and/ or the proliferation of Schwann cells (Hall and Sanes,
140
Discussion
1993). In addition, the expression of SMDF in the adult rat spinal cord motor neurons suggests
that it may also act at mature neuromuscular junctions via reinnervation of muscle fibers by
motor neurons following nerve damage.
In our screening experiment on the BmKII fraction, we found that fractions RP5.2G1,
RP6.4F7 and RP6.3F4 were able to increase gene expression of SMDF. These fractions can be
tested on PC12 cells to see whether it may promote nerve growth following nerve growth
damage in vitro.
5.3.5 Fibroblast growth factor 2 (FGF2)
FGF-2 is a member of a large family of structurally related proteins that affect the growth,
differentiation, migration and survival of a wide variety of cell types (Bikfalvi et al, 1997).
FGF2 or also known as basic FGF is a major growth factor extracted from prostatic tissue
(Dow et al., 2000). This molecule belongs to a member of a large family of proteins that bind
heparin and heparin sulfate and modulate the function of a wide range of cell types.
FGF-2 interacts with specific cell surface receptor proteins derived from four separate genes.
FGF-2 has been proposed to have two separate receptor binding sites which may allow a single
FGF-2 to bind to two receptors or to interact with a single receptor in two separate positions
(Kan et al., 1993). FGF-2 is released from cells as result of cell damage, death and non-lethal
membrane disruptions and they are stored in the extracellular matrix and basement membrane
(Conrad, 1998; Folkman et al., 1988).
Extracellular FGF-2 binds to cell surface receptors and heparin sulfate proteoglycans (HSPG)
and is subjected to internalization and lysosomal degradation (Nugent and Iozzo, 2000). FGF2 plays major roles in development, remodeling and disease states in almost every organ
system (Bikfalvi et al, 1997). The most well studied function of FGF-2 is its ability to regulate
growth and function of vascular cells such as endothelial and smooth muscle cells. It has also
been implicated in the development and growth of new blood vessels (angiogenesis) and in the
pathogenesis of vascular diseases such as atherosclerosis (Bikfalvi et al, 1997). In addition,
FGF-2 stimulates wound healing, tissue repair, and hematopoiesis, and stem cell survival
141
Discussion
(Dow et al., 2000). FGF-2 also found to play a key role in tumorigenesis though its ability to
induce angiogenesis.
In the brain especially in cerebral cortex, hippocampus, cerebellum, retina, ciliary ganglion
and spinal cord, FGF-2 promotes proper function of the nervous system (Dow et al., 2000).
The authors postulated that basic FGF-2 stimulates glial cells to produce a trophic factor(s)
which in turn elicits enhanced neuronal survival and neurite outgrowth. FGF-2 has pronounced
effects on central nervous system astrocytes stimulating their proliferation and inducing the
synthesis of glial fibrillary acidic protein (GFAP; Morrison et al., 1986). The summary of
FGF2 intracellular signaling pathway is depicted in Figure 5.3.
FGF-2 gene expression was increased upon treatment with RP5.2G1, RP6.4F7 and RP6.3F4.
Since FGF2 is able to promote neurite outgrowth, these three fractions can be tested on PC12
cells as an assay for neurite outgrowth. In addition, the synthesis of GFAP can be measured by
immunostaining with a GFAP antibody in an astrocytic cell line such as U-373 MG.
142
Discussion
Figure 5.3: Schematic diagram of FGF-2 signalling pathways which has been shown to
activate a number of intracellular signalling routes. Binding of FGF-2 to its receptors is
enhanced by cell surface HSPG and leads to activation of autophosphorylation of the FGF-R
in several tyrosine residues. Some of the phosphotyrosine residues are binding sites for src
homology domain containing proteins such as phospholipase C- and others are binding sites
for proteins with phosphotyrosine-binding domains such as FGF receptor substrate 2 (FRS2)
and SHC. SHC and FRS2 function as docking proteins which bind the GRB2–SOS complex
which then activates RAS. RAS recruits RAF-1, a serine/threonine kinase that actives MEK.
MEK proceeds to activate the mitogen activated protein kinases (MAPK) which translocate to
the nucleus where they directly activate transcription factors by phosphorylation. The
activation of PLC-
also plays a major role in transmitting the eventual FGF-2-mediated
biological signals. PLC- activation results in hydrolysis of phosphatidylinositol to inositol-3phosphate and diacylglycerol (DAG) leading to Ca2+ release and activation of protein kinase C
(PKC). The biological end responses for FGF-2 may lead to stimulation of cell proliferation
and migration as well as neuronal survival. Image adapted from Nugent and Iozzo, (2000).
143
Discussion
5.3.6 Krit1
Krit1 (for Krev Interaction Trapped 1) is a protein of 529 amino acid, with a amino-terminal
ankyrin repeat domain and a novel carboxyl-terminal domain required for association with
Krev-1 (Serebriiskii et al., 1997). Krev-1 is an evolutionarily conserved Ras-family GTPase
whose cellular function remains unclear but has been proposed to function as a tumor
suprressor gene and may act as a Ras antagonist. Mutation of krit1 caused cerebral cavernous
malformations (CCM), a type of congenital vascular anomalies of the brain that can cause
significant neurological disabilities, including intractable seizures and hemorrhagic stroke
(Sahoo et al., 1999). Interaction of Krev-1 and Krit1 may involve in signaling pathway in
angiogenesis and cerebovascular disease.
So far, not much is known about Krit1 as it is not strongly homologous to any previously
described protein. Krit1 is suspected to interact weakly with a family of cytoskeletal and
membrane-associated proteins defined by erythrocyte protein 4.1 (Conboy et al., 1986). It may
also be a potential positive regulator of Krev-1 activity and provide an indirect mechanism to
enhance tumor progression (Serebriiskii et al., 1997). As additional evidence for Krit1
involvement in tumor suppression processes, krit1 ortholog was found to be transcriptionally
upregulated in Chinese hamster ovary (CHO) cell. In addition, Krit1 may act in localizing the
Krev-1 to an appropriate cellular compartment for function in tumor progression and cellular
growth control (Serebriiskii et al., 1997).
Northern analysis of krit1 showed weak expression in heart, muscle and brain but not in a
number of other tissues (Serebriiskii et al., 1997).
In the screening experiment, several
fractions of BmKII were found to alter Krit1 gene expression. BmKII downregulated the gene
expression of Krit1 by 5-fold and 22-fold in microarray and real-time studies respectively.
Intriguingly, subfractions of RP1–9 upregulated Krit1 gene expression. Fraction RP5.2 caused
the highest fold of changes of 7-fold gene upregulation.
144
Discussion
5.4 Conclusion and future studies
A proteomics study can be performed by checking the protein profile of the six genes using
Western Blot. Each of the fractions RP5.2G1, RP6.2F4, RP6.3F4, RP6.3F5 and RP6.4F7 can
be individually tested on the neuroblastoma cell line to check for presence of amphiregulin,
neuregulin, Krit1, GABA (A) receptor, FGF2 and galanin protein expression level using
specific antibody. This may provide useful evidence whether these toxin fractions caused any
translational regulation as it did at the transcriptional level.
So far, the role of these toxins focused largely on the physiological function at the Na+
channel. RP5.2G1 which shares sequence similarity to bukatoxin and BmK T toxins (Table
4.9). Bukatoxin belongs to α-toxins family that prolongs the action potential of excitable cells
by blocking sodium channel inactivation and neuronal transmission. It can also produce a
marked, concentration-dependent relaxation of precontracted rat anococcygeus muscle (ACM;
Srinivasan et al., 2001). BmK T has anti tumor effect and strong inhibitory effect on pain (Liu
et al., 2003). BmK toxin I, a voltage-gated Na+ channel modulator was found to evoke
profound change in c-Fos protein expression pattern in rat spinal cord (Bai et al., 2003). The
authors found that BmK toxin I may be a key contributor of c-Fos expression induced by BmK
venom. It is noteworthy that we also found certain protein fractions in BmKII venom was able
to modulate genes involved in neurogenesis, angiogenesis, and other ion channel genes.
145
Discussion
REFERENCES
94
References
Aiken, S.P., Sellin, L.C., Schmidt, J.J., Weinstein, S.A., and McArdle, J.J. (1992) A novel peptide
toxin from Trimeresurus wagleri acts pre- and post-synaptically to block transmission at the rat
neuromuscular junction. Pharmacol. Toxicol. 70, 459-462.
Alkondon, M., and Albuquerque, E.X. (1993) Diversity of nicotinic acetylcholine receptors in rat
hippocampal neurons. I. Pharmacological and functional evidence for distinct structural subtypes, J.
Pharmacol. Exp. Ther. 265, 1455-1473.
Altschul, S.F., Madden, T.L., Schaffer, A.A., Zhang, J., Zhang, Z., Miller, W., and Lipman, D.J.
(1997) Gapped BLAST and PSI-BLAST: a new generation of protein database search programs.
Nucleic Acids Res. 25, 3389-3402.
Amiranoff, B., Lorinet, A.M., Laburthe, M. (1989) Galanin receptor in the rat pancreatic beta cell line
Rin m 5F. Molecular characterization by chemical cross-linking. J. Biol. Chem. 264, 20714-20717.
Anderson, A.J., Harvey, A.L., and Mbugua, P.M. (1985) Effects of fasciculin 2, an anticholinesterase
polypeptide from green mamba venom, on neuromuscular transmission in mouse diaphragm
preparations. Neurosci. Lett. 54, 123-128.
Antil, S., Servent, D., and Menez, A. (1999) Variability among the sites by which curaremimetic
toxins bind to torpedo acetylcholine receptor, as revealed by identification of the functional residues of
alpha-cobratoxin. J. Biol. Chem. 274, 34851-34858.
Antil-Delbeke, S., Gaillard, C., Tamiya, T., Corringer, P.J., Changeux, J.P. Servent, D., and Menez,
A. (2000) Molecular determinants by which a long chain toxin from snake venom interacts with the
neuronal alpha 7-nicotinic acetylcholine receptor, J. Biol. Chem. 275, 9594-9601.
Bagetta, G., Nistico, G. and Dolly, J.O. (1992) Production of seizures and brain damage in rats by dendrotoxin, a selective K+ channel blocker. Neurosci. Lett. 139, 34–40.
Bai, Z.T., Zhang, X.Y., and Ji, Y.H. (2003) Fos expression in rat spinal cord induced by peripheral
injection of BmK I, an alpha-like scorpion neurotoxin. Toxicol. Appl. Pharmacol. 192, 78-85.
Balozet, L. (1971) Scorpionism in the old world. In: Venomous Animals and Their Venoms (Bucherl,
W. and Buckley, E.E, eds.) pp 349-371, New York, Academic Press.
Barker, W.C., Garavelli, J.S., Hou, Z., Huang, H., Ledley, R.S., McGarvey, P.B., Mewes, H.W.,
Orcutt, B.C., Pfeiffer, F., Tsugita, A., Vinayaka, C.R., Xiao, C., Yeh, L.S., and Wu, C. (2001) Protein
Information Resource: a community resource for expert annotation of protein data. Nucleic Acids Res.
29: 29-32.
Bartfai, T., Hokfelt, T., and Langel, U. (1993) Galanin--a neuroendocrine peptide. Crit. Rev.
Neurobiol. 7, 229-274.
Batista, C.V., Gomez-Lagunas, F., Lucas, S., and Possani, L.D. (2000). Tc1, from Tityus cambridgei,
is the first member of a new subfamily of scorpion toxin that blocks K(+)-channels. FEBS Lett. 486,
117-120.
146
References
Bauer, F.E., Ginsberg, L., Venetikou, M., MacKay, D.J., Burrin, J.M., and Bloom, S.R. (1986) Growth
hormone release in man induced by galanin, a new hypothalamic peptide. Lancet 2, 192-195.
Bawaskar, H.S and Bawaskar, P.H. (1989) Stings by red scorpion (Buthus tamulus) in Maharashtra
state, India: a clinical study. Ann. Trop. Med. Parasit. 81, 719-723.
Benishin, C.G. (1990) Potassium channel blockade by the β subunit of the beta-bungarotoxin. Mol.
Pharmacol. 38, 164-169.
Benraiss, A., Chmielnicki, E., Lerner, K., Roh, D., and Goldman, S.A. (2001) Adenoviral brainderived neurotrophic factor induces both neostriatal and olfactory neuronal recruitment from
endogenous progenitor cells in the adult forebrain. J. Neurosci. 21, 6718-6731.
Betzel, C., Lange, G., Pal, G.P., Wilson, K.S., Maelicke, A. and Saenger, W. (1991) The refined
crystal structure of alpha-cobratoxin from Naja naja siamensis at 2.4-A resolution. J. Biol. Chem. 266,
21530-21536.
Bikfalvi, A., Klein, S., Pintucci, G., and Rifkin, D.B. (1997) Biological roles of fibroblast growth
factor-2. Endocr Rev. 18, 26-45.
Boehm, S., and Betz, H. (1997) Somatostatin inhibits excitatory transmission at rat hippocampal
synapses via presynaptic receptors. J. Neurosci. 17, 4066-4075.
Bon, C., Changeux, J.P., Jeng, T.W., and Fraenkel-Conrat, H. (1979) Postsynaptic effects of crotoxin
and of its isolated subunits. Eur. J. Biochem. 99, 471-481.
Bouchier, C., Boulain, J.C., Bon, C., and Menez, A (1991) Analysis of cDNAs encoding the two
subunits of crotoxin, a phospholipase A2 neurotoxin from rattlesnake venom: the acidic non enzymatic
subunit derives from a phospholipase A2-like precursor. Biochim. Biophys. Acta 1088, 401-408.
Brejc, K., van Dijk, W.J., Klaassen, R.V., Schuurmans, M., van Der Oost, J., Smit, A.B., and Sixma,
T.K. (2001) Crystal structure of an ACh-binding protein reveals the ligand-binding domain of nicotinic
receptors. Nature 411, 269-276.
Brown, N.P., Leroy, C., and Sander, C., (1998) MView: a web-compatible database search or multiple
alignment viewer. Bioinformatics 14, 380–381.
Brusic, V., Zeleznikow, J., and Petrovsky, N. (2000) Molecular immunology databases and data
repositories. J. Immunol. Methods 238, 17-28.
Bucherl, W. (1971) Classification, biology, and venom extraction of scorpions. In: Venomous Animals
and their venoms (Bucherl, W. and Buckley, E.E., eds) pp 317-348, Academic Press, New York.
Catterall, W. A. (1980) Neurotoxins that act on voltage-sensitive sodium channels. Ann. Rev.
Pharmac. Toxicol. 20, 15-43.
Catterall, W.A. (1986) Molecular properties of voltage-sensitive sodium channels. Annu. Rev.
Biochem. 55, 953-985.
147
References
Cervenansky, C., Dajas, F., Harvey, A. L., and Karlsson, E (1991) Fasciculins, anticholinesterase
toxins from mamba venoms: Biochemistry and pharmacology. In: Snake Toxins (Harvey, A.L. ed.) pp
303-321, Pergamon Press, New York.
Chang, C.C. (1979) The action of snake venoms on nerve and muscle. In: Snake venoms, Handbook of
Experimental Pharmacology (Lee, C.Y. ed) pp. 30-376, Springer-Verlag, Berlin.
Chang, C.C., and Lee, C.Y (1963) Isolation of neurotoxins from the venom of Bungarus multicinctus
and their modes of neuromuscular blocking actions. Arch. Int. Pharmacodyn. 144, 241-257.
Chang, C.C., Huang, T.Y., Kuo, K.W., Chen, S.W., Huang, K.F., and Chiou, S.H. (1993) Sequence
characterization of a novel alpha-neurotoxin from the king cobra (Ophiophagus hannah) venom.
Biochem. Biophys. Res. Commun. 191, 214-223.
Chang, L. S., Lin, S. K., Huang, H. B. and Hsiao, M. (1999) Genetic organization of α-bungarotoxin
from Bungarus multicinctus (Taiwan banded krait): evidence showing that the production of αbungarotoxin isotoxins is not derived from edited mRNAs. Nucleic Acids Res. 27, 3970-3975.
Chang, L.S., Lin, S. K and Wu, P. F. (1998) Differentially expressed snoRNAs in Bungarus
multicinctus (Taiwan Banded krait) Biochem. Biophys. Res. Commun. 245, 397-402.
Changeux, J. P. (1993) Allosteric proteins: from regulatory enzymes to receptors – personal
recollections. Bioessays 15, 625-634.
Cher, C. D., Armugam, A,, Lachumanan, R., Coghlan, M.W., and Jeyaseelan, K. (2003) Pulmonary
inflammation and edema induced by phospholipase A2: global gene analysis and effects on aquaporins
and Na+/K+-ATPase. J. Biol. Chem. 278, 31352-31360.
Chiappinelli, V.A (1985) Actions of snake venom toxins on neuronal nicotinic receptors and other
neuronal receptors. Pharmacol. Ther. 31, 1-32.
Chiappinelli, V.A. (1991) Structure-function relationship of postsynaptic neurotoxins from snake
venoms, In: Snake Toxins (Harvey, A.L ed.) pp. 223-258, Pergamon Press, New York.
Chiappinelli, V.A. (1992) Snake venom kappa-neurotoxins distinguish subtypes of neuronal nicotinic
receptors, In: Recent Advances in Toxicology Research, (Gopalakrishakone, P. and Tan, C.K. eds.) pp.
103-120, National University of Singapore Press, Singapore.
Chicheportiche, R, Vincent, J.P., Kopeyan, C., Schweitz, H., and Lazdunski, M. (1975) Structurefunction relationship in the binding of snake neurotoxins to the torpedo membrane receptor.
Biochemistry 14, 2081-2091.
Clifford, H.P. (1955) The Reptile World: A natural history of the snakes, lizards, turtles, and
crocodilians. Alfred A Knopf, New York.
Coleman, M.H., Yamaguchi, S.-I. and Rogawski, M.A. (1992) Protection against dendrotoxin-induced
clonic seizures in mice by anticonvulsant drugs. Brain Res. 575, 138–142.
148
References
Collinson, N., Kuenzi, F.M., Jarolimek, W., Maubach, K.A., Cothliff, R., Sur, C., Smith, A., Out,
F.M., Howell, O., Atack, J.R., McKernan, R.M., Seabrook, G.R., Dawson, G.R., Whiting, P.J., and
Rosahl, T.W. (2002) Enhanced learning and memory and altered GABAergic synaptic transmission in
mice lacking the alpha 5 subunit of the GABAA receptor. J. Neurosci. 22, 5572-5580.
Conboy, J., Kan, Y.W., Shohet, S.B., and Mohandas, N. (1986) Molecular cloning of protein 4.1, a
major structural element of the human erythrocyte membrane skeleton. Proc Natl Acad Sci U S A. 83,
9512-9516.
Conrad, H.E.(1998) Heparin-binding Proteins, Academic Press, San Diego.
Consolo, S., Bertorelli, R., Girotti, P., La Porta, C., Bartfai, T., Parenti, M., and Zambelli M. (1991)
Pertussis toxin-sensitive G-protein mediates galanin's inhibition of scopolamine-evoked acetylcholine
release in vivo and carbachol-stimulated phosphoinositide turnover in rat ventral hippocampus.
Neurosci Lett. 126, 29-32.
Corfield, P.W., Lee, T.J. and Low, B.W. (1989) The crystal structure of erabutoxin a at 2.0-A
resolution. J. Biol. Chem. 264, 9239-9242.
Corringer, P.J., Le Novere, N., and Changeux, J.P. (2000) Nicotinic receptors at the amino acid level.
Annu. Rev. Pharmacol. Toxicol. 40, 431-58.
Couraud, F., Jover, E., Dubois, J.M.., and Rochat, H. (1982) Two types of scorpion receptor sites, one
related to the activation, the other to the inactivation of the action potential sodium channel. Toxicon
20, 9-16.
Crawley, J.N. (1993) Functional interactions of galanin and acetylcholine: relevance to memory and
Alzheimer's disease. Behav. Brain Res. 57, 133-141.
Crestani, F., Keist, R., Fritschy, J.M., Benke, D., Vogt, K., Prut, L., Bluthmann, H., Mohler, H., and
Rudolph, U. (2002) Trace fear conditioning involves hippocampal alpha5 GABA(A) receptors. Proc.
Natl. Acad. Sci. USA. 99, 8980-8985.
Dajas, F., Bolioli, B., Castello, M.E. and Silveira, R. (1987) Rat striatal acetylcholinesterase inhibition
by fasciculin (a polypeptide from green mamba snake venom). Neurosci. Lett. 77, 887-891.
DeBin, J.A., Maggio, J.E., and Strichartz, G.R. (1993) Purification and characterization of chlorotoxin,
a chloride channel ligand from the venom of the scorpion. Am. J. Physiol. 264, C361-9.
Deneris, E.S., Connolly, J., Rogers, S.W., and Duvoisin, R. (1991) Pharmacological and functional
diversity of neuronal nicotinic acetylcholine receptors. Trends Pharmacol. Sci. 12, 34-40.
Devi, A. (1968) The protein and non-protein constituents of snake venoms. In: Venomous animals and
their venoms (Bucherl. W., Buckley. E., and Deulofeu, V. eds.) pp 119-165, Academic Press, New
York.
Dewan, J.C., Grant, G.A., and Sacchettini, J. C. (1994) Crystal structure of kappa-bungarotoxin at 2.3A resolution, Biochemistry 33, 13147-13154.
149
References
Dow, J.K., and deVere White, R.W. (2000) Fibroblast growth factor 2: its structure and property,
paracrine function, tumor angiogenesis, and prostate-related mitogenic and oncogenic functions.
Urology 55, 800-806.
Drachman, D.B. (1981) The biology of myasthenia gravis. Annu. Rev. Neurosci. 4, 195-225.
Drake, C.T., Terman, G.W., Simmons, M.L., Milner, T.A., Kunkel, D.D., Schwartzkroin, P.A., and
Chavkin, C. (1994) Dynorphin opioids present in dentate granule cells may function as retrograde
inhibitory neurotransmitters. J. Neurosci. 14, 3736-3750.
Dufton, M.J, and Hider, R.C. (1991) The structure and pharmacology of elapid cytotoxins. In: Snake
Toxins (Harvey, A.L. ed.) pp. 259-302, Pergamon Press, New York.
Dufton, M.J. (1984) Classification of elapid snake neurotoxins and cytotoxins according to chain
length: evolutionary implications, J. Mol. Evol. 20, 128-134.
Dufton, M.J. and Hider, R.C. (1983a) Classification of phospholipase A2 according to sequence.
Evolutionary and pharmacologica implications. Eur. J. Biochem. 137, 545-551.
Dufton, M.J. and Hider, R.C. (1983b) Conformational properties of the neurotoxins and cytotoxins
isolated from Elapid snake venoms. CRC Crit. Rev. Biochem. 14, 113-171.
Eaker, D. and Porath, J. (1967) The amino acid sequence of neurotoxin from Naja nigricollis venom.
Jpn. J. Microbiol. 11, 353-355.
Ekins, R.P. (1989) Multi-analyte immunoassay. J. Pharm. Biomed. Anal. 7, 155-168.
Ekins, R.P. and Chu, F. (1992) Multispot, multianalyte immunoassay. The microanalytical ‘compact
disk’ of the future. Ann. Biol. Clin (Paris) 50, 337-353.
Endo, T., and Tamiya, N. (1991) Structure-function relationship of postsynaptic neurotoxins from
snake venoms. In: Snake Toxins, (Harvey, A.L., ed.) pp 165-222, Pergamon Press, New York.
Espiritu, D.J., Watkins, M., Dia-Monje, V., Cartier, G. E., Cruz, L.J., and Olivera, B.M. (2001)
Venomous cone snails: molecular phylogeny and the generation of toxin diversity. Toxicon 39, 18991916.
Falk, A., and Frisen, J. (2002) Amphiregulin is a mitogen for adult neural stem cells. J. Neurosci. Res.
69, 757-762.
Faure, G., Boulain, J.C., Bouet, F., Montenay-Garestier, T., Fromageot, P., and Menez, A. (1983) Role
of indole and amino groups in the structure and function of Naja nigricollis toxin alpha. Biochemistry
22, 2068-2076.
Fenton, J. (2002) Toxicology: a case-oriented approach, pp 537, CRC Press, Boca Raton.
Fiordalisi, J.J., al-Rabiee, R., Chiappinelli, V.A., and Grant, G.A. (1994) Affinity of native kappabungarotoxin and sitedirected mutants for the muscle nicotinic acetylcholine receptor. Biochemistry
33, 12962-12967.
150
References
Fischer, H.S. and Saria, A., (1999) Voltage-gated, margatoxin-sensitive potassium channels, but not
calcium-gated, iberiotoxin-sensitive potassium channels modulate acetylcholine release in rat striatal
slices. Neurosci. Lett. 263, 208–210.
Fletcher, J.E., and Rosenberg, P. (1997) The cellular effects and mechanisms of action of
presynaptically acting phospholipase A2 toxins. In: Venom Phospholipase A2 Enzymes: Structure,
Function and Mechanism (Kini, R.M. ed.) pp 373-391, John Wiley and Sons Ltd, Chichester.
Fodor, S.P., Read, J.L., Pirrung, M.C., Stryer, L., Lu, A.T., and Solas, D. (1991) Light-directed,
spatially addressable parallel chemical synthesis. Science 251, 767-773.
Fohlman, J., Lind, P., and Eaker, D. (1977) Taipoxin, an extremely potent presynaptic snake venom
neurotoxin. Elucidation of the primary structure of the acidic carbohydrate-containing taipoxinsubunit, a prophospholipase homolog. FEBS Lett, 84, 367-371.
Folkman, J., Klagsbrun, M., Sasse, J., Wadzinski, M., Ingber, D., and Vlodavsky, I. (1988) A heparinbinding angiogenic protein--basic fibroblast growth factor--is stored within basement membrane. Am.
J. Pathol. 130, 393-400.
Francis, B., Coffield, J.A., Simpson, L.L., and Kaiser I.I. (1995) Amino acid sequence of a new type of
toxic phospholipase A2 from the venom of the Australian tiger snake (Notechis scutatus scutatus).
Arch. Biochem. Biophys. 318, 481-488.
Francke, O.F. (1982) Studies on the scorpion subfamilies Superstitioninae and Typhlochactinae, with
description of a new genus (Scorpiones, Chactoidea). Bulletin of the Association of Mexican Cave
Studies 8, 51-61.
Fry, B., Wuster, W., Kini, R.M., Brusic, V., Khan, A., Venkataraman, D., and Rooney, A.P. (2003)
Molecular evolution and phylogeny of elapid snake venom three-finger toxins. J. Mol. Evol. 57, 11029.
Gallagher, P.G., Bao, Y., Serrano, S.M., Kamiguti, A.S., Theakston, R.D., and Fox, J.W. (2003) Use
of microarrays for investigating the subtoxic effects of snake venoms: insights into venom-induced
apoptosis in human umbilical vein endothelial cells. Toxicon 41, 429-40.
Garcia, C., Becerril, B., Selisko, B., Delepierre, M., and Possani, L.D. (1997) Isolation,
characterization and comparison of a novel crustacean toxin with a mammalian toxin from the venom
of the scorpion Centruroides noxius Hoffmann. Comp. Biochem. Physiol. B. Biochem. Mol. Biol. 116,
315-322.
Garcia, M.L., Hanner, M., Knaus, H.G., Koch, R., Schmalhofer, W., Slaughter, R.S., and
Kaczorowski, G.J. (1997) Pharmacology of potassium channels. Adv. Pharmacol. 39, 425-471.
Gong, J.P., Gong, Y.P., Liu, J.F., and Zhang, Y.S. (1992) Cardiovascular effects of scorpion venom
(Buthus martensi Karsch). In: Recent advances in toxinology research (Gopalakrishnakone, P. and
Tan, C.K. eds.) pp 50-60, National University of Singapore Press, Singapore.
151
References
Gong, N.L., Armugam, A., and Jeyaseelan, K. (1999) Postsynaptic short-chain neurotoxins from
Pseudonaja textilis: cDNA cloning, expression and protein characterization. Eur. J. Biochem. 265,
982-989.
Gong, N.L., Armugam, A., and Jeyaseelan K. (2000) Molecular cloning, characterization and
evolution of the genes encoding a new group of short-chain alpha-neurotoxins in an Australian elapid,
Pseudonaja textilis. FEBS Lett. 473: 303-310.
Gopalakrisnakone, P. (1990) A computer based colour-photo database system for dangerous animals
and plants: academic and public information. Toxicon 28, 1285-92.
Gordon, D., Martin-Eauclaire, M.F., Cestele, S., Kopeyan, C., Carlier, E., Khalifa, R.B., Pelhate, M.,
and Rochat, H. (1996) Scorpion toxins affecting sodium current inactivation bind to distinct
homologous receptor sites on rat brain and insect sodium channels. J Biol. Chem. 271, 8034-8045.
Gordon, D., Savarin, P., Gurevitz, M., and Zinn-Justin, S. (1998) Functional anatomy of scorpion
toxins affecting sodium channels. J. Toxicol. Toxin. Rev. 17, 131-159.
Goudet, C., Chi, C.W., and Tytgat, J. (2002) An overview of toxins and genes from the venom of the
Asian scorpion Buthus martensi Karsch. Toxicon 40, 1239-58.
Grant, G.A and Chiappinelli, V.A (1985) kappa-Bungarotoxin: complete amino acid sequence of a
neuronal nicotinic probe. Biochemistry 24, 1532-1537.
Grant, G.A., Frazier, M.W., and Chiappinelli, V.A. (1988) Amino acid sequence of kappa-flavitoxin:
establishment of a new family of snake venom neurotoxins. Biochemistry 27, 3794- 3798.
Grutter, T. and Changeux, J.P. (2001) Nicotinic receptors in wonderland. Trends Biochem. Sci. 26,
459-463.
Hains, P.G., Ramsland, P. A., and Broady, K.W. (1999) Modelling of acanthoxin A1, a PLA2 enzyme
from the venom of the common death adder (Acanthophis antarcticus) Proteins 35, 80-88.
Hall, Z.W., and Sanes, J.R. (1993) Synaptic structure and development: the neuromuscular junction.
Cell 72, 99-121.
Habert-Ortoli, E., Amiranoff, B., Loquet, I., Laburthe, M., and Mayaux, J.F. (1994) Molecular cloning
of a functional human galanin receptor. Proc Natl Acad Sci U S A. 91, 9780-3.
Hallbergson, A.F., Gnatenco, C., and Peterson, D.A. (2003) Neurogenesis and brain injury: managing
a renewable resource for repair. J. Clin. Invest. 112, 1128-1133.
Halpert, J. and Eaker, D (1975) Amino acid sequence of a presynaptic neurotoxin form the venom of
Notechis scutatus scutatus (Australian tiger snake). Biochimie 61, 719-723.
Harris, J. B. (1991) Phospholipases in snake venoms and their effects on nerve and muscle. In: Snake
Toxins (Harvey, A.L., ed.) pp. 91-129, Pergamon Press, New York.
Harvey, A. L. (2001) Twenty years of dendrotoxins. Toxicon. 39, 15-26.
152
References
Harvey, A.L. (2002a) Toxins ‘R’ us: more pharmacological tools from nature’s superstore. Trends
Pharmacol. Sci. 23, 201-203.
Harvey, A.L. (2002b) Toxins leading to medicines. In: Perpectives in molecular toxinology (Menez,
A., ed.) pp 377-382, Chichester, John Wiley & Sons, England.
Harvey, A.L., and Anderson, A.J. (1991) Dendrotoxins: Snake toxins that block potassium channels
and facilitate neurotransmitter release. In: Snake Toxins (Harvey, A.L. ed.) pp. 131-164, Pergamon
Press, New York.
Harvey, A.L., Anderson, A.J., Mbugua, P.M., and Karlsson, E (1984) Toxins from mamba venoms
that facilitate neuromuscular transmission. Toxin Rev. 3, 91-137.
Harvey, A.L., Bradley, K.N., Cochran, S.A., Rowan, E.G., Pratt, J.A., Quillfeldt, J.A., and
Jerusalinsky, D.A. (1998) What can toxins tell us for drug discovery? Toxicon 36, 1635-40.
Hawgood, B. and Bon, C. (1991) Snake pre-synaptic toxins. In: Handbook of natural toxins, Reptile
venoms and toxins (Tu, A.T., ed.) pp 3-52, Marcel Dekker, New York.
Hayes, C., Kelly, D., Murayama, S., Komiyama, A., Suzuki K, and Popko B. (1992) Expression of the
neu oncogene under the transcriptional control of the myelin basic protein gene in transgenic mice:
generation of transformed glial cells. J Neurosci. Res. 31,175-187.
Hider, R.C., Karlsson, E., and Namiranian, S. (1991) Separation and purification of toxins from snake
venoms, In: Snake Toxins (Harvey, A.L. ed.) pp1-34, Pergamon Press, New York.
Ho, W.H., Armanini, M.P., Nuijens, A., Phillips, H.S., and Osheroff, P.L. (1995) Sensory and motor
neuron-derived factor. A novel heregulin variant highly expressed in sensory and motor neurons. J.
Biol. Chem. 270, 14523-14532.
Hodgson, W.C and Wickramaratna, J.C. (2002) In vitro neuromuscular activity of snake venoms. Clin.
Exp. Pharmacol. Physiol. 29, 807-814.
Hori, H., and Tamiya, N. (1976) Preparation and activity of guanidinated or acetylated erabutoxins.
Biochem. J. 153, 217-222.
Howbrook, D.N., van der Valk, A.M., O'Shaughnessy, M.C., Sarker, D.K., Baker, S.C., and Lloyd,
A.W. (2003) Developments in microarray technologies. Drug Discov. Today 8, 642-51.
Ibanez-Tallon, I., Miwa, J.M., Wang, H.L., Adams, N.C., Crabtree, G.W., Sine, S.M., and Heintz, N.
(2002) Novel modulation of neuronal nicotinic acetylcholine receptors by association with the
endogenous prototoxin lynx1, Neuron 33, 893-903.
Ingels, M (2001) Snakes and other reptiles. In: Toxicology Secrets, (Ling L.J, Clark, R.F., Erickson,
T.B. and Trestrail, J.H., eds) pp 261-269, Hanley and Belfus, Philadelphia.
Ishikawa, Y., Menez, A., Hori, H., Yoshida, H., and Tamiya, N. (1977) Structure of snake toxins and
their affinity to the acetylcholine receptor of fish electric organ. Toxicon 15, 477-488.
153
References
Ji, R.R., Zhang, X., Zhang, Q., Dagerlind, A., Nilsson, S., Wiesenfeld-Hallin, Z., and Hokfelt, T.
(1995) Central and peripheral expression of galanin in response to inflammation. Neuroscience 68,
563-576.
Joubert, F.J. and Strydom, D. J, (1978) Snake venoms. The amino acid sequences of trypsin inhibitor E
of Dendroaspis polylepis polylepis (black mamba) venom. Eur. J. Biochem. 87, 191-198.
Jover, E., Couraud, F., and Rochat, H. (1980) Two types of scorpion neurotoxins characterized by their
binding to two separate receptor sites on rat brain synaptosomes. Biochem. Biophys. Res. Commun. 95,
1607-1614.
Kan, M., Wang, F., Xu, J., Crabb, J.W., Hou, J., and McKeehan, W.L. (1993) An essential heparinbinding domain in the fibroblast growth factor receptor kinase. Science 259, 1918-21.
Karlin, A. (2002) Emerging structure of the nicotinic acetylcholine receptors. Nature Rev. Neurosci. 3,
102-114.
Karlsson, E. (1979) Chemistry of protein toxins in snake venoms. In: Snake venoms, Handbook of
Experimental Pharmacology, (Lee, C.Y., ed.) pp. 159-212, Springer-Verlag, Berlin.
Karlsson, E. D., Mbugua, P., and Rodriguez-Ithurallde, D (1984) Fasciculins, anticholinesterase toxins
from the venom of green mamba Dendroaspis augusticeps. Pharmacol. Ther. 30, 259-276.
Kato, M., Inazu, T., Kawai, Y., Masamura, K., Yoshida, M., Tanaka, N., Miyamoto, K., and
Miyamori, I. (2003) Amphiregulin is a potent mitogen for the vascular smooth muscle cell line, A7r5.
Biochem. Biophys. Res. Commun. 301, 1109-1115.
Kemp, J.A. and McKernan, R.M. (2002) NMDA receptors as drug targets, Nat. Neurosci. 5, 10391042.
Kini, R. M., and Iwanaga, S. (1986) Structure-function relationships of phospholipase A2. I: prediction
of presynaptic neurotoxicity. Toxicon 24, 527-541.
Kini, R.M. (2002) Molecular moulds with multiple missions: functional sites in three-finger toxins.
Clin. Exp. Pharmacol. Physiol. 29, 815-822.
Kochva, E. (1987) The origin of snakes and evolution of the venom apparatus. Toxicon 25, 65-106.
Kondo, K., Narita, K., and Lee, C.Y. (1978) Chemical properties and amino acid composition of
beta1-bungarotoxin from the venom of Bungarus multicinctus (Formosan banded krait). J. Biochem
(Tokyo) 83, 91-99.
Kondo, K., Toda, H., Narita, K., and Lee CY. (1982a) Amino acid sequences of three betabungarotoxins (beta 3-, beta 4-, and beta 5- bungarotoxins) from Bungarus multicinctus venom. Amino
acid substitutions in the A chains. J Biochem (Tokyo) 91, 1531-48.
154
References
Kondo, K., Toda, H., Narita, K., and Lee, C.Y. (1982b) Amino acid sequence of beta 2-bungarotoxin
from Bungarus multicinctus venom. The amino acid substitutions in the B chains. J. Biochem. (Tokyo)
91, 1519-1530.
Kondo, K., Zhang, J.K., Xu, K., and Kagamiyama, H. (1989) Amino acid sequence of a presynaptic
neurotoxin, agkistrodotoxin, from the venom of Agkistrodon halys Pallas. J. Biochem. 105, 196-203.
Kuhn, H.G., Winkler, J., Kempermann, G., Thal, L.J., and Gage, F.H. (1997). Epidermal growth factor
and fibroblast growth factor-2 have different effects on neural progenitors in the adult rat brain. J.
Neurosci. 17, 5820-5829.
Kuhn, P., Deacon, A.M., Comsa, D.S., Rajaseger, G., Kini, R.M., Uson, I.I. and Kolatkar, P.R. (2000).
The atomic resolution structure of bucandin, a novel toxin isolated from the Malayan krait, determined
by direct methods. Acta Crystallogr. D. 56, 1401-1407.
Kwong, P.D., McDonald, N.Q., Sigler, P.B., and Hendrickson, W.A. (1995) Structure of beta 2bungarotoxin: potassium channel binding by Kunitz modules and targeted phospholipase action.
Structure 3, 1109-1119.
Langel, U., and Bartfai, T. (1998) Chemistry and molecular biology of galanin receptor ligands. Ann.
N. Y. Acad. Sci. 863, 86-93.
Le Du. M.H., Marchot, P., Bougis, P.E. and Fontecilla-Camps, J.C. (1989) Crystals of fasciculin 2
from green mamba snake venom. Preparation and preliminary x-ray analysis. J. Biol. Chem. 264,
21401 – 21404.
Lee, C.Y., Tsai, M.C., Tsaur, M.L., M.W., Carlsson, F.H.H. and Joubert, F.J. (1985) Pharmacological
study on augusticeps-type toxins from mamba snake venoms. J. Pharmacol. Exp. Ther. 233, 491-498.
Lee, C.Y. (1972) Chemistry and pharmacology of polypeptide toxins in snake venoms. Ann. Rev.
Pharmacol. 12, 265-281.
Lee, C.Y., and Ho, C.L. (1982) In: Advances in Pharmacology and Therapeutics II, Vol. 4,
Biochemical Immunological Pharmacology (Yoshida, H., Hagihara, Y., and Ebashi, S., eds.) p37,
Pergamon Press, New York.
Lee, D.C., Sunnarborg, S.W., Hinkle, C.L., Myers, T.J., Stevenson, M.Y., Russell, W.E., Castner, B.J.,
Gerhart, M.J., Paxton, R.J., Black, R.A., Chang, A., and Jackson, L.F. (2003) TACE/ADAM17
processing of EGFR ligands indicates a role as a physiological convertase. Ann. N. Y. Acad. Sci. 995,
22-38.
Lentz, T.L., Burrage, T.G., Smith, A.L., Crick, J., and Tignor, G.H. (1982) Is the acetylcholine
receptor a rabies virus receptor? Science 215, 182-184.
Lentz, T.L., Wilson, P.T., Hawrot, E. and Speicher, D.W. (1984) Amino acid sequence similarity
between rabies virus glycoprotein and snake venom curaremimetic neurotoxins, Science 226, 847-848.
Leung, Y.F., and Cavalieri, D. (2003) Fundamentals of cDNA microarray data analysis. Trends Genet.
19, 649-659.
155
References
Lin, W.W., Lee, C.Y., Carlsson, F.H.H. and Joubert, F.J. (1987) Anticholinesterase activity of
augusticeps-type toxins and protease inhibitor homologues from mamba venoms. Acta Pacific J.
Pharmacology. 2, 79-85.
Lippens, G., Najib, J., Wodak, S. J., and Tartar, A. (1995) NMR sequential assignments and solution
structure of chlorotoxin, a small scorpion toxin that blocks chloride channels. Biochemistry 34, 13-21.
Lipshutz, R.J., Fodor, S.P., Gingeras, T.R., and Lockhart, D.J. (1999) High density synthetic
oligonucleotide arrays. Nat Genet. 21, 20-24.
Liu, C.M., and Pei, G.Q. (1989) Analgesic effect of venom Buthus martensi Karsch. Journal of
Shenyang College of Pharmacy 6, 176-180.
Liu, C.S., Hsiao, P.W., Chang, C.S., Tzeng, M.C., and Lo, T.B. (1989) Unusual amino acid sequence
of fasciatoxin, a weak reversibly acting neurotoxin in the venom of the banded krait, Bungarus
fasciatus. Biochem. J. 259, 153-158.
Liu, H.X., and Hokfelt, T. (2002) The participation of galanin in pain processing at the spinal level.
Trends Pharmacol. Sci. 23, 468-474.
Liu, Y.F., Ma, R.L., Wang, S.L., Duan, Z.Y., Zhang, J.H., Wu, L.J. and Wu, C.F. (2003) Expression of
an antitumor-analgesic peptide from the venom of Chinese scorpion Buthus martensii karsch in
Escherichia coli. Protein Expr. Purif. 27, 253-258.
Loret, E.P., Martin-Eauclaire, M.F., Mansuelle, P., Sampieri, F., Granier, C., and Rochat, H. (1991)
An anti-insect toxin purified from the scorpion Androctonus australis Hector also acts on the alphaand beta-sites of the mammalian sodium channel: sequence and circular dichroism study. Biochemistry
30, 633-640.
Marchot, P., Frachon, P., and Bougis, P.E. (1988) Selective distinction at equilibrium between the two
alpha-neurotoxin binding sites of Torpedo acetylcholine receptor by microtitration. Eur. J. Biochem.
174, 537-542.
Martin-Eauclaire, M.F. and Couraud, F (1995) Scorpion neurotoxins: effects and mechanisms. In:
Handbook of Neurotoxicology (Chang, L.W. and Dyer, R.S, eds) pp 683-716, Marcell and Dekker,
New York.
Matsui, T., Fujimura, Y., and Titani, K. (2000) Snake venom proteases affecting hemostasis and
thrombosis. Biochim. Biophys. Acta. 1477, 146-156.
Mazarati, A., Langel, U., and Bartfai, T. (2001) Galanin: an endogenous anticonvulsant?
Neuroscientist 7, 506-517.
Mazzotti, L. and Bravo-Becherelle, M.A. (1963) Scorpionism in the Mexican Republic. In: Venomous
and Poisonous Animals and Noxious Plants of the Pacific Area (Keegan J.L., and MacFarlane, M.V.
eds.) pp 119-131, Pergamon Press, London.
156
References
McDowell, R.S., Dennis, M.S., Louie, A., Shuster, M., Mulkerrin, M.G. and Lazarus, R.A. (1992)
Mambin, a potent glycoprotein IIb-IIIa antagonist and platelet aggregation inhibitor structurally related
to the short neurotoxins, Biochemistry 31, 4766-4772.
McLane, K.E., Weaver, W.R., Lei, S., Chiappinelli, V.A,, and Conti-Tronconi, B.M. (1993)
Homologous kappa-neurotoxins exhibit residue-specific interactions with the alpha 3 subunit of the
nicotinic acetylcholine receptor: a comparison of the structural requirements for kappa-bungarotoxin
and kappa-flavitoxin binding. Biochemistry. 32, 6988-6994.
Mebs,D. (1989) Snake venoms: toolbox of the neurobiologist, Endeavour 13, 157-161.
Meldrum, B., and Whiting, P.J. (2001) Anticonvulsants acting in the GABA system. In: Handbook of
Experimental Pharmacology: Inhibitory Amino Acid Transmitters (Mohlet, H. eds) pp. 173-194,
Springer-Verlag, Berlin.
Menez, A. (1998) Functional architectures of animal toxins: a clue to drug design? Toxicon 36, 15571572.
Merchenthaler, I., Lopez, F. J., and Negro-Vilar, A. (1993) Anatomy and physiology of central
galanin-containing pathways. Prog. Neurobiol. 40, 711-769.
Meves, H., Simard, J.M., and Watt, D.D. (1984) Biochemical and electrophysiological characteristics
of toxins isolated from the venom of the scorpion Centruroides sculpturatus. J. Physiol. (Paris). 79,
185-191.
Minton, S.A. (1990) Neurotoxic snake envenoming. Semin. Neurol. 10, 52 -61.
Miwa, J.M., Ibanez-Tallon, I., Crabtree, G.W., Sanchez, R., Sali, A., Role L.W., and Heintz, N.
(1999) lynx1, an endogenous toxin-like modulator of nicotinic acetylcholine receptors in the
mammalian CNS, Neuron 23, 105-114.
Miyazawa, A., Fijiyoshi, Y., and Unwin, N. (2003) Structure and gating of the acetylcholine receptor
pore. Nature 423, 949-955.
Morrison, R.S., Sharma, A., de Vellis, J., and Bradshaw, R.A. (1986) Basic fibroblast growth factor
supports the survival of cerebral cortical neurons in primary culture. Proc. Natl. Acad. Sci. USA. 83,
7537-7541.
Mourre, C., Lazdunski, M. and Jarrard, L.E. (1997). Behaviors and neurodegeneration induced by two
blockers of K+ channels, the mast cell degranulating peptide and dendrotoxin I. Brain Res. 762, 223–
227.
Naguib, M., Flood, P., McArdle, J.J., and Brenner, H.R. (2002) Advances in neurobiology of the
neuromuscular junction: implications for the anesthesiologist. Anesthesiology 96, 202-231.
Neri, P., Bracci, L., Rustici, M., and Santucci, A. (1990) Sequence homology between HIV gp120,
rabies virus glycoprotein, and and snake venom neurotoxins. Is the nicotinic acetylcholine receptor an
HIV receptor? Arch. Virol. 114, 265-269.
157
References
Neubig, R.R., and Cohen, J.B. (1980) Permeability control by cholinergic receptors in Torpedo
postsynaptic membranes: agonist dose-response relations measured at second and millisecond times.
Biochemistry 19, 2770-2779.
Nirthanan, S. and Gwee, M.C.E (2004) Three-finger α-neurotoxins and the nicotinic acetylcholine
receptor, forty years on. J. Pharmacol. Sci. 94, 1-17.
Nirthanan, S., Charpantier, E., Gopalakrishnakone, P., Gwee, M.C.E., Khoo, H.E., Cheah, L.S.,
Bertrand, D., and Kini, R.M. (2002) Candoxin, a novel toxin from Bungarus candidus, is a reversible
antagonist of muscle (alphabetagammadelta) but a poorly reversible antagonist of neuronal alpha 7
nicotinic acetylcholine receptors. J. Biol. Chem. 277, 17811-17820.
Nugent, M.A., and Iozzo, R.V. (2000) Fibroblast growth factor-2. Int J. Biochem. Cell Biol. 32, 115120.
Okuda, D., Nozaki, C., Sekiya, F., and Morita, T. (2001) Comparative biochemistry of disintegrins
isolated from snake venom: consideration of the taxonomy and geographical distribution of snakes in
the genus Echis. J. Biochem. (Tokyo). 129, 615-620.
Paaventhan, P., Joseph, J.S., Nirthanan, S., Rajaseger, G., Gopalakrishnakone, P., Kini, M.R. and
Kolatkar, P.R. (2003) Acta Crystallogr. D Biol. Crystallogr. 59, 584-586.
Pencea, V., Bingaman, K.D., Wiegand, S.J., and Luskin, M.B. (2001) Infusion of brain-derived
neurotrophic factor into the lateral ventricle of the adult rat leads to new neurons in the parenchyma of
the striatum, septum, thalamus, and hypothalamus. J. Neurosci. 21, 6706-6717.
Peng, F., Zeng, X.C., He, X.H., Pu, J., Li, W.X., Zhu, Z.H., and Liu, H. (2002) Molecular cloning and
functional expression of a gene encoding an antiarrhythmia peptide derived from the scorpion toxin.
Eur J Biochem. 269, 4468-75.
Peterson, D.A. (2002) Stem cells in brain plasticity and repair. Curr Opin. Pharmacol. 2, 34-42.
Peterson, D.A., and Gage, F.H. (1999) Trophic factor therapy for neuronal death. In: Alzheimer disease
(Terry, R.D., Karzman, R., Bick, K.L., and Sisodia, S.S. eds.) pp 337-388, Philadephia, Pennsylvannia,
USA.
Phelps, T. (1989) Poisonous snakes. Blandford Press, New York.
Pillet, L., Tremeau, O., Ducancel, F., Drevet, P., Zinn-Justin, S., Pinkasfeld, S., Boulain, J.C., and
Menez, A. (1993) Genetic engineering of snake toxins. Role of invariant residues in the structural and
functional properties of a curaremimetic toxin, as probed by site-directed mutagenesis. J. Biol. Chem.
268, 909-916.
Pimenta, A.M., Stocklin, R., Favreau, P., Bougis, P.E., and Martin-Eauclaire, M.F. (2001) Moving
pieces in a proteomic puzzle: mass fingerprinting of toxic fractions from the venom of Tityus
serrulatus (Scorpiones, Buthidae). Rapid Commun. Mass Spectrom. 17, 1562-72.
158
References
Pirrung, M.C., Fallon, L., and McGall, G. (1998) Proofing of photolithographic DNA synthesis with 3’
5’-dimethoxybenzoinloxycarbonyl-protected deoxynucleoside phosphoramidites. J. Organic Chem.
63, 241-246.
Plowman, G.D., Green, J.M., McDonald, V.L., Neubauer, M.G., Disteche, C.M., Todaro, G.J., and
Shoyab, M. (1990) The amphiregulin gene encodes a novel epidermal growth factor-related protein
with tumor-inhibitory activity. Mol. Cell Biol. 10, 1969-1981.
Poh, S.L., Mourier, G., Thai, R., Armugam, A., Molgo, J., Servent, D., Jeyaseelan, K., Menez, A.
(2002) A synthetic weak neurotoxin binds with low affinity to Torpedo and chicken alpha7 nicotinic
acetylcholine receptors. Eur. J. Biochem. 269, 4247-56.
Possani, L.D. (1984) Structure of scorpion toxins. In: Handbook of natural toxins (Tu, A.T., ed.) pp
513-550, Marcel Dekker Inc, New York.
Possani, L.D., Becerril, B., Delepierre, M., and Tytgat, J. (1999) Scorpion toxins specific for Na+channels. Eur. J. Biochem. 264, 287-300.
Pramanik, A. and Ogren S.O. (1992) Galanin-evoked acetylcholine release in the rat striatum is
blocked by the putative galanin antagonist M15. Brain Res. 574, 317-319.
Ramon y Cajal, S (1928) Degeneration and regeneration of the nervous system. Haffner Publishing
Co. New York, USA, p750.
Ray, J., and Peterson, D. A (2002) Neural stem cells in the adult hippocampus. In: Neural stem cells
for brain and spinal cord repair. (Zigova, T., Snyder, E.Y., and Sanberg, P.R., eds.) pp 269-286,
Humana Press, New Jersey, USA.
Ricciardi, A., Le Du, M.H., Khayati, M., Dajas, F., Boulain, J.C., Menez, A., and Ducancel, F. (2000)
Do structural deviations between toxins adopting the same fold reflect functional differences? J. Biol.
Chem. 275, 18302-18310.
Rodriguez-Ithurralde, D., Silveira, R., and Dajas, F. (1981) Gel chromatography and anticholinesterase
activity of Dendroaspis augusticeps venom. Braz. J. Med. Sci. 14, 394.
Rodriguez-Ithurralde, D., Silveira, R., Barbeito, Land., and Dajas, F. (1983) Fasciculin, powerful
anticholinesterase polypeptide from Dendroaspis augusticeps venom. Neurochem. Int. 5, 267-274.
Rosahl, T.W., (2003) Validation of GABA(A) receptor subtypes as potential drug targets by using
genetically modified mice. Curr Drug Target CNS Neurol Disord. 2, 207-212.
Rossowski, W.J. & D.H. Coy. (1989) Inhibitory action of galanin on gastric acid secretion in
pentobarbital-anesthetized rats. Life Sci. 44, 1807-1813.
Sahoo, T., Johnson, E.W., Thomas, J.W., Kuehl, P.M., Jones, T.L., Dokken, C.G., Touchman, J.W.,
Gallione, C.J., Lee-Lin, S.Q., Kosofsky, B., Kurth, J.H., Louis, D.N., Mettler, G., Morrison, L., GilNagel, A., Rich, S.S., Zabramski, J.M., Boguski, M.S., Green, E.D., and Marchuk, D.A. (1999)
Mutations in the gene encoding KRIT1, a Krev-1/rap1a binding protein, cause cerebral cavernous
malformations (CCM1). Hum. Mol. Genet. 8, 2325-2333.
159
References
Schena, M., Shalon, D., Davis, R.W., and Brown, P.O. (1995) Quantitative monitoring of gene
expression patterns with a complementary DNA microarray. Science. 270, 467-70.
Schena, M. (2003) Introduction to microarray analysis, In: Microarray analysis, pp 20-23, John Wiley
and Sons, Inc.
Schoepfer, R., Luther, M., and Lindstrom, J. (1988) The human medulloblastoma cell line TE671
expresses a muscle-like acetylcholine receptor. Cloning of the alpha-subunit cDNA, FEBS Lett. 226,
235-240.
Sege, K. (1983) Cross-reactivity of anti-idiotypic antibodies as a tool to study nonimmunoglobulin
proteins, Ann. N. Y. Acad. Sci. 418, 248-256.
Seguela, P., Wadiche, J., Dineley-Miller, K., Dani, J.A., and Patrick, J.W. (1993) Molecular cloning,
functional properties, and distribution of rat brain alpha 7: a nicotinic cation channel highly permeable
to calcium, J. Neurosci. 13, 596-604.
Sekhon, H.S., Jia, Y., Raab, R., Kuryatov, A., Pankow, J.F., Whitsett, J.A., Lindstrom, J., and Spindel,
E.R. (1999) Prenatal nicotine increases pulmonary alpha7 nicotinic receptor expression and alters fetal
lung development in monkeys. J. Clin. Invest. 103, 637-644.
Senior, K. (1999) Taking the bite out of snake venom, Lancet 353, 1946.
Serebriiskii, I., Estojak, J., Sonoda, G., Testa, J.R., and Golemis, E.A. (1997) Association of Krev1/rap1a with Krit1, a novel ankyrin repeat-containing protein encoded by a gene mapping to 7q21-22.
Oncogene 15, 1043-1049.
Servent, D and Menez, A (2001) Snake neurotoxins that interact with nicotinic acetylcholine receptors.
In:, Handbook of Neurotoxicity, (Massaro, E.J. ed.) pp385-425, New Jersey, Humana Press.
Servent, D., Antil-Delbeke, S., Gaillard, C., Corringer, P.J., Changeux, J.P., and Menez, A. (2000)
Molecular characterization of the specificity of interactions of various neurotoxins on two distinct
nicotinic acetylcholine receptors. Eur. J. Pharmacol. 393, 197-204.
Servent, D., Mourier, G., Antil, S., and Menez, A. (1998) How do snake curaremimetic toxins
discriminate between nicotinic acetylcholine receptor subtypes, Toxicol. Lett. 102-103, 199-203.
Servent, D., Winckler-Dietrich, V., Hu, H.Y., Kessler, P., Drevet, P., Bertrand, D., and Menez,
A.(2000) Only snake curaremimetic toxins with a fifth disulfide bond have high affinity for the
neuronal alpha7 nicotinic receptor. J. Biol. Chem. 272, 24279-24286.
Shoyab, M., McDonald, V.L., Bradley, J.G., and Todaro, G.J. (1988) Amphiregulin: a bifunctional
growth-modulating glycoprotein produced by the phorbol 12-myristate 13-acetate-treated human
breast adenocarcinoma cell line MCF-7. Proc. Natl. Acad. Sci. USA. 85, 6528-32.
Shoyab, M., Plowman, G.D., McDonald, V.L., Bradley, J.G., and Todaro, G.J. (1989) Structure and
function of human amphiregulin: a member of the epidermal growth factor family. Science 243, 10741076.
160
References
Simard, J.M., and Watt, D.D. (1990) Venoms and toxins. In: The biology of scorpions (Gary, A.P. ed.)
pp 414-444, Stanford University Press, California.
Sixma, T.K., and Smit, A.B. (2003) Acetylcholine binding protein (AChBP): a secreted glial protein
that provides a high-resolution model for the extracellular domain of pentameric ligand-gated ion
channels. Annu. Rev. Biophys. Biomol. Struct. 32, 311-34.
Smit, A.B., Syed, N.I., Schaap, D., van Minnen, J., Klumperman, J., Kits, K.S., Lodder, H., van der
Schors, R.C., van Elk, R., Sorgedrager, B., Brejc, K., Sixma, T.K., and Geraerts, W.P. (2001) A gliaderived acetylcholine-binding protein that modulates synaptic transmission. Nature 411, 261-268.
Srinivasan, K.N., Gopalakrishnakone, P., Tan, P.T., Chew, K.C., Cheng, B., Kini, R.M., Koh, J.L.,
Seah, S.H., and Brusic, V. (2002) SCORPION, a molecular database of scorpion toxins. Toxicon 40,
23-31.
Stoesser, G., Baker, W., van den Broek, A., Camon, E., Garcia-Pastor, M., Kanz, C., Kulikova, T.,
Lombard, V., Lopez, R., Parkinson, H., Redaschi, N., Sterk, P., Stoehr, P., and Tuli, M.A. (2001) The
EMBL nucleotide sequence database. Nucleic Acids Res. 29, 17-21.
Strydom, D.J. (1977) Snake venom toxins. The amino-acid sequence of a short-neurotoxin homologue
from Dendroaspis polylepis polylepis (black mamba) venom. Eur. J. Biochem. 76, 99-106.
Sudhof, T.C. (1995) The synaptic vesicle cycle: a cascade of protein-protein interactions. Nature 375,
645-653.
Sur, C., Fresu, L., Howell, O., McKernan, R.M., and Atack, J.R. (1999) Autoradiographic localization
of alpha5 subunit-containing GABAA receptors in rat brain. Brain Res. 822, 265-270.
Sur, C., Quirk, K., Dewar, D., Atack, J., and McKernan, R. (1998) Rat and human hippocampal alpha5
subunit-containing gamma-aminobutyric Acid A receptors have alpha5 beta3 gamma2
pharmacological characteristics. Mol. Pharmacol. 54, 928-933.
Sutcliffe, M.J., Dobson, C.M. and Oswald, R.E. (1992) Solution structure of neuronal bungarotoxin
determined by two-dimensional NMR spectroscopy: calculation of tertiary structure using systematic
homologous model building, dynamical simulated annealing, and restrained molecular dynamics.
Biochemistry 31, 2962-2970.
Tamiya, N. and Yagi, T. (1985) Non-divergence theory of evolution: sequence comparison of some
proteins from snakes and bacteria, J. Biochem. (Tokyo) 98, 289-303.
Tan, N.H. (1983) Isolation and characterization of two toxins from the venom of the Malayan cobra
(Naja naja sputatrix). Toxicon. 21, 201-207.
Tan, P.T., Khan, A.M., and Brusic, V. (2003) Bioinformatics for venom and toxin sciences. Brief.
Bioinform. 4: 53-62.
Tatemoto, K., Rokaeus, A., Jornvall, H., McDonald, T.J., and Mutt, V. (1983) Galanin - a novel
biologically active peptide from porcine intestine. FEBS Lett. 164, 124-128.
161
References
Tateno, Y., Miyazaki, S., Ota, M., Sugawara, H., and Gojobori, T. (2000) DNA data bank of Japan
(DDBJ) in collaboration with mass sequencing teams. Nucleic Acids Res. 28, 24-26.
Tindall, R.S., Kent, M., Baskin, F., and Rosenberg, R.N. (1978) Nicotinic acetylcholine receptor of
mammalian brain: naja toxin binding to subcellular fractions of rat brain, J. Neurochem. 30, 859-863.
Toyama, M.H., de Oliveira, D.G., Beriam, L.O., Novello, J.C., Rodrigues-Simioni, L., and
Marangoni, S. (2003) Structural, enzymatic and biological properties of new PLA(2) isoform from
Crotalus durissus terrificus venom. Toxicon 41, 1033-1038.
Troyer, K.L., and Lee, D.C. (2001) Regulation of mouse mammary gland development and
tumorigenesis by the ERBB signaling network. J. Mammary Gland Biol. Neoplasia. 6, 7-21.
Tsai, I.H., Liu, H.C. and Chang,T. (1987) Toxicity domain in presynaptically toxic phospholipase A2
of snake venom. Biochim. Biophys. Acta 916, 94-99.
Tsai, I.H., Hsu, H.Y., and Wang, Y.M. (2002) A novel phospholipase A(2) from the venom glands of
Bungarus candidus: cloning and sequence-comparison. Toxicon 40, 1363-1367.
Tsetlin, V. (1999) Snake venom alpha-neurotoxins and other ‘three-finger’ proteins. Eur. J. Biochem.
264, 281-286.
Tu, A.T. (1973) Neurotoxins of animal venoms: snakes, Annu. Rev. Biochem. 42, 235-258.
Tyler, M.I, Barnett, D., Nicholson, P., Spence, I., and Howden, M.E. (1987) Studies on the subunit
structure of textilotoxin, a potent neurotoxin from the venom of the Australian common brown snake
(Pseudonaja textilis). Biochim. Biophys. Acta 915, 210-216.
Tytgat, J., Chandy, K.G., Garcia, M.L., Gutman, G.A., Martin-Eauclaire, M.F., van der Walt, J.J., and
Possani, L.D. (1999) A unified nomenclature for short-chain peptides isolated from scorpion venoms:
alpha-KTx molecular subfamilies. Trends Pharmacol. Sci. 20, 444-447.
Utkin, Y.N., Kukhtina, V.V., Kryukova, E.V., Chiodini, F., Bertrand, D., Methfessel, C. and Tsetlin
V.I. (2001) "Weak toxin" from Naja kaouthia is a nontoxic antagonist of alpha 7 and muscle-type
nicotinic acetylcholine receptors. J. Biol. Chem. 276, 15810-15815.
Valdivia, H.H. and Possani, L.D. (1998) Peptide toxins as probes of ryanodine receptor. Trends
Cardiovas. Med. 8, 111-118.
Van der Kloot, W., and Molgo, J. (1994) Quantal acetylcholine release at the vertebrate neuromuscular
junction. Physiol. Rev. 74, 899-991.
van Praag, H., Schinder, A.F., Christie, B.R., Toni, N., Palmer, T.D., and Gage, F.H. (2002)
Functional neurogenesis in the adult hippocampus. Nature 415, 1030-1034.
Velluti, J.C., Caputi, A. and Macadar, O., (1987) Limbic epilepsy induced in the rat by dendrotoxin, a
polypeptide isolated from the green mamba (Dendroaspis angusticeps) venom. Toxicon 25, 649–657.
162
References
Villar, M.J., Cortes, R., Theodorsson, E., Wiesenfeld-Hallin, Z., Schalling, M., Fahrenkrug, J., Emson,
P.C., and Hokfelt, T. (1989) Neuropeptide expression in rat dorsal root ganglion cells and spinal cord
after peripheral nerve injury with special reference to galanin. Neuroscience 33, 587-604.
Vrontakis, M.E. (2002) Galanin: a biologically active peptide. Curr Drug Target CNS Neurol. Disord.
1, 531-541.
Vrontakis, M.E., Torsello, A., and Friesen, H.G. (1991) Galanin. J Endocrinol. Invest. 14, 785-794.
Vrontakis, M.E., Yamamoto, T., Schroedter, I.C., Nagy, J.I, and Friesen, H.G. (1989) Estrogen
induction of galanin synthesis in the rat anterior pituitary gland demonstrated by in situ hybridization
and immunohistochemistry. Neurosci. Lett. 100, 59-64.
Wang, C.Y., Tan, Z.Y., Chen, B., Zhao, Z.Q., and Ji, Y.H. (2000) Antihyperalgesia effect of BmK
IT2, a depressant insect-selective scorpion toxin in rat by peripheral administration. Brain Res. Bull.
53, 335-338.
Weber, M., and Changeux, J.P. (1974) Binding of Naja nigricollis (3H) alpha-toxin to membrane
fragments from Electrophorus and Torpedo electric organs. I. Binding of the tritiated alpha-neurotoxin
in the absence of effector. Mol. Pharmacol. 10, 1-14.
Whiting, P.J. (1999) The GABA-A receptor gene family: new targets for therapeutic intervention.
Neurochem. Int. 34, 387-390.
Whiting, P.J (2001) Pharmacological subtypes of GABA-A receptors based upon subunit composition.
In: GABA in the Nervous System: A View at Fifty Years (Martin, D.L. and Olsen, R. W., eds) pp 113126, Lippincott, Williams and Wilkins.
Wiesenfeld-Hallin, Z., Xu, X. J., Hughes, J., Horwell, D. C., and Hokfelt, T. (1990) PD134308, a
selective antagonist of cholecystokinin type B receptor, enhances the analgesic effect of morphine and
synergistically interacts with intrathecal galanin to depress spinal nociceptive reflexes. Proc. Natl.
Acad. Sci. USA. 87, 7105-7109.
Wittwer, C.T., Hermann, M.G., Moss, A.A., and Rosmussen, R.P. (1997) Continuous fluorescence
monitoring of rapid cycle DNA amplification. Biotechniques 22, 130-138.
Wolf, K.M., Ciarleglio, A., and Chiappinelli, V.A (1988) kappa-Bungarotoxin: binding of a neuronal
nicotinic receptor antagonist to chick optic lobe and skeletal muscle, Brain Res. 439, 249-258.
Xu, Z.Q., Shi, T. J., and Hokfelt, T. (1996) Expression of galanin and a galanin receptor in several
sensory systems and bone anlage of rat embryos. Proc. Natl. Acad. Sci. USA. 93, 14901-14905.
Yang, C.C. (1994) Structure and function of cobrotoxin. J. Toxicol. Toxin Rev. 13, 275-290.
Yasuda, O., Morimoto, S., Jiang, B., Kuroda, H., Kimura, T., Sakakibara, S., Fukuo, K., Chen, S.,
Tamatani, M. and Ogihara, T. (1994) FS2. a mamba venom toxin, is a specific blocker of the L-type
calcium channels. Artery 21, 287-302.
163
References
Zamudio, F.Z., Conde, R., Arevalo, C., Becerril, B., Martin, B.M., Valdivia, H.H., and Possani, L.D.
(1997a) The mechanism of inhibition of ryanodine receptor channels by imperatoxin I, a heterodimeric
protein from the scorpion Pandinus imperator. J Biol. Chem. 272, 11886-11894.
Zamudio, F.Z., Gurrola, G.B., Arevalo, C., Sreekumar, R., Walker, J.W., Valdivia, H.H., and Possani
LD. (1997b) Primary structure and synthesis of Imperatoxin A (IpTx(a)), a peptide activator of Ca2+
release channels/ryanodine receptors. FEBS Lett. 405, 385-389.
Zeller, E.A. (1977) Snake venom action: are enzymes involved in it? Experientia 30, 143-150.
Zhang, F.T, Xi, Z.S., and Qi, Y.X. (1987) A preliminary research of the antineoplastisitic effects
induced by Buthus martensi Karsch of chinese drug: I. Observation of the effect on mice with tumor.
Journal of Wannan Medical College 1, 1-5.
Zhang, X., Nicholas, A. P., and Hokfelt, T. (1995) Ultrastructural studies on peptides in the dorsal
horn of the rat spinal cord--II. Co-existence of galanin with other peptides in local neurons.
Neuroscience 64, 875-891.
Zhang, X., Nicholas, A.P., and Hokfelt, T. (1993a) Ultrastructural studies on peptides in the dorsal
horn of the spinal cord--I. Co-existence of galanin with other peptides in primary afferents in normal
rats. Neuroscience 57, 365-384.
Zhang, X., Verge, V. M, Wiesenfeld-Hallin, Z., Piehl, F., and Hokfelt T. (1993b) Expression of
neuropeptides and neuropeptide mRNAs in spinal cord after axotomy in the rat, with special reference
to motoneurons and galanin. Exp. Brain Res. 93, 450-461.
Zhang, Z.W., Vijayaraghavan, S., and Berg, D.K. (1994) Neuronal acetylcholine receptors that bind
alpha-bungarotoxin with high affinity function as ligand-gated ion channels, Neuron 12, 167-177.
Zhou, X.H., Yang, D., Zhang, J.H., Liu, C.M., and Lei, K.J. (1989) Purification and N-terminal partial
sequence of anti-epilepsy peptide from venom of the scorpion Buthus martensii Karsch. Biochem J.
257, 509-17.
Zigova, T., Pencea, V., Wiegand, S.J., and Luskin, M.B. (1998) Intraventricular administration of
BDNF increases the number of newly generated neurons in the adult olfactory bulb. Mol. Cell
Neurosci. 11, 234-245.
164
[...]... catalase, amylase and B-glucosaminidase Table 1.2: Some of the common components in snake venoms of different species 1.3 Neurotoxins The first amino acid sequence of a neurotoxin, Tx α from Naja nigricollis was reported in 1967 (Eaker and Porath, 1967) Since then, a large number of neurotoxins have been isolated and characterized Reports on the amino acid sequences of a number of homologous neurotoxins... structural plasticity of the three fingers, the three-finger fold is also amenable to a variety of overt and subtle deviations, such as the number of the β-strands present, size of the loops and C-terminal tail as well as twists and turns of various loops, all of which may have great significance with respect to functional diversity and selectivity of molecular targets (Servent and Menez, 2001; Kini,... Introduction 1.5.4 Kappa neurotoxins κ-Neurotoxins form a new family of snake venom neurotoxins that are structurally related to long neurotoxins With the exception of κ-cobrotoxin, all the other κ-neurotoxins identified consist of 66 amino acid residues and on the basis of amino acid alignment, contain five disulphide bonds as in long neurotoxins (Figure 1.5D) The first κ -neurotoxin, κ-bungarotoxin... paired modified salivary glands which in most venomous snakes are located superficially beneath the scales in the posterior part of the head and eyes The gland is linked to the fang by a duct Contraction of muscles around the gland compresses the gland, forcing the flow of venom along the duct to the fang where the size of these structures depends on the size and species of the snakes 1 Introduction... other subtypes of nAChRs such as neuronal nAChR 1.5.3 Short- and long-chain neurotoxins Based on the length of their polypeptide chains, α-neurotoxins were initially classified as shortchain α-neurotoxins that have 60 – 62 residues and four conserved disulfide bonds in common positions, between Cys-3 and Cys-24, Cys-17 and Cys-45, Cys-40 and Cys-61, Cys-62 and Cys-68 (Figure 1.5A) These neurotoxins bind... patterns of all the neurotoxins 77 Figure 3.6 Classification of snake NTX based on structure, function and phylogenetic information 81 Figure 3.7 Phylogenetic trees performed using parsimony analysis of all the mature svNTX 90 Figure 3.8 Competitive binding studies of 125I-α-Bgt and native Bc-ntx4 to nAChR of Torpedo receptor 91 Figure 3.9 Output of the annotation tool in svNTXs 92 Figure 4.1 Separation of. .. homologous neurotoxins from snakes such as cobras, kraits, mambas and sea snakes have been added to this growing list of neurotoxins At present, more than 100 neurotoxin amino acid sequences are known and they form one of the largest families of protein with known primary structures (Endo and Tamiya, 1991) 5 Introduction Neurotoxins are capable of blocking nerve transmission by binding specifically to nicotinic... of a “broken-neck” syndrome reported after bites of snake with neurotoxic venoms (Minton, 1990) 8 Introduction 1.4 Classification of neurotoxins Snake neurotoxins can be divided into two groups: presynaptic or postsynaptic neurotoxins depending on their mode of action Presynaptic neurotoxins are either phospholipase A2 enzymes or contain these enzymes as an integral part of the neurotoxin complex and, ... arthropods and chordates These venomous animals produce a variety of toxins to defend themselves from predators, to subdue their prey and for digestion Therefore, animal toxins have immobilizing effects and killing functions towards a wide variety of creatures Snake venoms have attracted much medical attention since ancient civilizations and had been used in medical treatment for thousands of years In... structure and α-cobratoxin, 2.4-Å-crystal structure To date, more than 100 three-finger α-neurotoxins have been isolated and sequenced from Elapidae and Hydrophiidae snakes Depending on their amino acid sequence and tertiary structures, α-neurotoxins can be classified into short-chain, long-chain, kappa and weak neurotoxins Although the primary target of all these categories of the three-finger neurotoxins ... Professor Kandiah Jeyaseelan for his dedicated supervision, constant encouragement and continued support during the course of this project I am also grateful for the assistance of Associate Professor... Institute of Infocomm Research for his kind help in the construction of the neurotoxin database My sincere appreciation also goes out to Dr Arunmozhiarasi Armugam for her valuable input and advice... scales in the posterior part of the head and eyes The gland is linked to the fang by a duct Contraction of muscles around the gland compresses the gland, forcing the flow of venom along the duct to