• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/323

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

323 Cards in this Set

  • Front
  • Back
3 Components of a nucleotide
phosphate, sugar, nitrogenous base
features of ribonucleotide sugar
A pentose (5 carbon) sugar with carbon atoms numbered 1'C to 5'C
difference between RNA and DNA sugar
RNA has an OH group attached to the 2'C, DNA has only H
which bases are purines
adenine and guanine
which bases are pyrimidines
cytosine, thymine (and uracil)
purines and pyrimidines, which are 2 ring strucures
purines
where do sugar and base join and what type of bond
1'C of the sugar by a N-glycoside linkage
where do sugar and phosphate join and what type of bond
5'C of the sugar by an ester linkage
how many phosphates on a nucleotide
1-3
what is the structure of a nucleoside
nucleotide without phosphate groups, only sugar and base.
where do nucleotides join and what type of bond
through sugar and phosphate
groups by phosphodiester bonds at the 3'OH to the alpha phosphate group
what type of reaction joins nucleotides
condensation reaction
direction of nucleotide joining
5' to 3'
what are needed for nucleotides to join
free 5' phosphate group or a free 3'OH
what will a DNAchain with 5' polarity have
a free 5' phosphate
what will a DNA chain with 3' polarity have
a free 3' OH
what are bonds joining DNA strands
hydrogen bonds between anti-parallel chains
how do A and T bond between DNA chains
2 H bonds
how do C and G bond between DNA chains
3 H bonds
4 things required for DNA synthesis
template strand, primer sequence, free, dNTP's, DNA polymerase
features of a DNA primer
upto 20 nucleotides, complementary to template, free 3'OH
what happens as a dNTP is attached to growing DNA chain
complementary dNTP arrives, DNA polymerase catalyses 3'OH nucleophilic attack of alpha phosphate on incoming dNTP, phosphodiester bond formed and extra pyrophosphate released
preparation steps for DNA synthesis
unwinding, separation, primer binds
direction of DNA synthesis
5' to 3'
what consequence does ribose sugar rather than 2'-deoxyribose sugar have in RNA
2' to 5' bonds can form as well as 3' to 5', succeptible to alkaline hydrolysis at high pH = less stable
what structures tend to form when RNA forms double strands
hairpin loops within a single strand
hierarchical structure of chromosome
nucleosome of 8 histone proteins with DNA wound round.
in eukaryotes where does protein biosynthesis occur
cytoplasm
stages in protein biosynthesis
transcription to mRNA, goes to cytoplasm, translated to protein
transcription requires
RNA polymerase, free NTP's, NO PRIMER, template DNA, promoter, terminator.
where does translation occur
ribosomes
what happens at translation
ribosome binds 5' end of mRNA and moves along until AUG start codon, tRNA attached to AA binds to AUG, tRNA leaves and next codon is 'read' and polypeptide chain forms until stop codon when it is released.
direction of RNA synthesis in transcription
5' to 3'
how many total codons and what do they code
64, 3 stop, 61 encode 20 AA's
what is RNA processing
pre-RNA has 5' cap and 3' poly-A tail added, introns removed and exons spliced to make mRNA
where does RNA processing occur
nucleus
6 points where genes can be regulated
• Initiation of transcription
• Elongation of transcription
• mRNA modification
• mRNA stability and transport
• Translation
• Protein stabilit
at what stage does most gene regulation occur
control of transcription
what does RNApol1 do
transcribes rRNA
what does RNApol2 do
transcribes mRNA
what does RNApol3 do
transcribes tRNA
how does RNA polymerase bind to promoter
accessory proteins, the general transcription factors.
what binding would repress RNA polymerase transcription
The binding of a transcriptional repressor to a silencer site would block RNA polymerase binding
what binding would enhance RNA polymerase transcription
binding of a transcriptional activator to an enhancer site
what may regulate enhancers and silencers of transcription
intra- and extra-cellular signals.
hormones, growth factors, stress signals (e.g. increased temperature), or nutritional
signals (e.g. glucose levels or a lack of a particular amino acid).
2 important domains of transcriptional activators
DNA binding domain, activation domain
what bonds are cut by endonucleases
phosphodiester bonds
what are free on the cut ends of DNA after digestion with a restriction enzyme
3'OH and 5'P
how may DNA be treated to protect from endonuclease digestion
methylation at recognition site
types of restriction enzyme used in recombinant DNA tech and features of it.
type 2, recognises 4-6 bp palindrome
what restriction enzyme will tend to make more cuts, a 4 bp recognition sequence one or a 6 bp
4, more chance of this sequence occuring
3 types of ends produced by restriction enzyme digestion
sticky (5' or 3' overhang) and blunt
how are restriction fragments separated
gel electrophoresis
what causes restriction fragments to move and separate
charge (overall negative charge due to P groups) causes travel towards +ve pole and separation by molecular size
features of an agarose gel
They allow the separation of DNA fragments typically between ~100bp and 20kb, large pores, organic
features of a polyacrymalide gel
smaller, more homogeneous pores, They allow the separation of shorter DNA fragments (1-1000bp range), good resolution (1bp discrimination)
3 determinants of separtion on gel by electrophoresis
charge, size, shape
what factor influences restriction fragment separation and why
size, charge and shape are constant due to double helix shape
what is necessary to estimating restriction fragment sizes
DNA size ladder
what is used to stain restriction fragments separated on gel
ethidium bromide (fluoresces in UV light)
what is used to join DNA fragments
DNA ligase
what does DNA ligase do
reforms DNA breaks by reforming the phosphodiester bond
necessities of DNA fragments for ligation
identical overhangs (or blunt ends) free 3'OH and 5'P. must be dsDNA
what is recombinant DNA
cut and ligated DNA from different sources
2 types of DNA cloning
cell-based and cell-free
cell based DNA cloning
recombinant DNA cloned, usually in e Coli
cell free DNA cloning
PCR
3 features of a cloning vector
origin of replication, selectable marker, cloning site
origin of replication
in a cloning vector- allows vector to replicate independent of host
selectable marker of vector
allows survival of only transformed vectors, most common is B lactam resistant e coli infected by ampicillin resistant vectors, B lactamase cleaves these drugs
onecloning site on vector
a single site for inserting DNA using specific restriction enzyme, only on recognition site
4 types of vectors
1. Bacterial plasmids
2. Bacteriophage vectors
3. Cosmids
4. Artificial chromosomes
features of plasmids
circular dsDNA, have an origin of replication, multiple cloning sites for different restriction enzymes, recombinant marker -typically a insertional inactivation (lac Z causes untransformed vectors to produce blue ecoli colonies)
2 methods of transformation
electroporation- bacteria washed, plasmid added and pulses of electricity creates small pores. chemical- at low temp CaCl2 added to bacteria, plasmid added, short 42 degree heat shock, membrane transiently disrupted
most commonly used phage vector
lambda phage
how does cloning with lambda replacement vector happen
15kb section of phage genome removed (stuffer fragment), desired dna ligated into phage, recombinant phage mixed with ecoli and plated together with uninfected ecoli, cell lysis and infection cascade, plaque formed.
how are untransformed phages identified/eliminated
without stuffer fragment phage genome is too small to survive so only those with an insert of 12-20 kb survive.
features of cosmid vectors
upto 45kb inserts, act as plasmids but infected in a phage head, collonies not plaques form.
yeast artificial chromosomes
hybrid of plasmid and yeast dna can accept 1mb inserts.
artificial chromosome types
yeast and bacterial
bacterial artificial chromosomes insert size
up to 300 kb
max plasmid insert
10kb
max phage insert
20kb
max cosmid insert
45kb
max BAC insert
300kb
max YAC insert
1MB
4 possible ligation products in non-directional cloning with a plasmid vector
untransformed vector, uninserted circularised insert, transformed plasmid with insert in one of 2 directions.
why might direction of insert insertion into a plasmid be important
if a protein product expressed by insert is desired then 5' end of insert must be next to promoter
directional cloning
2 restriction enzymes with non-complementary sticky ends, a small section of plasmid is excised but restriction sites should be close together to minimise this
why might direction cloning be impossible and how would desired ligations be achieved in this situation
lack of suitable restriction sites, alkaline phosphatase treatment of linearised plasmid suppresses self-ligation by removing 5'P, this prevents DNA ligase from joining ends, untreated insert is still inserted and the missing P nicks are later repaired by bacterial repair systems
2 enzymes involved in blunt end ligation and their purpose
5'-overhanging ends filled in by Klenow fragment of E.coli DNA polymerase I, 3'-overhanging ends trimmed by 3'→5' exonuclease activity of T4 DNA polymerase
T4 DNA polymerase does what
3'→5' exonuclease activity, trims 3' overhang
Klenow fragment of E.coli DNA polymerase I does what
fills in 5' overhang
5 requirements for PCR
excess of dNTP's, template DNA, primers to bind 3' end and provide free 3' OH, heat stable DNA polymerase, magnesium ions
3 steps of PCR and temperatures
1. DNA denatured at 95°C (unwinds and H bonds break)
2. primers annealed at 55°C
3. primer extension at 72°C, 5' to 3'
where is PCR carried out
a thermocycler
what will a PCR control involve
a parallel run with no template
after PCR is finished what happens to product
gel electrophoresis separation versus a DNA ladder
2 principle limitations of PCR
only fragments up to 2kb, sequence of flanking region must be known to design primers
differences in first 2 cycles of PCR
original template will produce complementary strands with overhang, therafter only this will occur once with every cycle, defined fragments will have a primer at each end and will exponentially produce defined product.
determinants of good primer design for PCR
1. length 18-24 nucleotides
2. base composition, lots of G and C, more stable due to extra H bonds, avoid long runs of single nucleotides
3. primer melting temps, make both primers melt at similar temps, affected by length and base composition
4. primer binding sequence but not occur elsewhere in DNA
5.primers that do not form secondary structures or self anneal (no complementary ends)
how to stop low temperature non-specific low temperature primer extension by Taq polymerase
hot start, Ab conjugated DNA polymerase that is inactivated
what are the consequences of Taq polymerase lacking exonuclease activity
no proof reading, introduction of error that will be propagated to differing amounts depending on which cycle they are introduced in.
low Mg2+ in PCR will cause
low polymerase activity
too high Mg2+ in PCR causes
non-specific annealing, multiple PCR prducts
3 ways manipulation of PCR can be used to insert suitable restriction sequences for directional cloning
1. attaching a restriction site sequence to the 5' ends of the PCR primers, only the 3' end may anneal but the restriction sequence will be copied into PCR product.
2. TA cloning, Taq polymerase tends to attach an extra single nucleotide to the 3' end on PCR product (terminal transerase activity creates a tiny sticky end, most commonly A), linearised vector will bind if it has a 3' T overhang, this can be created by treatment of linearised vector with Taq polymerase and dTTP's.
3. Pfu DNA polymerase produces blunt ended DNA that may be inserted to blunt ended vector or TA cloning can be used.

2&3 are non-directional
detection of genetic alterations involving a deletion, insertion or rearrangement by PCR
using primers to flank the regionPCR amoplification id performed and compared to a control by electrophoresis separation, different length in disease, 2 different lengths in diseased sample if heterozygous.
detection of point mutation by PCR
allele-specific PCR, requires the mutation to be known, 2 PCR's run each with a primer specific for mutated or wild type forms (3' ends terminate at point mutation), only correctly base paired primer will extend, a second pair of primers that flank the mutated region are used as a control to ensure the PCR process is working.
example of allele specific PCR detection
K-ras oncogene mutations in colorectal cancer detected in faecal DNA, specific point mutations known to occur early at codon 12 and 13, useful diagnostic tool
diagnostic uses of PCR
genetic mutations separated by length v's control, point mutations detected by allele specific PCR, viral or bacterial detection of infection eg TB faster than culture or Ab
what is the starting template material for RT PCR
a single stranded piece of RNA
2 stages of RT PCR
1. RT reaction. primer 1 complementary to 3' end of desired mRNA, reverse transcriptase extends this to a complementary cDNA strand

2. PCR. in cycle 1 primer 2 extends sscDNA to ds. thereafter primer 1 and 2 extend both strands
2 clinical/research uses of RT PCR
diagnostic eg HPV specific mRNA products & in research to determine the transcription of specific genes under
different experimental conditions
basic principal of real time PCR
new DNA produced in each cycle is detected as it is produced, number of cycle until cycle threshold - Ct is reached is udes to calculate either the absolute or relative quantity of starting mRNA/cDNA
how is measurement done in real time PCR - 2 methods
special thermocycler that can detect fluorescent product
1. SYBR Green fluorescent dye will fluoresce only when bound to dsDNA.
2. fluorescent reporter probes e.g. TaqMan probes, probe is designed to bind specifically to the target DNA sequence
between the two PCR primers, it has a 5' fluorescent reporter dye and a 3' quencher. At annealing stage it binds to ssDNA and at extension stage the e 5'→3' exonuclease activity of Taq DNA polymerase chops off the quencher so fluorescence occurs.
how is PCR used in DNA sequencing
dideoxy chain termination
what is needed to perform dideoxy chain termination PCR
a high fidelity DNA polymerase, only one primer, dNTP's and labeled ddNTP's, template.
how do dNTP and ddNTP's differ
ddNTP is 2' 3' di-deoxyribose and has a 3' H rather than OH
how is the product of dideoxy chain termination PCR read
run on polyacrymalide gel (gives one nucleotide resolution), each possible length of temple fragment will have been copied with a fluorescing ddNTP at the end which is scanned and is complemtary to the initial template.
2 types of DNA library
1. genomic DNA library composed of fragemtns of DNA inserted into plasmid or phage λ that constitute the entire genome including regulatory regions, introns and noncoding areas.
2. cDNA library, cDNA fragments corresponding to all mRNA present in a cell at sampling this is both tissue- and developmental stage- specific
4 steps in constructing a genome library
1. Isolation of genomic DNA, usually from WBC
2. Fragmentation by partial digestion to produce random, ovelapping similar sized fragments.
3. isolation and ligation, gel separation and extraction then vector ligation
4. Plating out, vectors infect usually ecoli, with plasmid, cosmid and BAC vectors each colony represent a fragment with phage it is a plaque.
what does a genome library look like
A genomic library consists of a large number of agar plates containing colonies or
plaques, each of which carries a different insert of genomic DNA
what factors determine how many colonies are needed to represent a genome in a genome library
size of genome and size of vector insert
Construction of a cDNA Library
1. isolation of mRNA, using
oligo-dT's to isolate polyadenylated mRNA
2. ss cDNA synthesis using reverse transcriptase and oligo-dT primer to bind poly A tail, treatment with alkali NaOH to destroy RNA but leave DNA.
3. ds cDNA synthesis, to allow one primer to bind all cDNA present Homo-polymer tailing is done by incubating with terminal transferase and one dNTP (eg C) which adds a poly C tail to 3' end without a primer. A oligo-dG primer can then be used to bind all.
4. ligation into vector
5. plating out.
genome library screening by nucleic acid hybridisation
1. colony lift onto nitrocellulose filter
2.treated to lyse the bacteria
3.alkali (NaOH) to denature the dsDNA
4. hybridised with a radioactively-labeled probe
5. autoradiography detects colony that binds to desired probe.
how to screen a genome library using a probe from another species
a homologous probe is totally specific and will be used at higher temperature and lower salt concentration (high stringency) a heterologous probe needs lower temperature and higher salt concentrations in hybridisation and washes but the sequence homology should be sufficient to still get a match between eg mouse and human
to screen acDNA library if the protein product is known but not the DNA sequence
back-translation from AA sequence of the protein to DNA (several codons encode most AA's so must use all possible) this is a degenerate probe -all possible
nucleotide sequences.
choose a section of at least 6 AA's preferably with methionine and tryptophan (only one codon each) and use as a probe. Run at high stringency to eliminate partial matches.
immuno screening a cDNA library
antibody raised to cDNA protein product, λ vectors used so nitrocellulose filter placed on plaque, ecoli lysed in plaque so proteins in plaque transfer to filter, incubated with primary antibody, washed, secondary labeled eg anti-rabbit ab applied and read.
what is southern blotting used for
analyse complex mixtures of DNA fragments
procedures of southern blotting
1. genomic DNA digested to fragments by restriction enzyme
2. separated by gel electrophoresis
3. NaOH to denature the dsDNA fragments on gel.
4. ssDNA fragments blotted onto nitrocellulose membrane (blotting solution drawn up blotting paper by cappilary action, through gel and carries fragments onto membrane to make transfer of gel)
3. fragemnts baked or UV onto membrane
4. membrane hybridized with a labeled probe complementary to area of interest
5. exposed to X-ray film to visualise
what information can be obtained from a southern blot
• The size of the fragment that binds to the probe.
• The copy number of a particular genomic sequence (i.e. the number of copies
that are present). For example, a large genomic sequence may be absent or
amplified (present in greater number) in cancer cells.
• Deletions, insertions or large scale rearrangements (e.g., a translocation)
within the genomic fragment of interest.
• The presence of specific ‘forei
how can a DNA probe be labeled
5' P removed from probe by alkaline phosphatase and radioactive phosphate added by polynucleotide kinase
function of northern blotting
analyse complex mixtures of RNA.
type of probe used in northern blotting
cDNA
northern blotting proceedures
1. total cellular RNA removed
2. separated by electrophoresis with formaldehyde to prevent secondary structure formation
3. stained with ethidium bromide (intact RNA sample will have sharp bands that correspond to
the two large rRNA molecules)
4. transferred and fixed to nitrocellulose membrane as in Southern blot.
5. probed with labeled
6. xray visualisation cDNA
information from northern blot
1. The size of a specific mRNA molecule (transcript).
2. The relative abundance of a particular mRNA
3. Expression (transcription) patterns between tissues or developmental stages
function of Western blotting
analyse complex mixtures of protein.
probe type used in western blotting
Ab
steps in Western blotting
1. protein extracted
2. sodium dodecyl sulphatepolyacrylamide gel electrophoresis (SDS-PAGE) separation
3. transferred onto a nitrocellulose membrane by
electro-blotting
4. Ab probe
5. secondary Ab
6. visualisation
information from western blot
size of protein, If a protein has been modified e.g. phosphorylated (will alter size, or detection by Ab to phosphorylated form), relatie protein abundance, expression pattern between tissues or developmental stages.
what is the transcriptome
all the mRNA molecules (transcripts) of a particular cell population under specific conditions.
oligonucleotide
a short sequence od bp's commonly synthetic in origin
What is a Microarray?
an ordered arrangement of DNA molecules immobilised usually on a glass slide. cDNAs, PCR fragments or oligonucleotides. each spot contains multiple copies of a short stretch of DNA
preparation for using a microarray
mRNA reverse transcribed to ss cDNA using reverse transcriptase, an oligo-dT primer, and fluorescently-labelled dNTPs
how is microarray analysis quantitative
each spot contains millions of copies and will not be saturated by hybridisation with probe so degree of fluorescence indicates relative abundance. This is relative quantity.
what is an expression profile
the results of a microarray analysis which shows the mRNA (DNA expression levels) of thousands of genes in a cell population
comparative microarray analysis
comparison of expression profile of 2 cell populations eg cancerous and not or different developmental stages
how are the expression profiles of 2 cell populations compared on one microarray
One of the mRNA populations is reverse transcribed into green fluorescing cDNAs by incorporating dNTPs labelled with the green fluorescent marker the other red. equal amounts hybridised and washed off then imaged. spots will be either unstained, red, green or yellow (both stain)
what broad uses may microarray analysis have clinically
diagnosis (versus known disease profiles), prognosis (versus known staging examples), effectiveness of therapy (retesting)
Serial Analysis of Gene Expression (SAGE)
cDNA tags unique for each gene, ligated, cloned, sequenced
creating SAGE
mRNA removed and RT into cDNA, tag created for each cDNA, tags ligated, put in vector, cloned and sequenced, software analysis of sequence counts number of times a tag occurs
use of SAGE
usually comparison of 2 cell populations
size of a tag for SAGE
10-14 bases
meaning of SAGE results
The number of times a particular tag is identified corresponds to the abundance of its parental cDNA in the starting mRNA. 2 cell populations comparable.
what is the principal difference between SAGE and microarray
Sage is based on sequencing, microarray is based on hybridisation. Therefore SAGE can detect previously unknown genes, microarray can only detect what is know.
what are the limitations of mRNA analysis
although most gene control is done at the level of transcriptional control so mRNA well represents cell status proteins that require post translational modifications or genes regulated post translationally are not discriminated
proteomics
is the large-scale study of proteins, particularly their structures, functions and interactions.
number of estimated human genes
30000
number of estimated human proteins
500000
by what means are more proteins produced than genes exist
alternative promoters, polyadenylation sites, alternative splicing, post-translational phosphorylation or glycosylation
2-dimensional polyacrylamide gel electrophoresis (2D PAGE)
Proteins from a particular cell sample can be extracted and separated into more than 1,000 discrete protein spots by two-dimensional polyacrylamide gel electrophoresis
2D PAGE method
Proteins are separated by net charge in one dimension and by size in the second dimension.
problems with 2D PAGE
not sensitive enough to detect rare proteins, and many proteins will not be resolved on the gel
how are spots generated by 2D PAGE analysed
mass spectrometry
protein identification following 2D PAGE
1. protease digestion to produce peptides (trypsin)
2. peptides identified by mass spectrometry giving a unique peptide mass fingerprint

identification only possible if protein is previously known
how does mass spectrometry detect post-translational modifications
phosphorylation etc will distinctively alter mass in a detectable way.
method for analysing protein-protein interactions
Yeast 2-hybrid analysis (Y2H)
what is learned by finding protein protein interactions
may indicate function especially regulatory networks or signal transduction cascades
what is the DNA binding domain
region of a protein that recognizes and binds to a specific recognition sequence in DNA
what is the activation domain
protein region that interacts with other proteins to activate transcription of the target gene.
basic Y2H set up
1. yeast transcriptional activator, GAL4 binds to a GAL4-recognition sequence with its DBD just in front of its target gene promoter and activates transcription with its AD.
1. GAL4 positioned beside HIS3 gene (only activation by GAL4 will produce histidine thus allowing yeast to grow on histidine free medium.
3. hybrids of 2 proteins to be tested attached to either GAL4 DBD or GAL4 AD are created by recombinant plasmids
4. when vectors inserted into yeast products produced and GAL4 DBD binds to GAL4 recognition sequence but AD will only attach if 2 proteins interact.
creating hybrid proteins for Y2H
inserting the cDNA of the DBD and the cDNA of protein X into a vector so that it can express a hybrid protein then the same with protein Y and cDNA of GAL4 AD
how to use Y2H analysis to screen a cDNA library for protein interactions
protein of interest is hybridised to GAL4 DBD in bait plasmid. All cDNA in library hybridised with GAL4 AD cloned into prey plasmids and translated to produce hybrid protein protein. Bait and prey transformed into yeast and selectable marker used to identify only yeast containing both. Yeat growing on histidine-free medium indicated protein-protein interaction has occurred. Prey plasmid then isolated and sequenced to identify.
Gene knock-outs
genetically engineered mice with a specific gene removed, the effect can then be observed
Gene knock-down advantages
1. can study essential genes
2. can be applied to most eukaryotes including cultures
3. can be a more accurate model of disease
what is RNA interference (RNAi).
small non-coding dsRNA target complementary mRNA transcripts for destruction so regulate gene expression.
how does RNA interference (RNAi) work. 4 steps
1. dsRNA is chopped up by the enzyme Dicer into shorter fragments known as short interfering RNAs
2. incorporated into a multi-protein complex - RNA-induced silencing complex (RISC).
3. ds siRNA then becomes single stranded and one of the strands (the one complementary to the mRNA) guides the RISC to the target mRNA
4. The ss siRNA binds to the mRNA and directs the RISC to degrade the mRNA
how are genes knocked down
siRNA oligos introduced to bind and destroy specific mRNA. This is done in culture.
Recombinant protein production
an expression vector used to produce recombinant protein
uses of recombinant proteins
1. industrial production eg insulin
2. analysis of protein function, can introduce protein with known mutations or overexpress.
where are recombinant proteins made
ecoli or yeast, insect or mammal cells
what would cause a recombinant protein to need to be made in eukaryotic cells
1. requires post-translationally glycosylated (ecoli cannot do this)
2. prokaryotic proteins may be misfolded
3. if protein is toxic to bacteria
2 types of gene therapy
1. germ cell, changes may be inherited, illegal in humans
2. somatic
challenges of gene therapy
1. must be delivered to specific target cells
2. must be inserted in the DNA so that it will be expressed (transcribed and translated) permanently and stably at a level which is of benefit
2 somatic cell gene therapy strategies
1 ex vivo
2 in vivo
ex vivo somatic gene therapy
patients cells removed, altered and reintroduced. must be able to be cultured (eg stem cells in BM), good for targeting
in vivo somatic gene therapy
gene delivered direct into body, very hard to target, only option for lung epithelial cells, which have been targeted for therapy of cystic fibrosis
targeting in vivo somatic gene therapy
viral vectors
problems with viral vectors
targeted by immune system, reversion to virulence
types of viral vector and features
1. retroviruses: RNA genome is converted into dsDNA by the retroviral enzyme reverse transcriptase and stably integrates into host. can only infect dividing cells,only small gene inserts due to stability problems,Random integration of virus carries the risk of inactivating genes at the site of insertion.
2. Adenoviruses, DNA, can infect dividing and non-dividing cells, larger inserts, do not integrate with host genome so repeat delivery required.
Non-viral gene therapy
liposomes
what are liposomes and what is their function in potential therapy
tiny lipid bubles, may be able to deliver DNA for gene therapy, fuse with cell membranes, not target of immune system, large DNA inserted, no infection risk, modifying bubble may allow targeting, most lost prior to arrival at target, weak and transient action
cancer gene therapy
introduce several genes into tumour cells. aim to kill cells, slow growth, cause expression of a protein to attract immune response, sensitise to chemo and radio therapy. Also target immune cells to induce sensitivity and action against tumour cells.
external oncogenic stimuli
smoking, radiation, sunlight and various chemicals, viruses, baceria
3 basic stages of cancer progression
1. initiation of a first genetic change
2. promotion, clonal expansion and accumulation of further mutations
3. progression, uncontrolled proliferation, switch from benign to cancerous
example of a tissue, mutation type and source of radiation damage
ionising radiation eg Xray, DNA strand breaks, Production of ROS, causes Base damage, Deletions, Transitions, Transversions. Skin cancer
example of a tissue, mutation type and source of chemical damage
Polycyclic Aromatic hydrocarbons from Combustion of organic
compounds cause DNA adducts, Transversions, Frameshifts & Point mutations causing Lung cancer
example of a tissue, mutation type and source of viral damage
Human Papilloma virus a DNA tumour virus causes Inappropriate gene expression & Cervical cancer
internal oncogenic stimuli
spontaneous mutation
1.DNA replication errors eg frameshift insertion/deletion, point mutations
2.chemical modification of bases eg deamination or alkylation
3. mutation by metabolism products. reactive oxygen species (superoxide, hydroxyl radical, and hydrogen peroxide), these can also be external stimuli. Antioxidants eg vitamin C and E are defense
what is a missense mutation
frameshift causing wrong AA to be made
nonsense mutation
frameshift resulting in stop codon
what is a transition mutation
a point mutation when a pyrimidine is replaced by a pyrimidine or a purine by a purine e.g. G-C becomes A-T
what is a transversion mutation
a point mutation which occur when a purine replaced by a pyrimidine or vice versa (e.g. A-T becomes T-A or C-G)
what is deamination
amine group on a base is replaced by carbonyl group, this can alter the base type eg C turned to U
what can remove U bases from DNA
U-DNA-glycosidase
partial internal partial external ongogenic stimuli
metabolism products of drugs etc ingested Xenobiotics
how are Heterocyclic amines (HCAs) linked to cancer
bbq meat
action of UV on DNA
cross-links between adjacent pyrimidines, most frequent are cyclobutane pyrimidine dimers (CPDs), usually formed between adjacent Thymine bases, pull bases closer distorting DNA and increasing errors at subsequent replication
what are (6-4) photoproducts
Dimers formed by a single covalent bond between the 6 position of one pyrimidine and the 4 position of the adjacent pyrimidine on the 3' side, distorts DNA leads to errors
what are cyclobutane pyrimidine dimers
cross links usually between adjacent thymine bases, distrots DNA leads to errors
5 types of DNA repair
•Repair by Alkyl Transferase (AT)
•Base Excision Repair (BER)
•Nucleotide Excision Repair (NER)
•DNA mismatch repair
•Recombinational Repair
what is a DNA adduct
DNA covalently bonded to carcinogen
what is alkyl transferase
a form of DNA repair where alkylated bases are repaired by transferring the alkyl group to a cysteine residue in this this enzyme
base excision repair. 5 steps
repairs oxidative damage, often spontaneous
1. N-glycosidase removes damaged base
2. endonuclease cuts 5' end of abasic site
3. DNA polymerase B cuts 3' abasic end
4. DNA polymerase B fills site
5. DNA ligase seals ends
nucleotide excision repair corrects what type of damage
complicated, multi-protein process to repair DNA lesions eg UV CPDs and 6-4 photoproducts.
5 basic steps in nucleotide excision repair
1. Detection of DNA damage is carried out by XPA, XPC, RPA
2. helix opened and TF-IIH (Transcription Factor IIH) macromolecule extends single strand
3.The DNA is cleaved on either side of the lesion by XPF & XPG
4. gap is filled in by DNA Polymerase ε, PCNA (Proliferation Control Nuclear Antigen)
5. DNA ligase seals chain.
NER damage detection
XPA, XPC, RPA (Replication Protein A)
NER strand separation
TF-IIH (Transcription Factor IIH). This composes of 6 subunits including XPB, XPD, ERCC
NER DNA cleavage
XPF & XPG
NER gap filling and sealing
DNA Polymerase ε, PCNA (Proliferation Control Nuclear Antigen) and the pieces ligated by DNA ligase
2 types of NER
transcription coupled repair (TCR) and global genomic repair (GGR)

TCR ensures that transcribed strand damage is repaired first.
differences in proteins used in TCR NER and GGR NER
TCR uses CS-A and CS-B to recognise damage not XPC
cockayne syndrome is caused by
CS-A or CS-B defects result in functional GGR NER but defective TCR NER
DNA MISMATCH REPAIR (MMR), 3 steps
wrong base has been inserted
1. MSH2:MSH6 heterodimer binds to mismatched base
2. MLH1:PMS1 heterodimer binds MSH2:MSH6, exonuclease cleaves strand
3. gap filled by DNA polymerase and DNA ligase
recombinational repair
a mutated section of dna (eg pyrimidine dimer) is skipped in replication and the gap is filled using the normal strand as a template. This is not repair but a way to tolerate damage.
what is the mutator phenotype
inactivity or deficiency of genes involved in repairing damaged DNA making the cell more error-prone and lead to increased mutation rate in other genes. These cells then outcompete neighbours.
4 main stages of cell cycle
G1, S, G2, M
what happens at G1
Growth and preparation of the chromosomes for replication
what happens at S
(synthesis) Replication of DNA
what happens at G2
Preparation for mitosis
what happens at M
(mitosis) Division of cell into 2 daughter cells
what is the function of cyclin dependant kinases
catalyse the phosphorylation of other proteins, thereby activating them. This regulated the cell cycle
what are the principal regulators of G1
cyclin D & E
how is a a CDK activated and produce its action
binds with cyclin, activated by phosphorylation, this activates a transcription factor by removing an inhibitor. This induces gene transcription for cell cycle progression and also next CDK and cyclin genes
RB example of CDK system
p16 INK4a inhibits cyclin D1/CDK4 complex, if uninhibited D1/CDK4 hyperphosphorylates pRB in the pRB/E2F complex, this dissociates activating the transcription factor E2F which stimulates transcription of various growth genes eg cdc2, myc. This activates other transcription factors eg Fos, jun in a cascade.
if erros occur in the cell cycle what are used to halt progression, examples of these
cyclin dependant kinase inhibitors, P21 family
key cell cycle checkpoints
•before the cell enters S phase (a G1 checkpoint)
•during S phase
•after DNA replication (a G2 checkpoint).
proto-oncogene
a normal gene that can become oncogenic if mutated
5 ways a proto-oncogene may become and oncogene
•Retroviruses
•Protein-Protein interaction (from DNA viruses)
•Chromosome translocation
•Gene amplification
•Point mutation
2 modes of retrovirus activation of proto-oncogene to oncogene
transduction- host genome gene is incorporated into virus and produced (acute transforming viruses)
transfection - the virus inserts its genome into the host genome (non-acute transforming viruses)
what are long terminal repeats (LTR)and what is their role in oncogenic activation
retroviral genomic elements that enhance gene expression. If a host gene is transfected into a retrovirus it may upregulated by viral LTR and if viral RNA is converted to DNA and inserted into host genome the viral LTR may act on host genes
DNA viruses in oncogene activation
viral proteins may bind host proteins preventing action eg HBV E7 protein binds host Rb (tumour suppressor) so preventing it from inhibiting E2F transcription factor
chromosome translocations in oncogene activation, 2 modes of action
1. increase normal product, if a proto-oncogene is translocated to be influenced by a strong promoter
2.expression of combined product, chimeric protein
example of chromosome translocation increasing normal product to induce oncogene
burkitts lymphoma, c-myc proto-oncogene on C8 is translocated to C14 and influenced by Ig gene enhancer
example of chimeric protein causing oncogenic action
CML, Philadelphia gene, c-abl on C9 combines with BCR on C22. The BCRabl chimeric product is a tyrosine kinase homologous to abl but with mutant qualities
gene amplification and oncogen activation
multiple copies of a proto-oncogene develop in chromosome due to defective start signalling in replication, copies are called double minute chromosomes. if all of these are transcribed then protein product will be greatly amplified
double minute chromosomes
multiple copies of a proto-oncogene that may lead to amplified transcription and so protein translation
homogenous staining regions
a chromosome region containing double minute chromosomes, gene copies that were amplified aberrantly
example of gene amplification in oncogen activation
HER-2 is over expressed in some breast cancers, this receptor protein-tyrosine kinase is overexpressed and binds epidermal growth factor, which stimulates cell proliferation. Target of Herceptin
point mutations in oncogen activation
single base pair mutation that alters protein enough to change function
example of oncogenic point mutation
RAS protein involved in signal transduction cascades, point mutated forms of the ras gene produce RAs protein that is not deactivated and constant stimulation drives cell cycle progression
5 classes of oncogen
•Secreted Growth factors
•Cell surface growth factor receptors
•Components of intracellular signal transduction system
•DNA binding gene regulatory proteins
•Components of cell cycle : cyclins/CDKs/CDKIs
secreted growth factor oncogene example
c-sis overexpression leads to overabundance of platelet derived growth factor -sarcomas and gliomas
Cell surface growth factor receptor oncogene example
erbB gene encodes EGF receptor, when mutated the usual phosphorylation does not inactivate so permanently on signalling for growth and division. Breast cancer
Components of intracellular signal transduction system as oncogene- example
ras, when mutated activated as normal by GTP binding but GTPase does not work on it so permanent activation, phosphorylation of downstream protein kinases inducing proliferation. Found in many cancers
Transcription factors as oncogenes, example
myc, fos, and jun TF's mutated forms mean growth factor stimulation is not required to induce transcription.
Components of cell cycle: cyclins/CDKs/CDKIs as oncogenes, example
D-type cyclins regulate progression through the early stages of G1. Cyclin D1 produced by oncogene PRAD-1
features of apoptosis
controlled programmed cell death, shrinkage, nuclear condensation, blebbing, fragments into vesicles, phagocytosed by macrophages
2 important proteins in apoptosis
bcl-2 and bax in balance. If bcl-2 levels become elevated it is encouraged to continue proliferating. Conversely, if bax levels are elevated the cell is encouraged to die
example of apoptotic pathway mutation causing cancer
follicular cell lymphoma, bcl-2 is over expressed due to translocation so is increased relative to bax and proliferation is promoted.
example of oncogenic teamwork
C-myc transcription factor and ras signalling protein, if bothmutated cancer is nearly 100% assured
how can tailored changes be made to a genome, 2 stage process
engineered endonucleases can cut dsDNA at desired point and DNA fragments containing th edesired change to the sequence are also inserted to the nucleus, these are then used as templates for filling in the gap by homology-directed repair (HDR)
knudson model
2 hit model of cancer progression, both copies of a gene must be knocked out or mutated
classic 2 hit example
retinoblastoma, an inherited recessive loss-of-function mutation to Rb on C13
proportion of cancers with mutated p53
60%
features of p53
TSG on C17, often deleted in cancers
most common type of somatic mutation of p53
point mutation
inherited p53 mutation is commonly found as what disease
Li-Fraumeni Syndrome, usually inherited one mutated allele and other then deleted. exhibit a wide range of tumours, including breast, brain, leukaemia and sarcomas.
function of p53
genomic guardian, the main player in the complex pathways that mediate cell cycle control and DNA repair
effect of DNA damage on p53
normal 20 minute half life is extended as it is stabilised so accumulates
how is p53 normally controlled
mdm2 binds p53 targeting it for ubiquitinylation and so destruction, also inhibits p53 activity as an activator of transcription.
how does DNA damage lead to p53 build up
amino terminus is phosphorylated so association with mdm2 does not happen
how is p53 in an autoregulatory system and how is this mutated
p53 activates the expression of its own regulator mdm2, mutated p53 this does not happen
3 biological activities of p53
1) Induction of G1 arrest and Suppression of Tumour cell growth
2) Induction of Apoptosis
3) Differentiation of B-cells
p21 function at G1 checkpoint
cell cycle inhibitor which works by binding and inactivating the cyclin/CDK dimers that regulate the cell cycle
GADD45 function at G1 checkpoint
GADD45 protein inhibits DNA synthesis by binding PCNA and also promotes DNA excision repair. Over-expression of GADD45 suppresses cell growth.
function of p53 in G1 arrest and tumour growth suppression
1. cell cycle checkpoint protein. in response to damage activates the expression of several genes including GADD45, and p21 at G1 checkpoint.
2. Activated in response to over-expression of gene products from myc, ras, jun oncogenes to check unregulated proliferation
function of p53 in inducing apoptosis
activation induces transcription of bax and inhibits bcl2 expression thus changing the ratio and inducing apoptosis
function of p53 in B cell differentiation
not fully known but aids development to mature form
6 biochemical functions of p53
1) Regulation of DNA synthesis
2) Sequence-Specific DNA binding
3) Transcriptional Activation
4) Transcriptional Repression
5) Promotion of DNA re-naturation and Strand Transfer
6) Translational Control
p53 in regulation of DNA synthesis
binds DNA adjacent to origin of replication blocking attachment of initiation proteins, prevents helicase unwinding, binds and inactivates replication factor RPA.
Sequence-Specific DNA binding by p53
most p53 mutations occur in the DNA binding domains
p53 Transcriptional Activation
N-terminal domain of p53 is a strong transcriptional activator of genes with a p53-binding motif on their promoter. Rb1, GADD45, EGF, creatine kinase, mdm2 and p53
p53 Transcriptional Repression
repress the transcription of growth promoting genes- c-fos, c-jun and c-myc. normal levels do not suppress but when elevated by DNA damage it does.
p53 Promotion of DNA re-naturation and Strand Transfer
catalyze re-naturation of complementary strands of ss DNA
p53 Translational Control
associate with the cellular translation machinery eg TGF-β mediates down-regulation of CDK4 synthesis at the translational level and TGF-β controlled by p53
functional p53 response to DNA damage detection
stabilised, lengthened half life acts to halt cell cycle and induce DNA repair at G1 or G2 checkpoint, if repaired then cycle continues, if not then apoptosis initiated
how may p53 be involved in failure of chemotherapy and radiotherapy
chemotherapy and radiotherapy aim to cause just enough cellular damage to activate the in-built cellular mechanism for over-expression of p53 and so apoptosis, this will not work if p53 itself is mutated.
p53 as therapy
in cancers where p53 is mutated chemo and radio therapy will be less effective so adenovirus vector introduction of unmutated p53 is being trialed
are oncogenes or TSG associated with hereditary cancers
TSG, think RB
what is the distinction between a benign tumour and a malignancy
is it confined within a capsule
at what stage are oncogenic genetic alterations likely to occur and why
mitogenesis (the induction of mitosis), short time i not allow sufficient time for repair. Also ss DNA is more sensitive to damage
how can epigenetics affect tumour development
DNA methylation, transcriptional activation or translational control may all control gene expression eg DNA methylation silences genes (perhaps TSG), if this process is disturbed TSG may be overly silenced or correct silencing of proto-oncogenes may not occur
example od a compound that is thought o affect epigenetics
folic acid, B12
what happens in the promotion phase of tumour development
clonal expansion of originally mutated cell, further mutations develop (especially if original mutation confers competitive advantage) replication becomes less accurate.
what natural process may act to stall growth or induce apoptosis in a growing tumour mass
central hypoxia induces p53
features of malignant cells
incompletely differentiated, rapid growth, invasive, loss of capsule, prone to metastasis
what cell feature makes cells a target of chemotherapy drugs and what is the problem with this
rapid division, means that chemo drugs also affect normally rapid dividing cells eg hair and GIT
stages of metastasis
1. detachment
2. release of angiogenic factors
3. dissemination
4. establish new foci
Detachment
first stage of metastasis development. loss of CAM's eg e-cadherin. normally proliferation requires adherence to ECM, this is Anchorage dependance.
what is anchorage dependance
cells must normally be correctly adhered to the ECM to proliferate
2 types of cellular adherance must be lost for detachment
1. intercellular adhesion
2. adhesion with cell and ECM
how do cell normally attach to basement membrane
integrins
how may a tumour cell break adherance to basement membrane yet still be able to adhere again at a new loci
tumour cell produces lamanin and lamanin receptor with lamanin already bound.Lamanin has heparin and collagen binding sites on it allowing ECM binding. To move Type IV collagenase is released by tumour cell breaking lamanin-collagen bond
what is anchorage independence
normally detached cells have inactivated regulatory protein Cyclin E-CDK2 which stalls growth, in tumour cells oncogenes relay false signals to the nucleus maintaining Cyclin E-CDK2 activity
what is Cyclin E-CDK2
a growth regulatory protein that normally stalls growth by inactivating in detached cells. This action is maintained in metastasising tumour cells, anchorage independence. Normal function involves dissociating Rb protein from transcription factor that initiates transcription of cell cycle proteins
how do tumour cell in the centre of a mass obtain oxygen
release of angiogenic factors
how is angiogenesis normally controlled
p53 controls transcription of TSP-1 angiogenic inhibitor thrombospondin 1, loss of P53 removes this control, seen in Li-fraumeni
4 modes of tumour dissemination
blood, lymph, locally, across bod cavities
haematogenous metastasis
tumour releases matrix metalloproteases MMP that break down ECM and basement membranes travel in blood, aggregate platelets and lymphocytes, attach to wall and after further protease breakdown extravasation
what is intravasation
tumour breakdown of blood vessel wall to enter
what is extravasation
tumour release of proteases to break down vessel wall ECM of blood vessels to leave circulation
how do tumour cell survive in the blood
must attach to the lining of the blood vessels and aggregate platelets and lymphocytes to protect from killer T cells and increase adhesion. this aggregate then travels and re-adheres elsewhere
what is a carcinoma and how does it usually disseminate
epithelial cancer usually with lymphatic metastasis
what is a sarcoma and how does it usually disseminate
connective tissue cancer usually with haemaotologenous metastasis
why are the lungs common points for metastasis
the first point of contact for vasculature leaving many tissues is the lungs
what factors are thought to underly the preference of some tumour types for certain tissues for metastasis, example
the concentrations of growth factors and hormones and surface receptors eg prostate cancer and bone marrow,
tumour staging
TNM method
what do TNM stand for and how is it scored
T- primary tumour site
N - extent of lymph node involvement
M - extent of metastasis

each scored 1-4
what are metastatic factors and an example
patterns of gene expression within tumour cells that are predictive of metastatic spread eg HER-2 in breast cancer, many p53 mutations lead to agressive and invasive cancers
XERODERMA PIGMENTOSUM
severe sunlight sensitivity, 2000x skin cancer risk. UV-induced lesions in DNA are inefficiently removed or not at all
XERODERMA PIGMENTOSUM genetic cause
nucleotide excision repair (NER) pathway defect, 7different types - A-G
cockayne syndrome
inability of cells to carry out transcription-coupled repair (TCR). so although general NER is functional errors occur specifically at transcribed regions