Techniques Flashcards

1
Q

Compare the sizes of bacterial and eukaryotic genomes

A

Bacterial 0.6-10Mb
Eukaryotic 10-100,000 Mb

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What are the steps of Sanger sequencing?

A

Denature DNA
Anneal the primer
Chain extension in presence of a limited number of radioactively labelled dideoxy NTPs (ddNTP)
Incorporation of ddNTP stops DNA strand extension (OH on 3’ of sugar is replaced with H)
E.g. if T is on the template strand, occasionally ddA will be added and chain will terminate. All the products of varying lengths end with an A
Radioactivity can be detected by X-ray film following polyacrylamide gel electrophoresis
Reading sequence from bottom up gives sequence from primer into unknown DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Explain modifications to Sanger’s method

A

Thermal Cycle Sequencing - The original method uses the template once as removing E.coli polymerase kills it - Use of thermostable polymerases meant the reaction could be recycled and reused
Improving Fragment Separation - Using capillary separation means smaller amount of DNA needed, more consistent, faster run times
Fluorescent Detection - Original method meant 4 reactions required for one section of DNA. GE of Taq enzyme allowed ddNTPs labelled with fluorophores means one reaction, detect which light peak produced by each dye as they fall out the tube

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Pros and Cons of Sanger sequencing

A

Pros:
- Cheap per experiment (~£4) - 1 sequence
- Very good single read accuracy (99.9%)
Cons:
- Need to know the first primer
- High cost per million bases (£4000)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What Is The Process Of Shotgun Sequencing?

A

Break DNA into pieces (enzymatic or physical)
Clone pieces and sequence both ends
Lots of reactions in parallel means lots of fragments that need to be put together and have overlapping sequences that look similar
These are aligned in contiguous sequence (contigs)
Higher number of overlapping sequences means higher chance it is the correct assembly
Bigger pieces are easier to be put together
Interspersed repeats prevent contig expression, Tandem repeats cause contig compression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is the concept of coverage in relating to DNA sequencing and how accurate is it?

A

1x coverage = having sequenced DNA of equivalent length to genome size (not all sequenced, some will repeat what’s already sequenced)
Even at 10x coverage, sequence will have many gaps - even 1 small section can make sequence unusable

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Explain Illumina Sequencing

A

Most dominant next gen sequencing method
1. Prepare genomic DNA by smashing it to bits and adding adapters to ends using ligases
2. Attach DNA to surface of glass slide - sparsely bind ss fragments randomly to inside surface of flow cell channels
3. Bridge amplification by PCR reaction - unlabelled nucleotides and enzyme
4. Fragments become ds - bridges on solid phase substrate
5. Denature ds molecules - ss templates anchored to substrate
6. Complete amplification - several dense clusters of dsDNA (all DNA in cluster is the same) generated in each channel of flow cell
7. Add 4 labelled reversible terminators, primers and polymerase, reaction stops due to terminator - wash of unincorporated nucleotide and polymerase
8. Image 1st base by laser excitation, detect fluorophore to see what was incorporated, cleave dye and unblock 3’ OH
9. Sequence over multiple cycles determines bases one at a time
10. Align data and compare to reference

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Pros And Cons Of Illumina Sequencing

A

Cons:
- Not as simple to apply as Sanger
- Set up is expensive (£1-20k per experiment)
- Limit to fragment size (~500bp) – have to be short enough to generate a small cluster and not reach across to other side of the glass chip
- Limit to how many times you can repeat the chemical cycle (up to 300b)
Pros:
- Capable of generations 10s to 1000s of million sequences in parallel
- Cheap cost per million bases (2-20p)
- Very good single base accuracy (99.9%)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Explain Nanopore Sequencing

A

Individual nanopores embedded in synthetic membrane
Voltage creates current through pore
Molecules passing through affect magnitude of current
Loop adapter joins 3’ end of one strand to 5’ end of other strand
5’ adapter provides binding for enzyme which feeds DNA into pore (slows DNA down)
Ss moves through pore
Different DNA causes different deflection of current

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Pros And Cons Of Nanopore Sequencing

A

Pros:
- Not sequencing by synthesis – don’t have to worry about number of cycles or if polymerase falls offf
- Can process 100s of bases per second
- Can also sequence modifications to DNA/RNA
- Sequencing can continue until nanopore blocks to membrane breaks down
- In theory, can sequence extremely long fragments of DNA
Cons:
- Much noisier than sequencing 1000s of copies at once as nucleotides don’t go through the pore one at a time - approx. 5 bases in pore at a time so (45) 1024 possibilities
- Single read accuracy is much lower than Sanger/Illumina (90% but improving)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are the methods of measuring mRNA Levels?

A

Northern Blotting
Quantitative Reverse Transcriptase PCR
RNA Sequencing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Explain Northern Blotting

A

Separate RNA molecules by size by agarose gel electrophoresis then transfer RNA to supportive membrane
Add labelled ssDNA which hybridises to cmRNA
Wash off unbound DNA
Amount of bound prove proportional to amount of mRNA in sample
Can use labelled probe to see levels of specific transcripts
Complex target, simple probe
Shows size as well as amount
Test level of one transcript at a time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Explain qRT-PCR

A

Regular PCR repeated denaturation (94C), annealing of primers (55-65C) and DNA synthesis (72C) amplifies in exponential fashion
RT produces cDNA from RNA, RT tends to snap back at end, making hairpin loop
Endpoint amount depends on available resources not necessarily amount of input material
Transforming volume to log scale means you can trace back to original amount. observe at which point DNA amount crosses threshold
Can detect amplified DNA by using DNA-binding dyes (e.g. SYBR Green I) – Unbound dye low fluorescence; bound dye high fluorescence- fluorescence increases as more dsDNA is produced – works with any amplicon but can’t distinguish between real product and mis-priming

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Explain RNA-Seq

A

With NGS, alternative is to sequence all cDNA; under ideal conditions, number of sequences arising from each cDNA will be proportional to the level of mRNA
Allows analysis of all mRNAs (transcripts) at once – transcriptomics (not measuring transcription) – lots of power – much more expensive than single-gene/mRNA measurement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Why are there difficulties in measuring protein levels?

A

Proteins cannot be generically isolated from cells based on chemical characteristics
Generally aren’t linear in natural state so won’t run neatly in gel, don’t have consistent charge
Can’t use binding of complementary sequences to detect

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Explain Western Blotting

A

Separate proteins by size by SDS-PAGE (sodium dodecyl sulfate) which has negative charge and binds to protein evenly, keeping it linear
SDS overpowers any natural charge
Use charge to send through polyacrylamide gel mesh
Transfer denatured protein to supporting membrane e.g. nitrocellulose
Block non-specific sites on membrane
Incubate with primary antibody and wash
Incubate with secondary antibody (with marker e.g. fluorophore) and wash and detect by horse radish peroxidase by chemiluminescence
Reagent = H2O2 and luminol
HRP catalyses breakdown of H2O2 to H2O and O2
Luminol is oxidised and emits photon of light, detected on film/camera

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How can protein tagging be used as an alternative to making a specific antibody?

A

Fuse CDS for protein of interest to a genetically-encoded tag e.g. GFP
Can follow as it goes along the cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Recombinant Protein Production - what does a generic plasmid look like?

A

Coding Sequence (CDS) for protein
DNA encoding protein of interest often dropped onto MCS
Machinery around the gene
DNA to enable replication of plasmid (ori - specific to bacteria)
Need to be able to select for bacteria that have taken up the plasmid e.g. resistance to drug most common

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Using lac operon

A

Contains genes for lactose metabolism, operator (lacO), regulatory gene for repressor (LacI)
Lactose binds to repressor protein which falls off operator and allows operon to be transcribed
Can use to make toxic product combining machinery from lac operon and polymerase from bacteriophage
Isopropyl beta-D1-thiogalactopyranoside = non-metabolisable analog of lactose
When IPTG is present in hose cell, it causes lac repressor dissociation and allows T7 RNA polymerase to initiate target gene transcription

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

How to purify proteins?

A

Create fusion of CDS with DNA encoding a protein tag; expressed as one protein (tag must be in same RF); can then use properties of tag to purify protein of interest
Many exist but one of simplest and most used is His tag (6x histidine residues)

21
Q

Explain His-tag purification

A

Poly-histidine residues bind to Nickel ions (Ni2+)
Passing crude lysate from bacteria (mix of proteins) over beads attached to Ni2+
His-tagged protein sticks to nickel, others flow through
After washing – elute His-tagged protein with competitor (imidazole – histidine analogue)
All other proteins fall through and we’re left with purified protein of interest that can be injected or used for a reaction

22
Q

Directed approaches of altering protein function

A

Make 2 primers (containing mismatch mutations) running in opposite directions around plasmid
Remove unmutated template by digesting with Dpnl (cuts at adenine methylase sites)
Transform mutated molecule into competent cells for nick repair
Version of this approach can be used to make lots of different mutations
Need to close circles before transformation

23
Q

Mutagenic Approaches of altering protein function

A

Harness what nature does - random rare mutations and selection of function from population
Slightly poison polymerases to force greater mutation rate - biased dNTP mixture, increased Mg2+, addition of Mn2+ (but don’t want to do this to entire plasmid)
Error prone PCR (lots of random mutated copies)
Clone fragments into plasmid
Express mutant proteins and select for function

24
Q

Explain Gibson assembly as a method to assemble DNA fragments

A

Relatively long dsDNA fragments with overlapping sequences at the ends
T5 exonuclease shortens 5’ ends
ssDNA strands at ends anneal and T5 exonuclease becomes inactive
Thermostable DNA polymerase (e.g. Taq) fills in gaps and DNA ligase seals nicks

25
Q

What is the difference between nothern blots and dot blots?

A

Northern blots: complex target, simple probe, quantitative, shows length as well as amount, test level of one transcript at a time
Dot blots: Simple target, complex probe, quantitative, one mRNA at a time but can easily increase number of targets

26
Q

Explain how microarrays can be used

A

Lots of dot plots miniaturised on a chip or slide
Use 2 dyes, control and experiment hybridised to same slide
E.g. Control mRNA labelled with green fluorescent dye and Mutant mRNA labelled with red fluorescent dye – hybridise onto DNA chip – laser scanner – Dots can be green (control-specific), ret (mutant-specific) or yellow (common to both)

27
Q

Explain how RNA-Seq can be used

A

RT mRNA into cDNA, sequence lots of these sections and align reads to genome
Estimate of mRNA levels from number of reads mapping to each gene but count from each RNA depends on how much sequencing we have done (normalise data for sequencing depth)

28
Q

How can ChIP-Seq be used as a functional genomics tool?

A

Chromatin-ImmnuoPrecipitation Sequencing
Chemically link DNA to bound proteins, crosslink, fragment and use antibodies to enrich for DNA bound by specific protein then reverse crosslinking and sequence
Peaks in mapped reads show where protein was bound to DNA at time of cross-linking e.g. can find binding motifs, promoter sequences, open/closed chromatin etc.

29
Q

How can morpholinos be used to investigate gene function?

A

Synthetic RNA analogues can perform base pairing with modified backbones and can form hybrids with DNA/RNA without being degraded
morpholine ring where ribose was
Phosphorodiamidate morpholino oligomers can be used to block translation (upstream of AUG) or alter splicing (force more/less by blocking splice donor or acceptor sequence)
Need to be introduced into cells e.g. microinjection of eggs of zebrafish, xenopus

30
Q

What is RNA interference (RNAi)?

A

Mechanism for post-transcriptional silencing of genes by patrolling for presence of dsRNA
Long dsRNA cleaved into short siRNAs by Dicer enzyme
Each siRNA unwound into 2 ssRNA (incorporated into RISC)
Guide strand pairs with complementary mRNA and induces cleavage by Argonaute
Target transcript is destroyed
If we can directly introduce siRNA into cells/organisms that target mRNA, we can hijack machinery to selectively degrade/switch off expression of all copies of a specific gene without removing genes from genome

31
Q

What are the two methods of inducing RNAi?

A
  1. Exogenous Source - microinjection, electroporation, in food (C. elegans)
  2. Endogenous Production - introduce genetic material encoding dsRNA
32
Q

What is pooled screening?

A

Lots of short hairpin RNAS, add barcode
Mix and package in lentiviral vector to infect lots of cells
grow cells, induce RNAi, apply pressure
Knocking down specific genes causes better/worse growth
sequence barcode of each cell which tells us how much of each cell we have
Can analyse abundance of each individual RNAi in population by NGS screening of barcode
Don’t have to test thousands of individual RNAis separately - make one library

33
Q

What are some examples of cell lines?

A

NIH-3T3 mouse embryonic fibroblasts
CHO Chinese Hamster Ovary epithelial cells (commonly used for production of recombinant proteins where bacterial expression makes wrong final product)
hTERT-RPE1 -Retinal Pigment Epithelial cell line, transformed by expression of human telomerase

34
Q

Transgene expression and shuttle vectors

A

Structure is similar in organisation to bacterial expression vectors but different sequences and selection markers.
Vectors get very big very quickly as many things are required to grow them in bacteria
Mammalian promoters are complex and transcription may require multiple TFs, mediator proteins and activators so are much too big to put into plasmids

35
Q

Inducible system: Tet-on

A

Tetracycline repressor (TetR) bind to Tetracycline operator (TetO)
Presence of Tetra- or Doxycycline releases TetR from TetO so transcription occurs
Mammalian polymerase isn’t very sensitive and just codes through proteins in the way (e.g. Chromatin has loads attached all the time)

36
Q

Inducible systems: Tet-off

A

Can use VP16 (T activator from herpes simplex vector)
DNA recruits Oct-1 which recruits VP16 which recruits transcription machinery to fire gene
Can attach TetR to VP16 - protein activates transcription when bound to promoter and no activation in presence of tetracycline

37
Q

Describe two chemical methods of transfection

A
  1. Calcium Phosphate:
    Cells washed in phosphate buffer, DNA added and precipitated using CaCl2, precipitate internalised by endocytosis
  2. Liposome-mediated:
    Put DNA into Liposome to form Lipoplex which is endocytosed into the cell (forms Endosome)
    Most of the time it’s taken straight into lysosomal pathway and degraded by cell degradation pathway
    Occasionally endosome bursts, releasing DNA into the cell
38
Q

Describe a physical method of transfection

A

Electroporation:
Cells are exposed to transient electric field, causing small pores to form in PM
DNA can enter
Pores can reseal once electrical field is removed

39
Q

Describe Adenoviral transduction

A

Do not integrate into host genome
dsDNA packaged into a capsid and endocytosed
Capsid proteins break off, stimulate burst and release of virion from package
Protein covering DNA specifically designed to carry DNA into nucleus
Machinery circumvents normal system and make lots of protein to make more viruses
Can remove parts responsible for infection and add transgenes
However people tend to have a strong immune response to adenoviruses

40
Q

Describe adeno-associated viral transduction

A

ssDNA, non-enveloped viruses with very small genome
Elicit very weak immune response
Good at infecting non-dividing cells
Can’t take host machinery to replicate
Most are episomal (DNA maintained as circular extrachromosomal element)

41
Q

Describe retroviral transduction

A

ssRNA or dsDNA viruses
Use RTase to make cDNA and integrate into host genome
e.g. HIV, human T-cell lymphotropic virus
Main issue is that they permanently modify most genomes
Separation of virus packaging genes from genetic material to be transferred means they can’t replicate in host cell - Viral vector and foreign gene transfected into packaging cell line that expresses viral RTase (pol), capsid (gag) and surface and transmembrane (env)  allows production of viral particles containing vector sequences (as RNA) which can infect target cells but once inside cannot replicate

42
Q

What are lentiviral plasmids?

A

Virus + Bacterial plasmid + Mammalian plasmid
Can do modifications in bacteria and shuttle into mammalian cell line
example is pLentiCRISPR
Retroviral genome, transgenic genes to be introduces into target cells (+selection marker), bacterial resistance genes

43
Q

Describe how transgenic mice are created

A

Isolate ESCs from trophoblast
GM ESCs put into different embryo and transplanted into surrogate
Offspring contain mix if wild type cells and chimera GM cells
Mate chimera type with wild type (some offpsring will have one full GM allele - hetero)
Crossing of heterozygotes to hopefully get offspring homozygous for knockout (only if mutant isn’t lethal)

44
Q

Describe Cre/Lox recombination

A

Cre = site specific recombinase which recognises LoxP sites and creates Holliday junctions between them, causing switches of DNA between the sites
If LoxP sites are in the same orientation, a section is excised. If the sites are opposite, a section is inverted
If we make LoxP insertions into the genome, we can then change when/where LoxP insertions manifest by controlling expression of Cre with a tissue specific promoter

45
Q

Describe how you would breed a transgenic mice for Cre/Lox system

A

A = homozygous mouse containing cre recombinase under control of a tissue specific promoter
B = homozygous mouse containing LoxP flanking target
Breed A and B together
F1 mice are hetero so would only delete one copy of the gene when expressed
Backcross F1 mice with parental homozygous floxed mice
F2 25% inherit Cre and 2 copies of floxed gene
Expression of Cre, both genes deleted

46
Q

What are Zinc Finger Nucleases and how do they work?

A

Zinc finger domains are found in lots of DNA-binding proteins - different domains have different specificity depending on residues in alpha helix - most bind to 3-6bp of DNA
Zinc Finger Nucleotide is a fusion of multiple Znf domains with Fokl nuclease domain
ZFN one bind upstream, one downstream
Two domains come together to cut any location in human sized genome (theory)
Realistically, specificities of Znf overlap and recognition depends on context of surrounding Znf and DNA

47
Q

What are TALENs (Transcription activator-like effector nucleases) and how do they work?

A

Molecules produced by Xanthamonas bacteria when infecting plant cells
Recognise and turn on DNA sequences in promoters of some plant genes
Contains many 34aa TALE repeat domains
Residue at positions 12 and 13 define binding to 1bp of DNA
2 required for fusion
Less context dependent (how DNA is packaged)

48
Q

How does CRISPR work in the context of genetic engineering?

A

Cleavage of foreign DNA achieved with CRISPR RNA (crRNA), trans-activating crRNA and Cas9 protein.
Cas9:crRNA-tracrRNA complex target Cas9 to create a ds break specifically in DNA matching part of crRNA
Simplify down by binding CRISPR RNA directly to a version of tracrRNA
Using designed gRNA to direct Cas9 – was enough to create ds break in genome – repaired previously mentioned methods.
Protospacer adjacent motif (PAM) must be next to genetic sequence and upstream of the cut site