Genetic Technologies Flashcards

1
Q

What is PCR?

A

Amplifying a specific known reigon of DNA

primers are used to flank the region you want to amplify

Each cycle doubles the amount of DNA copies of your target sequence

Amplify enough DNA molecules so that we have sufficient sample material

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

What is fragment analysis?

A

A PCR based assay

PCR followed by capillary electrophoresis

Here we are sizing the PCR product to detect repeat expansions or other small size changes

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

What is Huntington’s disease?

A
  • Server neurodegenerative disorder
  • caused by CAG repeat expansion in the Huntington Gene
  • pathogenic when there are MORE THEN 35 copies
  • expanded protein is toxic (accumulates in neurones causing cell death)
  • Diagnosed with FRAGMENT ANALYSIS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is Sanger sequencing?

A
  • cycle sequencing
  • different dye for each four nucleotides
  • reading dyes to obtain DNA sequence
  • single nucleotide polymorphisms (SNPs) or mutations are identified
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is FISH (fluorescent in situ hybridisation)?

A
  • using cells from patients and using a metaphase spread
  • to detect large microscopic abnormalities
  • detect large deleted segments
  • detect translocations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

How is FISH carried out?

A
  1. Design fluorescent probe to chromosomal region of interest
  2. Denature probe and target DNA
  3. Mix probe and target DNA (hybridisation)
  4. Probe binds to target
  5. Target fluoresces
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is Array CGH (comparative genomic hybridisation) used for?

A
  • for detection of sub-microscopic chromosomal abnormalities

Patient DNA - green
Control DNA - red

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

What occurs during Array CGH?

A

2 florescently labelled samples are mixed together

They hybridise to the microarray

Microarray scanner measures fluorescent signals

Computer software analyses the data and generates a plot

Increased green signal over a chromosomal segment in the patient DNA indicates a gain in the patient sample not present in the parents.

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

What is MLPA.

A

Multiplex ligand dependant amplification

Variation of PCR that permits amplification of multiple targets

Used to detect abnormal copy numbers at specific chromosomal locations

Can detect sub microscopic gene duplications/ partial gene deletions

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

How does MLPA function?

A

Two primers are used and detect different oligonucleotide sequences (forward primer and reverse primer)

Only when BOTH OLIGONUCLEOTIDES are hybridised to their respective targets can they be ligated into a complete probe

Amplified target

Then fragment analysis of MLPA to determine relative ploidy (how many chromosome copies?) at specific location

The signal strength of the probes are compared with those obtained form a reference DNA sample known to have two copies of the chromosomes

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

What is whole exome sequencing?

A
  • used for identifying disease causing genes
  • only interested in the protein coding exons (more efficient)
  • cheaper
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

How is whole exome sequencing carried out?

A

By target enrichment

Capture target regions of interest with baits (biotinylated RNA library)

Potential to capture several Mb genomic regions

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

Why wouldn’t all tests automatically move to whole genome sequencing?

A
  • panels/single gene tests may still be more suitable for some diseases
  • capillary based methods: repeat expansions, MLPA, family mutation confirmation Sanger sequencing
  • Array-CGH: large sized chromosomal aberrations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the limitations of Exome and Genome sequencing?

A

Result interpretation is the greatest challange
- large numbers of genetic variants are detected at once

Ethical considerations
- modified patient consent process
- data analysis pathways (analyse relevant genes first)
- strategy for reporting “incidental” findings

Infrastructure and training (time consuming to manually do)

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

What is the 100,000 genome Prodject?

A

Being direct benefit of WGS and genetics to patient

Enable new scientific discovery and medical insights in disease - eg. Personalised medicine

Genomes are collected from genomic medicine genders

In rare diseases - index cases + families

Cancer - germaline (blood) and tumour samples

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

What are the three tiers the panel is divided into?

A

Tier 1:
- known pathogenic
- protein truncating (mechanism known)

Tier 2:
- protein altering
- intronic

Teir 3:
- Loss of function in genes not on the disease gene panel

17
Q

What role does the NHS diagnostic laboratory have?

A

Help consultants reach a genetic diagnosis for individuals and families to help guide treatment and clinical management.

Perform specific tests with:
- clinical validity: how well the test predicts the phenotype
- clinical utility: how the test adds to the management of the patient

UKGTN approved tests

18
Q

What kind of testing does the NHS laboratory do?

A

Diagnostic

Predictive

Carrier(recessive)

Informed consent - genetic counselling (implications for other family members)

Diagnostic testing is available for all consultant referrals

19
Q

What are the the 3 diagnostic test outcomes?

A

Pathogenic mutation

Normal variation - polymorphism

Novel variant - investigate clinical significance

20
Q

How do we establish if a mutation is pathogenic?

A
  • mode of inheritance
  • genetic databases of published and unpublished data
  • nonsense, frameshift, splice site
  • missense/intronic mutation
21
Q

What are the advantages and disadvantages of Sanger sequencing?

A

Advantages:
Up to 800bp per sequence reaction (good for single exons of genes)

Disadvantages:
Slow
Low-throughput
Costly to perform on large numbers of samples