Week 8 Flashcards

1
Q

Transfection

A
  • Viral infection initiated by naked nucleic acid
  • Has come to mean transfer of any nucleic acid into eukaryotic cells
  • Uses molecular conjugates e.g. CaPO4 precipitate, Cationic liposomes
  • By viral vectors = transduction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How to initiate viral replication

A
  1. Viral particles (infection)
    • viral RNA (transfection)
  2. cDNA synthesis and cloning + reverse transcription –> DNA –> plasmid (transfection) –> new progeny virion
    • sense strand of RNA, lacking VPG protein (transfection)
  • Can use cDNA cloned into plasmid with suitable RNA pol II
  • RNA synthesised enzymatically using bacteriophage polymerase e.g. T7 RNA pol, SP6 RNA pol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

When using viral vectors, you need to….

A
  • Remove capacity of virus to replicate and release new infectious particles
  • Made defective by deletion of one or more genes required for replication
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Viral vector design requirements

A
  • Appropriate promoter e.g. CMV
  • Terminate RNA e.g. polyA signal
  • Maintenance of genome size within packaging limit of virus - delete non essential genes
  1. Remove viral sequences responsible for pathogenicity and replication
  2. Separate viral sequences required for replication and production of virus particles–> packaging construct
  3. Flank transgene by essential cis-acting sequences and packaging signals
  4. Provide viral proteins required for packaging and replication in packaging cell
  • ​Need RNA packaging signal = retrovirus containing foreign gene
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Comparison of different viral vectors

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

Retro and lentivirus vectors

A
  • Inserts integrated (permanently transduce cells)
  • Lentivirus has a lot more inessential genes - get rid of many of genes
  • VPR - assists in infecting non-dividing cells
  • psi signal = packaging signal
  • Rev retained in lentivirus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Reverse transcription of RNA to DNA

A
  1. tRNA primer extended to form DNA copy of genomic 5’ end, tRNA primer binds to PBS region (primer binding signal), repeated sequence at either end (R), make a copy of R and U5
  2. RNaseh removes hybridised RNA
  3. cDNA hybridisees to R sequences at 3’ end of genome
  4. Extension of DNA strand
  5. Most of hybrid RNA removed
  6. 3’ end of second DNA strand synthesised
  7. Balance of RNA genome and tRNA primer removed
  8. PBS sequences hybridise
  9. both DNA strands completed
  10. LTR repeats - made up of u3, R, U5
  11. Integrates into cellular DNA integrated form called provirus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Issues with retroviral gene transfer vectors

A
  • Regeneration of replication competent retrovirus
    • Inactivate LTR as a promoter, express structural proteins for packaging from separate plasmids
    • Packaging cells must not express endogenous retoviruses
  • Use alternative internal promoter (self-LTR inactivating vector, doesn’t rely on LTR) or remove Tat from HIV
  • Insertional mutagenesis - carcinogenic
  • Limited envelope tropism - add pseudotype vector particles, VSV-G protein (pantropic), can bind to sialic acid –> capable of infecting any cell
  • Heteologous gene expression wanes with time
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Vector plasmid vs integrated vector after RT

A
  • Vector plasmid: remove LTR to serve as a promoter
    • don’t need all of R sequence or all of UR but DO need ends of U5 (att) which interacts with integrase and retain packaging site
    • U3 has other attachment site for integrase, delte TATAA box and enhancer sequences that form promoter
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Applications of retroviral/lentiviral vectors

A
  • Gene correction therapy: no vector-induced immune response or cytotoxicity
  • Cancer therapy: transient high-level expression
  • Vaccines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

DNA virus expression vectors

A
  • Most have very big genomes - too big to clone into plasmid
  • Make shuttle vector e.g. in e.coli
  • Contransfect of recombinant plasmid with viral DNA or transfection of recombinant plasmid DNA into virus infected cells to facilitate homologous recombination
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Adenovirus vectors

A
  • Replication defective form of human adenovirus type 5
  • Retain essential elements (inverted terminal repeats)
  • Non-essential : E3
  • Supplied by packaging construct/cell line : E1A, E1B
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Jesse Gelsinger

A
  • Liver disease: OTC deficiency
  • treated with adenoviral vector high dose (e1 and e4 genes deleted), carried normal copy of OTC gene
  • Death: toxicity of high titer, high immunogenicity of adenoviral vector (immune response)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Safer adenoviral vectors (gutless)

A
  • Remove structural genes by using cre-lox remobination system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Adenovirus expression vectors

A
  • Efficient transduction, highly immunogenic, high titres
  • Gene therapy, vaccines, cancer therapy
  • LImitations: pre-existing immunity, strong T cell response (short term expression), srong humoral response against viral capsid prevents re immunization
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

ZFNs

A
  • Tethered to fok1 - when dimerised, cleaves ccr5 part of DNA
  • Make CCR5 defective –> HIV can’t enter
17
Q

CRISPR- Cas9

A
  • Guide RNA directs annealing, recruits binding of Cas9 proteins which direct editing
18
Q

adeno advantages vs. disadvantages

A

Advantages:

  • High titres
  • Dividing and non-dividng cells
  • Wide tissue tropism
  • Can modify penton spikes to tune tropism

Disadvantages:

  • Transient expression
  • Highly immunogenic
  • High titers of virus can be toxic
  • Suitable for cancer immunotherapy
19
Q

AAV vectors

A
  • Parvovirus
  • Have TR sequences at either end, rep and cap protein
  • Vectors only need to retain TR, can include RNA pol II specific promoter, cDNA, polyA
    *
20
Q

AAV vector: advantage vs disadvantages

A

Advantages:

  • Integration and persistent expression
  • No insertional mutagenesis
  • Infect dividing and non-dividing
  • Safe

Disadvantages:

  • Small size
  • Low virus titer, low gene expression
  • 1 trial recipient death
21
Q

Herpes vectors

A
  • Low risk
  • Big genome - 40kb of foreign DNA
  • Neurotropic
22
Q

DNA recombination to prepare herpesvirus

A
  • Homologous DNA recombination produces mutant strains of Herpesvirus for production of packaging cell lines
  • Vectors produced by recombination with highly deleted defective virus or using large plasmids with an HSV Ori S and packaging sequence
23
Q

RNA replicons

A
  • Self-replicating Rnas
  • replication in cytoplasm
  • High level expression of heterologous genes
  • Can be delivered as: VLP, naked RNA, naked DNA
  • e.g. +ssRNA vectors - togavirus, flavivirus
24
Q

Kunjin vector

A
  • West nile flavivirus
  • High level of expression, noncytopathic, cytoplasmic replication (no integration)
  • No recombination
  • Vaccines for HIV, ebola, cancer
  • Limitations: packaging constrains, pre-existing immunity