Viruses Flashcards

You may prefer our related Brainscape-certified flashcards:
1
Q

Give examples of the different structures viruses can have:

A
  • Helical capsid, filament like (TMV)
  • Icosahedral capsid (Polio)
  • Icosahedral capsid with lipid envelope (HSV)
  • Helical capsid with lipid envelope, spherical (Influenza)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How can the type of nucleic acid determine the size of a virus?

A
  • Number of sections (monopartite = one, segmented)
  • Error rate: RNA genomes are error prone (particularly RNA dependent RNA polymerases)
  • Some viruses have developed checking capabilities
  • Therefore the largest viruses are dsDNA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Detail ways that viruses increase coding efficiency in their genome:

A

Coding increases efficiency of information to reduce genome size.
- Small intergenetic spacing (few non-coding regions)
- Overlapping reading frames: ambi-sense RNAs where +ve and -ve sense produce different proteins or ORF within larger ORFs
- Post-transcriptional mRNA editing (hijack the host splicing system to create different polyproteins).
- Ribosomal frameshift: nucleotides can be added by polymerase stutter, resulting in higher diversity of protein formation.

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

Name some methods to study virus structure

A
  • Cryo-EM: Shows crystalline structure and can be used to calculate virion concentration (mix sample with known concentration of small beads and count)
  • PCR: when section of sequence is known, specific primers used to identify and quantify genomes (does not measure infectivity).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Suggest some ways to detect a specific virus:

A
  • PCR: using specific primers used to identify and quantify genomes
  • Haemagglutination: for RBC binding viruses
  • Immunological evidence of infection: adaptive immune response will leave antibody/T cell traces to specific virus
  • Cryo-EM to determine structural markers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

How might viral concentration and infectivity be determined (4 methods)?

A

Concentration:
- PCR (number of cycles produced)
- Cryo-EM: concentration estimated by mixing virions with beads to compare count (in virion/mL)
- Haemagglutination: for RBC binding viruses, crude concentration measured by mixing with known RBC concentration. Determine maximum dilution where haemagglutination still occurs.
Infectivity:
- Plaque Assay: A series of dilutions are applied to lawns of susceptible cells. Eventually a visible area of cells are destroyed (plaque forms due to cytopathic effect of virus). Each plaque is from one infective virus. Infectivity = pfu/mL (particle/pfu ratio important)
- Observe effect in host: ID50 number

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

What are the general stages of viral replication (giving specific examples)?

A
  1. Find appropriate host (shows tropism)
  2. Adsorption to host cell: bind to cell surface receptor e.g. HIV gp120 binds to CD4 receptor. Influenza HA binds to sialic acid
  3. Penetration: either endocytosis, macropinocytosis or fusion of virus envelope and cell membrane
  4. Eclipse phase: viral genome inside cell but no viral particles yet.
  5. Assembly of new virions
  6. Release (giving release burst size)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Detail the difference ways that viruses can enter a cell, giving examples:

A
  • Extracellular Uptake: Endocytosis (e.g. macropinocytosis)
  • Fusion (enveloped virus) with cell membrane:
    Influenza: penetrates (endocytosis) allowing acidification of endosome, causing movement of HA1, exposing HA2 and allowing fusion pore formation.
    HIV: gp120 binds to CD4, causing a conformational change to gp120/41 (bringing virus closer to cell)
  • Disruption to host membrane: picornavirus (FMDV, Polio) binds to receptor causing a conformational change to disrupt the membrane
  • Injection of nucleic acid: bacteriophage T4 injects using a syringe like sheath
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Describe the general genome replication cycle of -ve sense ssRNA viruses and give some examples.

A
  1. RNA transcribed to +ve sense by virus encoded RdRp
  2. +ve RNA used as a template strand for new -ve strands, replicates in cytoplasm
  3. Packaged into new virus
    Purified viral RNA is not infectious
    Examples: rabies, measles, ebola
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Describe the general genome replication cycle of +ve sense ssRNA viruses and give examples.

A
  1. RNA impersonates mRNA so translated directly
  2. Translated proteins include RdRp (since they use -ve strand as template)
  3. Either translated into proteins or packaged into new virions

Purified viral RNA is infectious
Examples: poliovirus, FMDV, hepatitis A, rubella

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

Describe the general genome replication cycle of retroviruses (e.g. HIV)

A
  1. +ve ssRNA converted to intermediate DNA using viral encoded reverse transcriptase
  2. This DNA is inserted into host genome = a provirus
  3. mRNA produced using host RNA polymerase (from the provirus)
  4. Full length transcripts are either translated, spliced or packaged into new virus capsids
    Examples: HIV, Visna virus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Describe the general genome replication cycle of dsDNA viruses and giving examples.

A
  1. Viral capsid transported to nucleus (except for Poxviruses!)
  2. Transcribed using host RNA polymerase II
  3. Some proteins (e.g. DNA polymerase and capsid) are taken back to nucleus for virion formation. DNA replicated – through rolling circle replication concatemeric DNA (later cleaved).
  4. Progeny genomes produced and packaged.

Viral DNA alone NOT infectious (needs to get to nucleus)
Examples: HSV, Adenovirus, HPV

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

What is rolling circle replication and which viruses may use it?

A

Method of replicating circular DNA, used by T4 bacteriophages as well as human dsDNA viruses such as HSV and HPV
1. One strand of ds circular DNA is ‘nicked’
2. ‘unnicked’ circle acts as template strand and is elongated, displacing 5’ end of nicked strand
3. Displaced DNA (‘nicked’) is effectively lagging strand and is replicated using Okazaki fragments
4. Displaced DNA is circularised
5. Replicated circles may be cleaved if DNA was concatemeric (repeating copies)

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

Describe poxvirus replication

A

Complex and large DNA viruses
- Have replication ‘factories’ = creates a microenvironment for DNA replication.
- Segregates DNA in cytoplasm (DNA in the cytosol is a powerful PAMP)
- Contain their own vDdRp for capping their RNAs
- They perform cap-snatching from host mRNA. Makes them less likely to be recognised (RLRs do not bind)

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

What are the ways that viruses control their gene expression (name 5)?

A
  • Temporal control: proteins for host cell modification produced earlier than capsid proteins
  • Quantitative control: early proteins (e.g. viral DdRp in a poxvirus) produced in low concentration as reusable enzymes, whereas large amounts of capsid proteins needed (noroviruses produce 180VP1 (capsid), 12VP2 and 1 viral RNA)
  • Polyprotein processing: many proteins from one mRNA (post-translationally cleaved)
  • RNA splicing: coding region placed close to 5’ end (increases chance of ribosome reading it). Splicing efficiency further controls protein concentration
  • Ribosomal Frameshift: ribosomes pause and may slip onto another frame to restart (-1 nucleotide) producing a different protein. Retroviruses exploit this to make polyproteins.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

How might transformation of a host cell lead to a tumour?

A
  • HPV 16 or 18 induce proliferation in host cells before viral replication (increase susceptible hosts): causes host cell to not respond to contact inhibition from neighbouring cells.
  • Retrovirus oncogene capture: virus can obtain a host oncogene during replication and express it at high levels (with no regulation).
  • Disruption of tumour suppressor genes: due to integration of retrovirus DNA in the middle of gene (rare)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What are acute transforming retroviruses and why are they more likely to cause cancer?

A

Oncogene capture and expression can occur:

  • Retroviruses which acquire a host gene during replication, allowing that gene to be expressed at unregulated and high levels
  • When this is an oncogene, a tumour may result e.g. Rous sarcoma virus expressing src (a tyrosine kinase) at high levels – involved in proliferation and differentiation.
  • Oncogene capture often associated with loss of viral sequences so may require co-infection by a helper virus to replicate.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What conditions to viruses need for assembly and how do they assemble themselves?

A

High concentrations of dNTPs (e.g. for large DNA viruses (poxviruses)):
- Have viral thymidine kinases
- Have ribonucleotide reductase (makes dNTP from ribonucleotides)
- Secretes moleculest o increase host production of dNTPs

Virion assembly:
- Spontaneous (non-catalytic) and may involve progressive addition of subunits (e.g. TMV)

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

Discuss some ways that viruses have a cytopathic effect (7 general points):

A
  • Subversion of host cellular metabolism
  • Stimulation of biochemistry to enhance viral yield
  • Increase nucleotide concentrations for viral synthesis
  • Cell membrane modifications/morphological changes (e.g. hypertrophy or viral protein insertion into membrane)
  • Evasion of host sensing of infection (blocking innate immunity)
  • Cell transformation
  • Suppression of host innate immune signalling pathways.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Give examples of how a virus has a cytopathic effect on host genomes.

A

Altering mRNA
- Poliovirus mediated host shut off: cleaves 5’ end off host mRNA so they cannot recruit ribosomes.
- Poliovirus also inserts internal ribosome entry sites (IRES) into its RNA so it can be translated in a cap-independent manner = instead of host.
- Destruction of host mRNA/DNA
- Poxviruses de-cap host mRNA, using it as a signal to switch from early to late gene expression and subverting host proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What are the methods of release used by viruses?

A

Budding: enveloped capsid viruses ‘bud’ from host cell over prolonged periods, stealing cell membrane. E.g. influenza
Cell fusion (cell associated viruses): viruses passes directly from one cell to the other e.g. measles)
Cell lysis: spreads virus. Common for bacteriophages. Proteins causing lysis must be expressed late in infection.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is a latent viral infection? Contrast latent HIV and HSV

A

Time when virus is quiescent – cell contains viral genome but no virus replication occurs. Virus has potential to switch from latency to productive replication cycle.

Retroviruses (HIV): provirus in host chromosome cannot be recognised by host immune system.
- Enables vertical transmission

HSV: viral DNA is quiescent but not integrated = an episome. HSV-1 causes cold sores, VZV causing shingles.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What are HERVs?

A

Human endogenous retroviruses make up 8% of genome – relics of evolutionary infection.
E.g. synctin-2 which is essential for placental formation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

How do viruses beat physical barriers of a host? What are these physical barriers?

A

Replicate without beating barrier = superficial infection:
- Virus replication occurs in epithelium at initial entry site; short incubation, short duration. E.g. influenza, rotavirus

Systemic Infection: breaks through:
- Skin: structural barrier, microbiome (importance shown by sterile mice), sweat (contains lysozyme and dermcidin)
- Epithelial cells: cillia, mucous cells, producing mucus (high in carbs which is hard to navigate)
- Stomach acid (low pH and proteases)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

How might a virus spread in a human?

A

Primary viraemia = spread via lymph, blood or through nerves

Secondary viraemia = infects internal organs for excessive replication (+ high viral blood count)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Describe the ways that a cell recognises viral infection.

A
  • Nucleic acid in unusual place (e.g. DNA in cytoplasm)
  • Nucleic acid with unusual structure e.g. RNA with 5’ triphosphate
  • Sensed by PRRs which activates signalling cascade/TFs: E.g. TLR-4 detects dsRNA in endosome –> nuclear factor NF-kB/interferon response factors activated.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Name ways that the innate immune system works against a viral infection (8 ways)

A
  • Complement: destroys viral particles using MAP/opsonisation
  • Phagocytosis: of viral debris or infected cells
  • Apoptosis: stops viral replication
  • Chemokines: chemoattractant molecules to recruit leukocytes to infection site
  • Cytokines: particularly IL-1/12/18, TNF and IFNs)
  • Interferons: activate virus into an antiviral state and promote adaptive immunity.
  • NK cells: antigen independent killing of infected cells
  • Fever: restricts the replication of microorganisms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Detail how viruses evade the innate immune system (giving examples):

A

Against complement:
- Complement binding proteins (e.g. HSV produces glycoprotein C which binds Cb3).
- Steal host complement suppressor proteins and package into virions (CD46/55/59 produced by HIV and poxviruses)
Phagocytosis: some viruses replicate inside (want recruitment!) E.g. influenza.
Apoptosis blocking: block Bcl-2 functioning and caspase action.
Chemokines:
- Viral blocking e.g. CK protein binds glycosamide glycan (GAG) on endothelial cell
- Viral blocking of CK receptor on leukocyte
- Reduced immune cell recruitment
Against cytokines
- Transformation of host cell to secrete cytokine binding molecules
- Block signalling pathways
- EBV secretes a viral cytokine (vIl-10) to drive response towards Th2 rather than Th1.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Detail the pathway leading to IFNβ release from an infected cell:

A
  1. PRR detects viral PAMP, activating IRF3 and NF-kB.
  2. These move to nucleus to activate IFNβ genes
  3. IFNβ produced which can bind to type I IFN receptor (including as an autocrine molecule)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Detail the types of interferons released following virus detection:

A

Type I (secreted and responded to by all cells): IFNα and IFNβ released by infected cells to upregulate MHC I in neighbours.

Type II (all cells can secrete, but mainly immune cells respond): IFNγ promoting inflammation and Th1 immunity

Type III (all cells secrete but only epithelial cells respond): IFNλ down regulates proliferation.

31
Q

Detail how IFNβ is detected and the effect it has on the target cell.

A
  1. Binds to type I IFn receptor (can be autocrine molecule)
  2. Induces JAK-STAT pathway, leading to transcription of interferon stimulated gene factor 3 (ISGF-3)
  3. ISGF-3 binds to interferon stimulated response element (ISRE) on specific interferon stimulated genes (ISGs)
  4. E.g. protein kinase R is an ISG which inhibits protein synthesis in a cell (following dsRNA detection)
32
Q

How do viruses evade IFN action? Give examples.

A

Target every step of the pathway:

  • Stop PRR detection: cleavage of PRRs, release of soluble blocking proteins (e.g. vaccinia virus produces B18 which binds IFNα to prevent binding)
  • Target JAK-STAT signalling pathway (e.g. Zika virus degrades Stat2)
  • Target ISG protein action
33
Q

How do viruses evade the adaptive immune system?

A

Reduce PAMP presentation:
- Block generation of peptides by proteasome
- Block transport of those peptides to ER
- Block MHC signalling (increases NK recognition though): tethers MHCs to ER or destroys them (HCMV degrades MHC I)

Hide: latency

Prevent antibody binding:
- Antigenic variation (old antibodies useless)
- Express viral Fc receptors so antibody cannot bind to cell receptors (prevents ADCC from complement)

34
Q

How does the adaptive immune system fight viral infections?

A

Antibodies:
- Mucosal IgA particularly important for respiratory infections
- Some antibodies can go inside cells for intracellular help (e.g. Trim8)
Cytotoxic T-lymphocytes to kill infected cells:
- Very important for cell associated viruses (measles and HCMV)

35
Q

What factors may affect the outcome of infection? (6 points)

A
  • Viral dose
  • Route of entry (e.g. dermal vs. respiratory smallpox has large mortality variation)
    Previous infection:
  • Antibody dependent enhancement (ADE) occurs with second Dengue infection when low affinity initial antibodies bind receptors and virus = facilitates entry!
  • Previous infection with measles may cause immune amnesia as measles clears CD150+ memory T cells and some follicular B cells (Tfh have SLAM receptors which MV can bind to)
  • May help if secondary adaptive response ready (point of vaccination)
  • Age (e.g. VZV more severe as age increases)
  • Sex
    Physiological state of host:
  • Stress level (endorphins)
  • Genetic predisposition: E.g. those with HLA haplotypes of B27 or B57 are long-term slow progressors for HIV (show natural immunity)
36
Q

What factors affect the spread of a virus (both within and between hosts)?

A

Spread requires amplification and a mode of transmission.
- Tropism of virus: how many cells express correct receptor, pro-viral factors or expression of anti-viral factors
- Host response: some viruses require response to spread (e.g. HIV in CD4 cells)
- Temperature: most viruses show narrow optimum temp range (why endogenous pyrogens can be helpful). May effect tropism (e.g. rhinovirus optimum 32C so replicates in upper respiratory tract epithelium).
- Budding site; apical or basal? Apical allows easier host-host transmission (e.g. influenza into respiratory lumen) but basal gives higher chance of systemic infection

37
Q

What are the outcomes of latent viral infections?

A
  • Resolution
  • Reactivation: on stimulus such as cell differentiation (e.g. HCMV production on monocyte to macrophage change. Can reactivate with different morphology (VZV reactivates to cause shingles – dermatome spread since latent in sensory nerves)
  • Recurrence = disease after reactivation. Can persist as chronic (Hep B, Hep C, HIV) or latent (HerpesVs e.g. VZV, HSV, HCMV)
38
Q

Give factors which affect the transmission rate of a virus (5 points):

A
  • Particle stability in environment
  • Duration of shedding (from host)
  • Concentration of virus
  • Availability of susceptible hosts
  • Vertical transmission
39
Q

What factors affect the stability of a viral particle in the environment?

A
  • Enveloped viruses generally less stable (cannot survive in dry environments)
  • Temperature
  • Route of infection e.g. faecal-oral transmitted viruses are very stable (evolved to survive stomach acid, bile…)
40
Q

What are the type of vertical virus transmission?

A

Congenital infection: through placenta (HIV, HCMV, rubella)
Perinatal infection: during birth or through breast milk (HIV, HCMV)
Germ line transmission: presence of retrovirus as provirus to offspring (e.g. endogenous retroviruses - normally silent)

41
Q

What factors affect epidemiological transmission?

A
  • Number of susceptible hosts and host species (for zoonotic viruses)
  • Immunity in hosts
  • Mutation/antibody change rate
  • R values (R0 = innate infection rate; Rt = infection rate given measures)
42
Q

Give an example of the economic impact of viral disease:

A
  • 2001 FMDV cattle and sheep outbreak led to culling of infected animals and >£8bn damage
43
Q

What are the main structural components of influenza virus?

A

Genome: -ve ssRNA (8 segments) associated with nucleoproteins (similar to histones). Contains RdRp in capsid (made from PB1, PB2 and PA proteins)

Capsid: enveloped capsid

Surface proteins: haemagglutinin (HA) and neuraminidase glycoproteins (NA) with M2 ion channel exposed and M1 under envelope.

44
Q

Describe the life cycle of influenza:

A
  1. HA binds to sialic acid on target cell, facilitating endocytosis.
  2. Acidification of endosome causes conformational change to HA, allowing fusion of virus to endosomal membrane and uncoating of virus.
  3. Nucleocapsid moves to the nucleus
  4. Viral RdRp transcribes all RNAs to +ve sense (some (7&8) being polyproteins)
  5. Cap-snatching from host mRNAs adds 5’ cap and polyadenylate tails to viral RNAs making them more host-like
  6. New nucleocapsids move to cell periphery and bud through membrane (acquiring envelope from host cell)
  7. Release requires NA activity to cleave sialic acid to prevent aggregation of viruses.
45
Q

How does acidification of an influenza filled endosome facilitate infection?

A

Fusion:
- Haemagglutinin on influenza contains HA1 and HA2 subunits
- HA1 binds sialic acid and when acidified undergoes a conformational change, exposing HA2
- HA2 (fusion peptide) allows fusion between virus and endosomal membrane
Uncoating:
- Acidification also causes import of protons through M2 channel
- Promotos uncoating of virus

46
Q

Suggest some important drug targets for influenza:

A
  • Preventing entry by disrupting HA antigen
  • Preventing acidification of endosome (salinomycin blocks M2)
  • Preventing leaving by disrupting NA cleaving of sialic acid
47
Q

Why does Influenza need to replicate in the nucleus

A
  • Cap-snatching requires host DNA dependent Rna pol II (adds to host-cell shut-off)
  • Nucleus also protected from the the rest of the cell (and immune recognition)
48
Q

What are the two main ways antigenic variation has occurred in influenza?

A

Antigenic drift: mutations resulting in changes to amino acid sequence (gradual):
- E.g. changes to HA antigen (must still bind to sialic acid) allows affinity for different hosts (18 HA types, 11NA types)
- E.g. single change in PB2 subunit of RdRp from E627 to K627 allows better replication in humans as opposed to birds.

Antigenic shift: radical change due to acquisition of a new HA from another influenza virus.
- Can occur during co-infection (RNA recombination)
- Responsible for Spanish flu (H1N1)

Antigenic variation more likely in RNA viruses due to higher RdRp error rate and smaller number of target proteins

49
Q

Contrast the structures of hepatitis A, B and C

A

Hepatitis A: a picornavirus with +ve ssRNA.
- Half-naked; half enveloped.
- HBsAg lipoprotein complex surface antigen.

Hepatitis B: small (42nm) DNA reversivirus (uses reverse transcriptase to replicate).
- Enveloped.
- Virions present with large amount of free surface antigen

Hepatitis C: small (50nm) flavivirus with +ve ssRNA. Enveloped.
- E1 and E2 glycoprotein surface antigens

50
Q

Describe what the hepatitis B genome codes for:

A

Very complex genome (4 overlapping reading frames). Partially ds, partially ss.
- Reversivirus so codes for a reverse transcriptase to make DNA copies from template RNA
- Encodes capsid protein
- Transactivating protein
- Surface glycoproteins (produced in huge amounts, released as free clumps as well as virions)

51
Q

Compare/contrast disease caused by hepatitis A, B and C:

A
  • All affect the liver (hence hepatitis) so cause jaundice, fatigue…

Transmission: Hepatitis B/C transmitted through blood while A is faecal-aural.

Infection site:
- Hep A infects epithelium of intestine = diarrhoea, fever.
- Hep B is blood borne causing acute hepatocellular carcinoma (HCC)

52
Q

Contrast prevention strategies for Hepatitis A, B and C

A

Hep A: attenuated vaccine, good hygiene (since faecal-aural transmission)

Hep B: vaccine (effective but expensive and dangerous to produce)

Hep C: No vaccine, only treatment (expensive but effective).

53
Q

Describe the genomic structure of Sars-Cov-2 (adaptations to allow for a large genome):

A
  • 80-140nm (large) allowing for 29.9kb RNA
  • +ve ssRNA
  • Includes proofreading capabilities (allows for larger genome)
  • Codes for non-structural protein nsp14 (an exonuclease) and nsp10.
  • Codes for RdRp (with ribosomal shift potential)
54
Q

How does Sars-Cov-2 infect cells?

A
  1. Spike protein of virus binds to ACE-2 receptor in lungs facilitating endocytosis.
  2. Endosome transported to Golgi where furin (host enzyme) cleaves S1 from S2.
    - Spike protein made from S1 (includes receptor binding domain) and S2 which allows fusion.
  3. Allows release of viral material.
55
Q

Briefly describe prevention and treatment methods for Sars-Cov-2

A

Prevention: vaccination
- mRNA (first created!)
- Viral vector
- ‘Killed’ virus based

Treatment: worst responses were overactivation of immune system in lungs, hence immunosuppressive drugs used.
- Dexamethasone: anti-inflammatory
- Remdesivir: inhibitor of viral RdRp (effective but toxic to liver)
- mAbs against spike protein.

56
Q

Describe the structure of HIV:

A

Capsid:
- Cone shaped, enveloped virus
- p17 capsid with internal gag p24 inner capsid
- Surface gp120 and gp41 proteins
- Gp is heavily glycosylated (reduces affinity to antibodies by creating a glycan shield)

Genome:
- Retrovirus (+ss RNA)
- Includes 5’ cap and poly A tail (disguises as mRNA)
- Diploid genome with tRNA in virion used to prime reverse transcriptase (converts to DNA).

57
Q

Describe the replication cycle of HIV:

A
  1. gp120 binds CD4 on CD4+ cells (including DC and macrophages)
    - Co-receptor must also be present (CCR5 preferentially, or CXCR4)
  2. Conformational change in gp120 allows fusion of envelope to host membrane
  3. Virus moves to nucleus where tRNA in virion primes reverse transcriptase, converting RNA to DNA
  4. Integrates into host genome as provirus using viral integrase
    – Can lie latent here for years –
  5. Viral RNA replicated by host machinery (RNA pol II) to mRNA (including 5’ cap and poly A tail)
  6. mRNAs heavily spliced to form viral proteins (polyproteins)
    - Virus also reduces host mRNA production and blocks NF-kB pathway.
  7. Assemble into virions and release
58
Q

Explain the difference between a retrovirus and a reversivirus.

A

Both use reverse transcriptase but for different reasons

Retrovirus (e.g. HIV): turns +ve ssRNA into DNA to integrate into host genome

Reversivirus (e.g. Hep B): is a DNA virus that uses reverse transcription to form a template strand for new DNA molecules (does not integrate)

59
Q

What are the methods of HIV treatment?

A
  • Vaccine hard to produce as fast antigenic variation.
    Better to treat with post exposure drugs:
  • Inhibit viral replication (stop fusion, integration or use protease inhibitors) e.g. enfuvirtide
  • Viral load can be kept low enough to prevent transmission
  • Multiple drugs given simultaneously to prevent resistance = highly active anti-retroviral therapy (HAART)
60
Q

What are some non-vaccine methods of epidemiological control?

A
  • Quarantine/isolation/slaughter: useful for animal diseases (e.g. rinderpest and rabies)
  • Surveillance: early detection of an outbreak for notifiable viruses (e.g. measles, influenza, AIDS)
  • Sanitary Engineering/hygiene regulations: very important for faecal-oral transmission and blood borne infections (reused needles)
  • Vector control: e.g. mosquitos
  • Screening of blood products: prevents spread via blood transfusions
61
Q

What was the first developed vaccination?

A
  • Variola major virus (smallpox) with 30-40% death rate
  • Variola minor virus (cowpox) with 1% death rate
  • Therefore minor used to vaccinate against major.
62
Q

Which criteria makes eradication possible?

A
  • Virus contained to one/isolated species (no animal vector for human disease)
  • Acute infection (e.g. short latency with no persistent infection)
  • Easily recognisable disease (notified early)
  • Vaccine works against all strains (slow mutating virus; little antigenic variation)
  • Vaccine properties: potent as a single dose, low cost, abundant (e.g. smallpox vaccine could be freeze-dried for easier transportation)
  • Vaccine induces long-lasting immunity
63
Q

What are the types of vaccination? Give an advantage and disadvantage for each:

A

Live vaccination: attenuated mutants with same antigen/related less virulent strain.
- A = self-replicating so cheaper to produce. Long-lived immunity.
- D = virus might revert to virulent form (dangerous). Virus may be too dangerous for immunocompromised patients.
Dead: whole inactivated virus/subunit vaccine (e.g. mRNA)
- A = no infectivity risk
- D = may require multiple doses. Often requires cold storage.
Passive: giving pre-formed antibodies
- A = immediate protection (post exposure prophylaxis)
- D = serum sickness may result (type III hypersensitivity). Short-lived protection.

64
Q

Suggest some important factors to consider during vaccine development:

A
  • Which antigens/viral molecules are important to expose
  • When is the virus a problem? E.g. Rubella in pregnancy.
  • What type of immunity is needed? What type of antibody is best (e.g. IgA on mucosal membranes or systemic and free IgG)
65
Q

What is acyclovir and how does it work?

A

Treatment for HSV
Works by stopping genome replication
- Becomes activated in infected cells
- Activated on phosphorylation by HSV-tyrosine kinase (not host TKs) forming a tri-phosphate end.
- This NTP is incorporated into viral DNA by HSV DNA pol and terminates the sequence.
- Stops viable virus genome.

66
Q

Give examples of drugs which stop the entry or exit of a virus:

A
  • Maraviroc binds to CCR5 co-receptor on susceptible cells, stopping HIV binding.
  • Zanamivir binds to NA antigen on influenza, preventing sialic acid cleavage and therefore budding of virus.
67
Q

Give examples of drugs which interfere with genome replication of a virus:

A
  • Abacavir: inhibits HIV reverse transcriptase (incredible success story!)
  • Acyclovir: becomes phosphorylated only in infected cells and competitively inhibits dGTP - is a dideoxy version so ends polymerisation of viral DNA.
  • Baloxavir: inhibits cap-dependent endonuclease of influenza replication.
68
Q

What are some requirements for successful antiviral drugs?

A
  • Targets virus but not host (hard as many viruses hijack host machinery)
  • Have high bio-availability (absorbed quickly)
  • Relatively inexpensive
69
Q

Give examples of viruses which establish a latent infection:

A

Genome in nucleus as episome:
- HSV type-1: replicates in epithelial cells ➡ sensory neurons (hides in cell body) ➡ episome in nucleus of trigeminal nerve
- Uses latency associated transcript (LAT) transcribed (using host RNA pol II)
- Transported by retrograde transport to nucleus
- Varicella-Zoster Virus (VZV): primary infection systemic ➡ secondary infection is nervous in dermatome region (shingles)
(Whereas for HSV primary and secondary infection are the same)

Latency through genome integration (copy on division)
- Human herpes virus 6 (HHV6) integration near telomeres
- Retroviruses (HIV): replicate via reverse transcription from ssRNA ➡ RNA:DNA ds hybrid ➡ degraded by ribonuclease H ➡ dsDNA which is integrated into host chromosome (provirus)
- Endogenous retroviruses are evolutionary examples

70
Q

What are the strategies by which viruses hide from the immune system (4)?

A
  • Latency without integration (VZV, HSV-1)
  • Genome integration (retroviruses, HHV6)
  • Replication in privileged sites (ebola)
  • Hiding during spread (formation of cell syncytia by measles and HSV): reduces membrane:cytoplasm ratio and therefore T external exposure
71
Q

Describe the mechanisms by which viruses can suppress MHC I presentation:

A

Kaposi sarcoma herpes virus:
- K3 protein (expressed in cell membrane) adds ubiquitin on MHCI causing endocytosis (clathrin mediated)

Cowpox:
- CPX203 protein traps MHCI complexes in Golgi (reducing expression)
- CPX012 binds TAP: stops maturation, loading and reduces expression

72
Q

In what ways do viruses evade the innate immune system using effector proteins? (4 main)

A

Steric hindrance:
- VacV A46R blocks TLR signalling (binds IRAK)
- VSV A49 protein inhibits NF-kB signalling by binding β-TrCP and sequestering it

Degradation:
- Viral E3 ligases regulate post-transcriptional modifications of RLRs (e.g. stop ubi build up)
- HIV vpu induces ubiquitination and then degradation of CD4 in proteosomes

Decoy (mimic host proteins):
- Viral ‘stealing’ of Bcl-2 protein to perform different function (inhibit NF-κB) as surface has been changed

Cleavage:
- Enterovirus 3C protease cuts NLRP3 inflammasome receptor (reduces IL-1β secretion)
- Suicide substrates
- VaCV CrmA inhibits caspase action — stops apoptosis

73
Q

What are the main HPV viral proteins which assist in re-epithelialisation and immune evasion?

A

All have dual functions!

Re-epithelialisation:
- E5 = activates EGFR without ligand
- E6 = represses p53 and inhibits differentiation by NOTCH signalling
- E7 = drives cell cycle entry (binds pRB)

Immune evasion:
- E5: reduces MHC levels
- E6: disrupts IFN signalling ad reduces Langerhans cells (specialised DC cells)
- E7: Inhibits STAT pathway and IFn signalling and represses MHC presentation