Unit 2 Exam Recall Q's Flashcards

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

What do you know about viruses? Start generally and go into more applied observations.

Do not define.

A

Generally:
- eat and breathe billions of them regularly
- infect our pets, domestic food animals, wildlife, plants, insects (crossing species barriers constantly - zoonotic infections)
- most abundant entities in global ecosystems
- different viruses face radically diff. challenges in host & extracellular environments

Applied:
- can co-evolve with hosts
- we carry viral genomes as part of our own genetic material
- exchange genetic info between virus & host
- viruses capture human genes (can encode things needed in our genes to use to adv. or not at all)
- too big to diffuse across PM (must create mechanisms to bypass these layers)

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

Do marine viruses affect the global carbon cycle? If yes, how/why? Explain generally & with ex of whales.

A
  • Microbes represent large majority
    of biomass in the ocean
  • Every milliliter of seawater has at least 10 million virus particles

Why/how?
- pathogens steal energy from ocean bacteria, preventing them from sucking up the greenhouse gas carbon dioxide
- impact material cycles and energy flows in the food web

Whales
- massive shedding events
- infected by virus from calicivirus family

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

How infected are we really with viruses? Define VZV, HCMW and EBV as examples.

A
  • majority of viruses are innocuous (not harmful)
  • persistent, life long infections (for ex: herpes virus(es) don’t jump species, attended to our biology, reactivation possible)

Defining
- VZV: human herpesvirus 3, chicken pox & shingles
- HCMV: Cytomegalovirus, leads to prenatal effects
- EBV: Human gammaherpesvirus 4, Mono (triggered by multiple Sacroiliitis)

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

What is the evidence that viruses have been with us since ‘the beginning’? What percent of our genome has retro-virus sequences?

A
  • presence is evident in genomes of
    vertebrates
  • retroviruses integrate into host genomes, meaning they have shaped evolution & gene expression
  • 10% of the human genome consists of retrovirus-like sequences
  • Similar findings in other genomes (mouse, rat, etc.)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

How are these viral sequences ‘fixed’ in the human genome?

A
  • fixed as they infect cells that are apart of humans germ line cells
  • integrated and passed to progeny
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Explain how retroviruses are in the process of being ‘fixed’ in the Koala host genome? What is the virus called?

A
  • Koala retrovirus (KoRV)
  • Infectious cause of Koala Immune Deficiency Syndrome (KIDS) which is like AIDS
  • KoRV is being ‘endogenized’ or integrated in Koala genome
  • spreads ‘horizontally’ and ‘vertically
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Do integrated ancient viruses have an effect on us? Syncytin?

A

Syncytin
- viral gene (endogenous retrovirus)
- Ancient cell attachment protein
- Located in human chromosome 7

Overall:
- retroviral envelope protein involved in human placental development
- protects the fetus from the mother’s immune system until delivery
- creates fused synctiotrophoblast layer

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

Explain the process of Syncytin creating a synctiotrophoblast layer. Why important?

A

Process:
- takes cells and makes them fuse together
- fuses into 1 cell, many nuclei created

Why?
- needed to maintain semipermeable barrier between mother and fetus
during pregnancy

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

Define what a virus is and what it lacks. Explain what you know about virus particles?

A

Definition:
- toxic/poision
- submicroscopic (smaller than diffraction limit on most microscopes) , obligate intracellular parasites
- smaller than bacteria

Lacks:
- genetic info to generate metabolic energy and synthesize proteins (very dependant on overtaking host machinery, use this to learn about host/virus)

Virus Particles:
- physical structures
- molecular structures that package virus genomes in infected cells and transmit them to new host cells (virus makes components in host cells, assemble into package and release from cell)
- do not divide or grow

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

Why do viruses overtake cellular process? Would we consider them living/alive?

A
  • certainly viruses multiply, but they do not ‘grow’
  • viruses harness cellular processes to direct genome replication and assembly of progeny virions

Alive?
- inside the host cell: viruses are alive
- outside of the host cell: complex assemblages of metabolically inert
chemicals

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

How do we visualize viruses? Give two examples and explain.

A
  1. Plaque assays (indirect)
    - serial dilute inoculum
    - add to layer of susceptible cells
    - overlay with agar
    - wait for cytopathic effect (cell destruction)
    - plaques are holes in the single layer of cells.

Notes:
- virus destroys host cell on replication, so infects cells w/ viruses from sample
- overlay cells are in immobilizing agar, infectious cells grow laterally
- cytopathic effect allows visualization of cell death (intact = like intact cobblestone, infected = scattered)
- plaques show initial infection event via holes present

  1. Electron Microscopy (direct)
    - powerful method to visualize viruses
    - electrons pass through a stained, ultrathin sample
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What are physical properties of viruses?

A
  • capsids (protein shells)
  • genomes (dsDNA, ssDNA, dsRNA, +ssRNA, -ssRNA)
  • optional components: tegument and envelopes

Key:
ds = double stranded
ss = single stranded
+ = ready to be translated
- = must decode into + to translate

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

Explain what you know about virions. Give steps in order of reaching host cell.

A

Virion:
- a complete, infectious, virus particle

Steps:
1. Assembled correctly
2. Escape the cell (where made)
3. Survive extracellular environment
4. Attach to and enter another host cell
5. Uncoat and release the viral genome

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

Explain all you know about virus capsids. What do they protect from? What’s a capsomer?

A

Capsids:
- a rigid, symmetrical container for viral genomes

Protect fragile nucleic acid genome from:
1. Physical damage - shearing by mechanical forces
2. Chemical damage - UV irradiation (from sunlight) leading to chemical
modification
3. Enzymatic damage - nucleases derived from dead/leaky cells or
deliberately secreted by vertebrates as defence against infection

Capsomers:
- subunit of the capsid, an outer covering of protein that protects the genetic material of a virus.
- self-assemble to form the capsid.

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

Why do virus capsids need to be metastable?

A
  • strong enough to withstand environment and protect genome
  • able to be ’unlocked’ and disassemble during virus entry (uncoating)

capsid (box) –> cell cytoplasm –> nucleus (turn on virus genes)

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

What are the four virus morphologies? Explain each.

A
  1. Helical
    - simplest capsid
    - composed of single type of capsomer stacked around a central axis
    - forms a helical structure
    - hollow center of helix contains the viral genome (usually ssRNA)
  2. Icosahedral
    - animal viruses have icosahedral symmetry (e.g. poliovirus, adenovirus, and hepatitis A virus)
    - Icosahedron - a polyhedron with 20 faces, 30 edges and 12 vertices
    - 5 fold symmetry (differing proteins create variability of structure)
    - Many viruses with ‘naked’ icosahedral capsids are enteric viruses (affecting gastro. tract)
    - get rid of w/ bleach
  3. Enveloped
    - cover themselves in outer layer of host cell lipid membrane (plasma membrane, or internal membranes ie: NM, ER)
    - have membrane-spanning viral glycoproteins (important roles in infection)
    - Matrix proteins found just beneath the lipid envelope and provide link between the envelope and the nucleocapsid
    - a lot of examples (Rabies, SARS-CoV-2, Ebola, HIV, influenza, herpes)
    - sensitive to cleaning agents like detergents
  4. Complex
    - ex: Bacteriophage T4 (explain later)
    - have a capsid which is neither purely helical, nor purely icosahedral,
    - may have extra structures such as protein tails or a complex outer wall.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How do these capsomers bind to the viral genome? Is it likely to be sequence specific?

A

They bind to viral genome due to electrostatic interactions
- RNA (-)
- Amino acids (+)
- electrostatic int. aid in assembly as they guide RNA into protein subunits, each subunit needs amino acid inside

No, not sequence specific

<—- right to left assembly

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

What governs the length and diameter of the helix?

A
  • length of helix is determined by the length of the genome
  • diameter determined by turns in helix or width of the coil
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Explain Fraenkel-Conrat & Williams experiment of TMV and what they found out about (helical) capsids/viral particles?

A

Experiment:
- when mixtures of purified tobacco mosaic virus (TMV) RNA and coat protein were incubated together, virus particles formed.

Discovery/conclusion:
- virus particles could form spontaneously from purified
subunits w/o any extraneous information
- particle in the free energy minimum state
- some viruses are fragile and unable to survive outside the protected host cell environment, but many persist for long periods, in some cases for years (capsids aid in stability)

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

Why do so many viruses adopt the icosahedral capsid shape? Give four reasons.

A
  1. Strength of the icosahedral structure
  2. Resistance to shear forces (& fluid)
  3. Tight packaging of the genome – maximal volume:surface area ratio
  4. Genetic Economy – can be built from a few repeating subunits (can do a lot w/ small genome)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

How is the cosahedral structure advantageous for these enteric viruses? How should we decontaminate surfaces covered with these viruses? Give two examples.

A

Advantageous
- strong and resistance enzymes
- low hostile pH environment

Decontamination:
1. bleach
- oxidizes proteins
- lose ability to attach to host cell or replicate genome
- takes time

  1. detergent (possibly)
    - denatures proteins to an extent
    - disrupts lipid envelope
    - honestly better for enveloped viruses (don’t like -OH, more susceptible to agents)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What do you know about glycoproteins? Why sugar proteins important for protein function?

What are the two types of glycosylation?

A

Glycoprotiens
- Viral glycoproteins are modified with host sugars added in the ER and Golgi apparatus
- normal host process, many host integral membrane proteins are glycosylated (virus reworked normal system for benefit)

Types of glycosylation
1. N-glycosylation: addition of sugar chains on amide nitrogen of asparagine
2. O-glycosylation: addition of sugar chains on serine/threonine

These sugar decorations are often CRITICAL for protein function:
- won’t work if not properly glycosylated
- can’t hide from immune response
- fight to keep away antigens

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

Compare structural and non- structural proteins.

A

Structural Protein
- found in the virion
- even if it does not play a structural role its called structural protein
- ie: polymerase in ebola virus, only plays role once INSIDE host

Non-structural Protein
- made in the infected cell but not
found in the virion.
- can still be essential to the life cycle of the virus even if it is not part of the virion.
- ie: can help in making progeny but not passed on

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

Go into detail about the involved virus; Herpes Simplex Virus.

A
  • Herpesviruses have envelopes AND tegument proteins
  • Tegument proteins are delivered into cytoplasm upon virus entry (allows them to act early in infection, to prevent antiviral responses)

Tegument
- space for enzymes to be delivered and do work (virus may dump these enzymes out to antagonize host immune system)

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

Briefly explain structure of bacteriophage T4

Hint: Complex virus morphology

A
  • Icosahedral head: containing viral dsDNA genome
  • Helical tail
  • Hexagonal base plate
  • Protruding protein tail fibres
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Give an overview of the form and functions of different virus parts. There should be seven named.

A
  1. Spike protein (receptor binding)
  2. Envelope (entry and exit)
  3. Matrix protein (assembly & stability)
  4. Capsid (stability)
  5. Nucleocapsid (genome packaging)
  6. Genome - RNA or DNA
  7. Polymerase (genome replication)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What are the steps viruses follow in a single replication cycle? Acronym?

A

APUTTGAR

Attachment
Penetration
Uncoating

Transcription
Translation

Genome Replication
Assembly
Release

Note:
- first three apart of entry
- last three apart of maturation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What are the two biggest concepts relating to viral entry?

A
  • Where the virus infects with in the body, where the virus can enter, is
    dictated by the location in the body of the virus receptor
  • Where the virus is infecting within the body dictates the disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

How does bacteriophage T4 attach to E. coli?

A
  1. initial reversible binding via long tail fibres
    - browses cell surface to find right spot
  2. irreversible binding via short tail fibres
    - bottom of tail
    - interacts w/ cell receptors
    - attempt to infect cell

Note:
- Attachment, penetration and uncoating at occur at bacterial cell surface

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Explain the steps in bacteriophage entry, more specially, how Phage T4 binds to specific receptors on E. coli cell surface.

A
  1. Long tail fibres recognize outer membrane protein C (OmpC) or lipo-
    polysaccharide (LPS) of E. coli (REVERSIBLE)
  2. After at least 3 long tail fibers have bound, there is a conformational change in the baseplate, and short tail fibers extend and bind irreversibly to the core region of the host cell LPS. (IRREVERSIBLE)
  3. Contraction of tail sheath and penetration of outer membrane
  4. T4 lysozyme (virus structural enzyme) degrades (pokes holes in) protective peptidoglycan layer
  5. Inner membrane is degraded
  6. Phage DNA is delivered into cytoplasm
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Briefly describe viral infection of EUKARYOTIC cells. Attachments? Penetration types? Locations?

A
  1. Initial attachment
    - reversible electrostatic interactions
  2. Stable attachment
    - irreversible tight interactions with receptor(s)
  3. Viruses penetrate host cells by
    - Membrane fusion at the cell surface
    - or receptor-mediated endocytosis

Where beginning stages occur:
Attachment - cell surface
Penetration - cell surface or internal membrane(s)
Uncoating - cytoplasm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is Virus–Receptor Promiscuity and specificity? (VAP)?

A
  • VAP = Virus attachment protein
  • Different viruses can bind the same receptor (ie: Adenovirus and Coxsackievirus bind to same
    cellular receptor)
  • Related viruses in the same family can bind different receptors
  • Virus entry sometimes requires more than one receptor (there is an order to receptor engagement; receptors first, co-receptors second)
  • Receptor-coreceptor system

Ultimately just:
- Complex interactions with the different cell surface receptors
- Some require a combo of receptors to confer entry

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Describe Influenza Virus’s cell receptor; Sialic Acid.

Note:
- viral entry into human cells

A
  • a generic term for the N- or O substituted derivatives of neuraminic acid, a monosaccharide with a nine-carbon backbone
  • Sialic acids are found widely distributed in animal tissues mostly in glycoproteins
  • occur @ end of sugar chains connected to the surfaces of cells and soluble proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Sialic acid is ubiquitous on cell surfaces. What does this mean for influenza virus tropism?

A
  • able to infect many cell types.
  • diff. strains bind more strongly to diff. types of sialic acid chains
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Describe the steps the influenza virus does to achieve viral entry by endocytosis. There are six.

A
  1. Initial attachment of HA envelope glycoprotein to cell surface sialic acid
  2. Receptor-mediated endocytosis
  3. pH drops in endosomes (pH 7 to pH 5)
  4. Low pH induces conformational change in HA, exposing hydrophobic fusion peptide.
  5. Fusion..viral and cell membranes mix.
  6. Influenza virus genome delivered into host cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What does the Influenza viruses have in the viral envelope? What does this aid in? What protein mediates?

A
  • it has a proton channel in the viral envelope
  • allows acidification of the core which is essential for uncoating
  • occurs after membrane fusion & is mediated by the viral M2 protein.
37
Q

How can uncoating be prevented in Influenza viruses? Is this used in clinical trials?

A
  • uncoating can be prevented by amantadine (blocks the M2 proton channel)
  • single amino acid change in M2 causes amantadine resistance
    without affecting proton channel function.
  • M2 inhibitors are not used in the clinic due to rapid emergence of drug
    resistant viruses.
38
Q

When is entry is complete for Influenza viruses? What do segments do? How many of them?

A
  • Entry is complete when the viral genome is delivered to the cellular compartment where replication takes place (cell nucleus - influenza viruses)
  • After uncoating, each of 8 genome segments traverses nuclear pore.
  • Each segment is attached to viral polymerase enzyme.
  • The polymerase starts to copy RNA inside the nucleus
39
Q

What do you know about the human coronavirus structure?

A
  • Attachment and fusion is mediated by the Spike glycoprotein
  • so coronavirus VAP = spike protein
  • There are 7 human coronaviruses that have different cellular receptors (entry pathways are similar, despite utilizing different cellular receptors)
  • Spike must be cut by a host cell protease to be ‘activated’
40
Q

Host protease inhibitor drugs can block the fusion step of entry in coronaviruses, what are two examples of drugs not yet at the front-line?

A
  1. Paxlovid inhibits a viral protease
  2. Remdesivir inhibits a viral RNA- dependent RNA polymerase
41
Q

Multiple proteases in different cellular
compartments can cleave Spike. What are the three used in coronaviruses? Where are their cleavage sites?

A
  1. Furin
    - first opportunity for cleavage event
    - proteolytic enzyme
    - S1/S2 site
  2. Transmembrane protease serine 2 (TMPRSS2)
    - second opportunity
    - on target cell
    - exposes fusion peptide
    - S2’ site
  3. Cathepsin
    - if spike is still not cleaved, this is 3rd option
    - abundant lysosomal proteases
    - both sites
42
Q

For coronaviruses, when entry is complete, which cellular compartment does it go to? Why?

A
  • entry is complete when viral genome is delivered to the cytoplasm.
  • RNA genome can be directly translated by host protein synthesis machinery to initiate infection.
  • ie: This machinery is in the cytoplasm.
43
Q

Describe an overview of bacteriophage Replication.

A
  1. after penetration, viral DNA is delivered into host E. coli
  2. Host gene expression is arrested (immediately)
    —> ie: host DNA/RNA degradation and inhibition of protein synthesis
  3. Enzyme synthesis (starting after 5 minutes)
  4. DNA replication (starting after 10 minutes)
  5. Formation of new virus particles (starting after 12 min)
  6. Lysis of host and release of viral particles (at 30 min)
44
Q

What are the two types of antiviral defense? Explain each briefly.

A
  1. Restriction Endonucleases
    - they are enzymes, ‘molecular scissors’, that can cut dsDNA nucleotide sequences (restriction sites) in a
    sequence-specific manner.
    - cut up foreign DNA (staggered)
    - useful in cut-and-paste gene cloning

Overview:
- host DNA is methylated by a host cell enzyme, which prevents cutting by host
restriction endonuclease
- to cut DNA, the enzyme makes two incisions at each sugar phosphate backbone.

  1. CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)
    - immune memory for prokaryotes
    - DNA Sequencing of a variety of bacteria revealed many viral short viral
    DNA sequences integrated into the genome
    - can edit animal genomes
    - ethics in human gametic cell editing

Overview:
- Viral DNA separated by non-viral repetitive sequences
- Always nearby a cluster of genes encoding DNA cutting enzymes (cas
genes)

45
Q

What the basic steps of CRISPR in antiviral defense?

A
  1. Cutting of foreign viral DNA
  2. Insertion of DNA as a novel spacer in array
  3. Transcription of array, creation of RNAs that can target incoming viral DNA in a sequence-specific manner
  4. Cleavage of incoming viral DNA
46
Q

Explain RNA-guided cleavage of foreign viral DNA.

A
  1. Guide RNA is transcribed from CRISPR array on bacterial genome
  2. Loaded onto Cas9 nuclease (dsDNA-cutting enzyme)
  3. RNA guides Cas9 enzyme to cut homologous incoming viral dsDNA
47
Q

Why was the discovery of antiviral defences important?

A
  • shows these antiviral defense systems are complementary
  • discovery has revolutionized molecular biology and medicine
  • provides solid argument for basic, curiosity-driven research
48
Q

How do EUKARYOTES combat infection since they do not have restriction enzymes or CRISPR arrays? What are PPRS?

Why are non-host nucleic acids (viral aspects) foreign to the host?

A
  • have an immune system, triggered by pattern recognition receptors (PRR)

PPRS
- recognize molecular patterns that are foreign
- include molecules that recognize non-host nucleic acids, including viral dsRNA and 5’-triphosphate RNA
- These viral targets are called “pathogen-associated molecular patterns”, or “PAMPs”

Why foreign to the host?
- RNA in humans gets 5’ cap structure preventing agents degradation
- if this structure is lacking, it is deemed as foreign

49
Q

What is important to know generally about PAMPs and PRRs?

A

There are:
1. multiple viral patterns (PAMPs) that the cell can recognize, including nucleic
acids and proteins.
2. Multiple cellular pattern recognition receptors (PRRs) that do the sensing.

50
Q

How do viruses make gene products (RNA and protein) and replicate their genomes?

A

Viruses are obligate intracellular parasites:
- they depend on many parts of host biosynthetic machinery
- so viruses need to either:
(i) conform to the machinery (e.g. use host ribosomes for translation), or
(ii) alter that machinery for their own use.

51
Q

What are the three aspects of the central dogma of molecular biology? How does this operate usually?

A
  1. DNA-dependent DNA polymerase:
    - uses dsDNA as a template to make more dsDNA.
    - use enzymes
  2. RNA polymerase:
    - uses dsDNA as a template to make (+)-sense RNA
  3. Ribosomes:
    - translates (+)-sense RNA into an amino acid chain (protein)
    - +: translation competent, ready to be translated

Usual operation:
- DNA copied and decoded into RNA then decoded into protein

52
Q

What are the ways in which viruses often break the central dogma of molecular biology?

A
  • viruses can take RNA template(s) and copy them into more RNA
  • viruses can take RNA and copy it into DNA (retroviruses, reverse translation)
53
Q

What do you know about virus genomes?

A
  • All are either DNA are RNA
  • Their composition and structures are diverse (many possible tactics for encoding and decoding information in
    nucleic acid)
  • must make mRNA that can be decoded by host ribosomes
  • ie: seven types of viral genomes all have processes that end with viral mRNA production)
54
Q

What kind of information is encoded in viral genomes? What are the five factors the information is required for?

A

Gene products (RNA, protein) and regulatory signals. Which are required for:
1. Replication of the viral genome
2. Assembly and packaging of the genome into viral particles
3. Regulation and timing of the replication cycle
4. Defeating host defenses
5. Spread to other cells and hosts

55
Q

Describe all that you know of (+)ssRNA viruses. Use Poliovirus as an example.

A
  • directly translated in the cytoplasm (generating polyproteins, which subsequently get cleaved by either
    viral or host proteases into the full complement of viral proteins)
  • replicate their genomes by Viral RNA-dep RNA pol (RdRp)
  • can’t use host RdRp since doesn’t exist
56
Q

What would happen if you transfected (introduced) naked poliovirus RNA into a cell? Would you make progeny virions?

A

Yes, this mimics the encoding step within viral replication so all the viral DNA needs is to hijack translation and replicate creating progeny virions.

57
Q

Proteases and RDRP make excellent drug targets. Why?

A

Proteases
- humans don’t have proteases
- specificity can be achieved, inhibiting viral proteases

RDRP
- humans don’t have RDRP
- lots of strong chemicals may be used appropriately to inhibit function (ie: low toxicity antiviral drugs)

58
Q

How can we say that ‘polyprotein processing is co-translational’?

A

As soon as any portion of viral protein exists (finishes being transcribed), local folding occurs which creates protease active sites.

59
Q

What do you know of (+)ssRNA virus gene expression? How are long viral polyproteins are chopped up & by what?

A

Question 1:
- Genome mimics host mRNA and is directly translated in the cytoplasm to make long polyproteins.
- Enveloped (+)ssRNA viruses typically make polyproteins at the endoplasmic
reticulum, with many transmembrane domains.

Question 2:
- Long viral polyproteins are chopped up into individual proteins by host and viral proteases
- viral proteases typically release themselves from the polyprotein

60
Q

What do (+)ssRNA viruses produce to transcribe genomes? What RNA products result?

A
  • They make an RdRp required to transcribe the genome
  • different RNA products are (-)ssRNA intermediates and (+)ssRNA genomes
61
Q

How do (+)ssRNA viruses make a (-)ssRNA intermediate? What catalyzes these steps, is this enzyme susceptible to anything?

A

initial genome -> replicative intermediate -> progeny genomes

+ssRNA -> -ssRNA -> +ssRNA

  • Viral RdRp catalyzes these steps in genome replication
  • susceptible to the effects of ribavirin
62
Q

What are the mechanisms of action for Ribavirin?

A
  • nucleoside analogs
  • targets a lot (ex: influenza, hepatitis C, polio, measles, yellow fever, West Nile and dengue viruses)
  • inactive pro-drug which is (active once it is) phosphorylated in cells
  • incorporated into viral RNA during genome replication.
63
Q

What does it mean that Ribavirin can cause lethal hypermutation? Give quick example using Zika virus.

A
  • It can induce hypermutation in viruses that depend on RdRp (can be lethal to RNA viruses)
  • Base analog for adenine (A) and guanine (G), and can bind either uracil or cytosine

ex: Zika virus (+)ssRNA genome, replicates via (-)ssRNA intermediate

64
Q

What do you know about Hepatitis C virus? How is it transmitted? Why/how is there a potential for a cure?

A

Known:
- Transmitted from host to host via blood (also.. unprotected sex, bodily fluid exposure and IV drug use)
- Causes hepatitis (liver disease), which can sometimes progress to hepatocellular carcinoma (liver cancer).
- Liver transplant often necessary, but a challenge to prevent disease in new liver.

Cure?
- New anti-HCV drugs can be more than a treatment.
- Difficult to transmit
- No animal reservoir (only humans)

65
Q

What is the result of the drug Sofosbuvir inhibiting RdRp in Hep. C?

What is targeted by Hep. C virus drugs? Explain function generally?

A

Result?
- No viral RNA synthesis

Drug Target?
- Rdpd
- Sofosbuvir is a pro-drug that is converted into triphosphate form by cellular enzymes
- incorporated into growing HCV RNA molecule, causing chain termination

66
Q

Describe Coronavirus gene
expression? Give detailed steps.

Exception: Do not explain Ribosomal frame shifting in detail w/ this question, will be asked next.

A
  1. The incoming CoV genome is +ssRNA and ready-for-translation.
    - Translated directly from capped and
    polyadenylated genomic (+) RNA as two large polyproteins (aka many proteins w/o stop codons)
  2. CoV polyproteins are cleaved by multiple viral proteases
    - Need protease activity to release RdRp.
  3. Coronaviruses produce 2 different polyproteins using ribosomal frameshifting
    - Ribosomes slip & move into a new reading frame (often -1), and
    continue translation in a new frame.
    - Sequences in the viral genome encourage ribosomal frameshifting
  4. An RNA pseudoknot causes programmed ribosomal frameshifting
    - generates longer protein
  5. ER-localized transmembrane coronavirus proteins
    - These proteins drive creation of
    viral ‘factories’ in the cytoplasm
    known as ‘Replication Organelles’
    - Replication Organelles are made
    from viral proteins and stolen ER
    membranes
67
Q

Explain RNA pseudoknots & programmed ribosomal frameshifting.

Hints: What structure causes the ribosome to struggle to proceed? What does this stalling lead to? In what reading frame does protein synthesis resume? What codon is no longer encountered?

A
  • Ribosome struggles to proceed because of RNA secondary structure
  • While stalled, the ribosome slips on poly U/A sequence, shifting the reading frame by one position.
  • Protein synthesis resumes, but in the -1 reading frame.
  • Stop codon is no longer encountered in the new frame.
68
Q

What do viral drugs such as Sofosbuvir (HCV) and Remdesivir (coronavirus) have in common?

A
  • inhibit RdRp synthesis (aka no viral RNA synthesis)
  • causes chain termination
69
Q

What two things does RdRp do in viral Replication Organelles? How is it produced generally?

A
  1. Copies the genome
    - (+)ssRNA genome to (-)ssRNA intermediate to (+)ssRNA product
  2. Make shorter “sub-genomic” +ssRNAs that encode structural proteins
    - ex: Spike

Produced?
- frame shifting is needed to make RdRp!

70
Q

What are two main features of viral Replication Organelles?

A
  1. Hides viral RNA from PRRs
  2. Crown-shaped pore
    - allows ribonucleotides in
    - lets viral RNAs exit to be translated in cytoplasm
    - or full-length genomes to be packaged into new viral particles.
71
Q

What is essential for Influenza A virus infection (sp. protein & host)? State anything you may know about Influenza A.

A

Infection & hosts:
- glycoproteins are essential for infection
- birds, bats, humans are hosts

Other influ. A virus info:
- new virus subtypes cause pandemics (deadly)
- subtypes are based on antigenically distinct HA and NA glycoproteins
- Masks were used during the 1918-19 influenza pandemic
- Influenza vaccines are reformulated each year because circulating strains
change (evolve rapidly)

72
Q

Explain HA & NA glycoprotein processes? How many of each for Influenza A viruses?

A

HA
- binds to sialic acid
- promotes attachment and entry
- Influ. A has 18

NA (neuraminidase)
- an enzyme that removes sialic acid from the cell surface
- allows progeny influenza viruses to escape the cell and spread to new
hosts
- Influ. A has 11

73
Q

What do you know about Influenza B viruses?

A
  • also divided based on antigenically distinct HA and NA glycoproteins
  • only circulate in humans and cause
    seasonal disease
  • Currently circulating influenza B virus strains are in the ’Victoria’ and ‘Yamagata’ lineages (& are antigenically distinct)
  • do not cause pandemics
74
Q

Explain Antigenic drift & Antigenic shift as it pertains to Influenza.

A

Antigenic Drift
- rapid mutation and selection of new strains that escape neutralizing antibodies
- ie: the antigens that drift are (mostly) located on HA and host antibodies can neutralize Influenza viruses

Antigenic Shift
- from spillover events from animal hosts into humans, and the segmented genome that allows swapping of genetic material between viruses.
- Entirely new viruses, not recognized by immune system
- major & abrupt changes

75
Q

In influenza, the structural RdRp is attached to what part?

What is it required to decode (–)ssRNA into? Give three examples.

A
  • structural viral RdRp is attached to each (-) ssRNA genome segment
  • RdRp is error-prone, which support rapid evolution

Required to decode (–)ssRNA into:
1. viral mRNAs with 5’ caps and poly(A) tails.
2. (+)ssRNA replication intermediates.
3. (-)ssRNA genome copies.

76
Q

Where is the location of commonly mutated amino acids in HA? Why is this?

A
  • The location of commonly mutated amino acids in HA is clustered in receptor-binding regions.
  • Why? Effective antibodies neutralize the virus and antibodies block attachment and entry

More notes:
- Successful viruses evade neutralizing
antibodies
- Antigenic drift is a frequent cause of vaccine mismatches

77
Q

H2N2 pandemic influenza A virus emerged in 1957. Was this the result of excessive mutation, or was there an animal reservoir? (What type of shift?)

A
  • Antigenic Shift is the major source of zoonotic influenza virus infections
  • Target cells can be co-infected by different strains of Influenza virus
  • This can allow the reassortment of genome segments in viral progeny
78
Q

What do you know about, antigenic shifts in terms of, reassortment(s) of influenza virus genome segments?

A
  • Most reassortments result in defective viruses that do not spread.
  • Some confer advantageous combinations of genes, giving viruses enhanced abilities to spread and evade antiviral defenses and neutralizing antibodies.
79
Q

How do we select the antigens a vaccine? Aka describe the genetic system steps used for creating flu vaccines!

A
  1. Clone selected HA and NA genes into plasmids.
  2. Combine with 6 remaining ’standard’ genome segments
  3. Insert 8 plasmids into cells
  4. Harvest hybrid viruses that have correct HA/NA antigens at the surface
  5. Use to inoculate eggs(?)/desired cells
80
Q

What antigens are in our current influenza vaccines?

A

The current quadrivalent egg-based vaccine has:
- Influenza A virus H1N1
- Influenza A virus H3N2
- Influenza B virus HA and NA (Victoria lineage)
- Influenza B virus HA and NA (Yamagata lineage)

81
Q

How do viruses escape the host cell? Differentiate between non-enveloped and enveloped.

A
  • Non-enveloped progeny viruses can escape by killing the cell (i.e. ”break stuff and get out”)
  • Enveloped progeny viruses escape the cell in the process of acquiring their lipid envelope:
    a) by budding into an internal membrane, followed by
    exocytosis (e.g. coronaviruses, herpesviruses)
    b) by budding directly from the plasma membrane (e.g. influenza viruses, HIV)
82
Q

How are Influenza viruses assembled?

A
  1. HA and NA proteins bear hydrophobic signal sequences then translated and processed in the endoplasmic reticulum (ER), and
    proceed through the Golgi apparatus, ultimately becoming embedded in
    the plasma membrane
    - glycoslayoted and cleaved by proteases in the Golgi apparatus
  2. HA and NA cluster in the plasma membrane
  3. Viral proteins and genome ‘home in’ on the HA/NA cluster and assembly
    takes place. Virion buds from the plasma membrane.
  4. NA (which has neuraminidase – or sialic acid cleaving activity) digests the
    local sialic acid sugars, which will allow the budding virus to escape the
    cell surface.
    - Antiviral drugs Tamiflu and Relenza are neuraminidase inhibitors (keep
    influenza virus tethered to cell surface)
83
Q

Each influenza virus genome segment has a packaging signal required for incorporation. Where is the signal?

A
  • Packaging signals are located at the ends of each genome segment.
  • Each segment is covered in NP and has RdRp bound to termini (structural proteins)
84
Q

How are Herpes viruses assembled?

A
  • Herpesvirus capsids are enveloped INSIDE the cell and use the normal protein secretory pathway for egress

Notes:
- Primary envelopment at nuclear envelope, purely to escape the nucleus
- Secondary envelopment at the trans Golgi network
- Uses secretory pathway to exit the cell

85
Q

During CoV assembly structural proteins are made but where? What type of RNA is produced?

A
  • Structural glycoproteins are made in the ER and accumulate in the ERGIC (the site of CoV assembly)…ERGIC = ER-Golgi Intermediate Compartment
  • CoV +ssRNA genome is coated with N
    and recruited to ERGIC for assembly
86
Q

How is the Coronavirus structure made and released from a cell?

A
  • Coronaviruses acquire their envelopes and structural proteins INSIDE the cell and escape the cell via secretory pathway.
  • Transcription and genome replication happen in a custom-built compartment in the cytoplasm
87
Q

In the cytoplasm, transcription occurs for Coronaviruses. But what two things does RdRp do?

A
  1. Copy the genome: +ssRNA genome to -ssRNA intermediate to +ssRNA product
  2. Make shorter “sub-genomic” +ssRNAs (mRNAs) that encode structural proteins
88
Q

BIG OVERVIEW of Coronavirus Assembly & Maturation…

A

CoVs have +ssRNA genomes (5’cap and polyA tail, resemble mRNA) that are
directly translated in the cytoplasm

15 non-structural proteins produced from polyprotein include 2 proteases and the RdRp

Some non-structural proteins build the cytoplasmic replication compartment.

Inside this ‘factory’ the RdRp synthesizes sub-genomic mRNAs and full-length +ssRNA genomes. Both types of RNA are exported from the factory via a crown-shaped pore.

  1. The subgenomic +ssRNAs encoding structural proteins are translated on
    ribosomes.
  2. The full-length genomic RNA is coated with N, and moves to the surface of the RGIC, where it binds to viral structural glycoproteins and assembles by budding into the ERGIC. Only the full-length genomic RNAs are selected, because they have a packaging site not found on the smaller sub-genomic RNAs.

These finished viral particles leave the cell via exocytosis.