Pages

Is there such a thing as a completely broad-spectrum antiviral?

I'm sure everyone is aware of the kind of effects virus infection and replication has on the health of humans and other animals (just scroll along my last blog posts and you'll see). 

It's really not good.

And, in most cases we don't have much to prevent or cure it: maybe a vaccine here, some antivirals there yet what we really would like is something that would act against ALL kinds of viruses, from influenzas to smallpox to ebola and even HIV. Most vaccines and antivirals target a very limited number of pathogens, usually only one. The search for this class of antiviral drugs has eluded us in the past yet a paper published this week in PLoS ONE reports (read paper here) the identification of an effective strategy termed Double-stranded RNA (dsRNA) Activated Caspase Oligomerizer or DRACO. See this MIT group's website here.

dsRNA - an important hallmark of viral infection
This approach takes advantage of chiefly two natural processes that occur within cells: the intracellular sensing of long double-stranded RNA molecules and the initiation of programmed cell death, known as apoptosis via what are called as caspase enzymes. Through linking these two pathways together via the construction of a novel protein molecule, this group aim to inhibit viral replication - and hence act as a broad-range antiviral as most known viruses generate dsRNAs.

How and why do cells 'see' ds-RNA?


ds-RNA binding domain from PKR protein
Our cells - and those of nearly every other organism - have a range of defenses to protect themselves from viral infection. One being, the ability to detect when they have been initially infected. For example: through the binding of double- stranded RNA molecules, which viruses produce a lot of. Think of it as a hallmark of virus replication and our cells naturally come equipped with a range of these detector modules that carry out this function using a double-stranded RNA binding motif found somewhere in their structure. Detection of ds-RNA sets off a serious of downstream reactions leading to the cell's normal antiviral response.



Examples:


What about apoptosis?

Apoptosis is a highly coordinated process leading to the self-destruction of the cell without any of the nasty inflammatory responses. The main players in this cellular suicide are the caspases - protease enzymes that specifically target and degrade hundreds of other proteins inside the cell in a well-defined cascade of activity. These proteins are activated through the construction of a larger complexes which themselves are produced as a response to certain noxious stimuli, one example is virus infection. One domain are the death effector domains (an example is a FADD domain) that recruit the initiator caspases into the death-induced signalling complex (DISC) thereby kick-starting the whole process of apoptosis and the cells' death.

The FADD death effector domain for example was added to the end of the ds-RNA binding protein

I said above that virus infection itself activates apoptosis so why doesn't every virus-infected cell just die? Well, viruses have evolved neat tricks to circumvent this response as in most circumstances the death of the cell leads to the end of the infection. The hypothesis that this group are working under is that what if we could couple these two processes more-so than they are naturally as to sidestep the viruses' strategies at combating apoptosis, in a way, short-circuiting this whole pathway. Instead of going through long networks of protein-protein interactions, make it rely only on a single binding. 

So what did they do?

These DRACO proteins are chimeras of the two classes of molecules described above: one half ds-RNA binding and the other a death effector domain. And, finally, in order to get this protein into the cell, they added a 'transduction tag' - a short peptide sequence that is naturally taken up inside cells. So, does it work?

They initially made a whole range of these proteins, using different RNA binding motifs combined with a number of dead effector domains (see below). These recombinant proteins were then tested to see which was the best at getting into cells, binding ds-RNA, triggering apoptosis and most importantly, which was the safest.

The chimeric DRACO proteins (transduction tag in yellow, ds-RNA binding in red and apoptosis in green and blue)

Then they show that these proteins can indeed get into cells and in the presence of ds-RNA they can initiate apoptosis - see below. Also, these proteins were generally well tolerated by these particular cells used.



But does this actually inhibit virus replication?

To test this, they first added their proteins to the cells and then infected them with a virus - in this case human rhinovirus 1B (an important human pathogen) - see data below. Virus infection without administration of DRACO's resulted in 0% cell viability, that is all the cells were killed by the virus. When the proteins were added, viability increased to about 80%. The authors suggest that this because the proteins kill off any virus-infected cells very early thus preventing infection from spreading any further. They also test how effective the strategy is when we add the proteins at different times. It works best when added before infection and then decreases substantially (when added 1 day after infection, viability dropped to 50%).

The antiviral effectiveness of 5 different DRACO proteins in an in vitro rhinovirus infection. The higher the cell viability the better the approach.
Nicely, they extended this same analysis to a very wide range of viruses: dengue, influenza, arenaviruses, adenoviruses, reoviruses - anything they could their hands on. And they all worked much the same as the rhinovirus example above. These viruses are incredibly different from each other, showing just how broadly active this approach is. Different cell lines were also investigated opening the possibility of this approach in a veterinary perspective.

We know that sometimes what works in cell culture doesn't necessarily work in an animal, so the group tried their system in a mouse model of lethal influenza virus infection. When administered prior to challenge, the DRACOs had great results 0 at most 80% of the mice survived, compared to 0 without the proteins. They didn't check out what would happen if they added the DRACOs after infection, which would more closely mimic the situation in real-life.


So, is there such a thing as a completely broad-spectrum antiviral? If there wasn't one before this, there certainly is now after these promising initial results. Although further work will have to establish the exact mechanism of this approach and also apply it to other viruses in more model systems.

To sum up with what the author's say:

DRACOs should be effective against numerous clinical and NIAID priority viruses, due to the broad-spectrum sensitivity of the dsRNA detection domain, the potent activity of the apoptosis induction domain, and the novel direct linkage between the two which viruses have never encountered. We have demonstrated that DRACOs are effective against viruses with DNA, dsRNA, positive-sense ssRNA, and negative-sense ssRNA genomes; enveloped and non-enveloped viruses; viruses that replicate in the cytoplasm and viruses that replicate in the nucleus; human, bat, and rodent viruses; and viruses that use a variety of cellular receptors

ResearchBlogging.org 
Rider, T., Zook, C., Boettcher, T., Wick, S., Pancoast, J., & Zusman, B. (2011). Broad-Spectrum Antiviral Therapeutics PLoS ONE, 6 (7) DOI: 10.1371/journal.pone.0022572

No comments:

Post a Comment

Markup Key:
- <b>bold</b> = bold
- <i>italic</i> = italic
- <a href="http://www.fieldofscience.com/">FoS</a> = FoS