back to index

Dmitry Korkin: Computational Biology of Coronavirus | Lex Fridman Podcast #90


small model | large model

link |
00:00:00.000
The following is a conversation with Dmitry Korkin.
link |
00:00:02.720
He's a professor of bioinformatics
link |
00:00:04.560
and computational biology at WPI,
link |
00:00:07.280
Worcester Polytechnic Institute,
link |
00:00:09.320
where he specializes in bioinformatics
link |
00:00:11.400
of complex diseases, computational genomics,
link |
00:00:14.600
systems biology, and biomedical data analytics.
link |
00:00:18.360
I came across Dmitry's work when in February,
link |
00:00:21.400
his group used the viral genome of the COVID 19
link |
00:00:25.040
to reconstruct the 3D structure of its major viral proteins
link |
00:00:29.040
and their interaction with the human proteins.
link |
00:00:32.360
In effect, creating a structural genomics map
link |
00:00:34.960
of the coronavirus and making this data open
link |
00:00:37.560
and available to researchers everywhere.
link |
00:00:40.200
We talked about the biology of COVID 19,
link |
00:00:42.360
SARS, and viruses in general,
link |
00:00:44.520
and how computational methods can help us understand
link |
00:00:47.760
their structure and function
link |
00:00:49.400
in order to develop antiviral drugs and vaccines.
link |
00:00:54.040
This conversation was recorded recently
link |
00:00:56.360
in the time of the coronavirus pandemic
link |
00:00:58.720
for everyone feeling the medical, psychological,
link |
00:01:01.080
and financial burden of this crisis.
link |
00:01:03.040
I'm sending love your way.
link |
00:01:04.800
Stay strong.
link |
00:01:05.880
We're in this together.
link |
00:01:06.920
We'll beat this thing.
link |
00:01:09.160
This is the Artificial Intelligence Podcast.
link |
00:01:11.560
If you enjoy it, subscribe on YouTube,
link |
00:01:13.720
review it with five stars on Apple Podcast,
link |
00:01:16.080
support it on Patreon,
link |
00:01:17.400
or simply connect with me on Twitter
link |
00:01:19.440
at Lex Friedman, spelled F R I D M A N.
link |
00:01:23.520
This show is presented by Cash App,
link |
00:01:25.600
the number one finance app in the app store.
link |
00:01:27.760
When you get it, use code LEXBODCAST.
link |
00:01:30.600
Cash App lets you send money to friends,
link |
00:01:32.880
buy Bitcoin, and invest in the stock market
link |
00:01:35.000
with as little as $1.
link |
00:01:36.720
Since Cash App allows you to buy Bitcoin,
link |
00:01:38.920
let me mention that cryptocurrency
link |
00:01:40.880
in the context of the history of money is fascinating.
link |
00:01:44.040
I recommend Ascent of Money
link |
00:01:46.160
as a great book on this history.
link |
00:01:48.240
Debits and credits on ledgers
link |
00:01:49.880
started around 30,000 years ago.
link |
00:01:52.800
The US dollar created over 200 years ago.
link |
00:01:55.720
And Bitcoin, the first decentralized cryptocurrency,
link |
00:01:58.480
released just over 10 years ago.
link |
00:02:00.640
So given that history, cryptocurrency is still very much
link |
00:02:04.160
in its early days of development,
link |
00:02:06.040
but it's still aiming to,
link |
00:02:07.600
and just might, redefine the nature of money.
link |
00:02:12.800
So again, if you get Cash App from the app store,
link |
00:02:15.160
Google Play, and use the code LEXBODCAST,
link |
00:02:18.280
you get $10, and Cash App will also donate $10 to FIRST,
link |
00:02:22.200
an organization that is helping to advance robotics
link |
00:02:25.040
and STEM education for young people around the world.
link |
00:02:28.480
And now, here's my conversation with Dmitry Korkin.
link |
00:02:33.320
Do you find viruses terrifying or fascinating?
link |
00:02:38.040
When I think about viruses, I think about them,
link |
00:02:42.320
I mean, I imagine them as those villains
link |
00:02:47.680
that do their work so perfectly well.
link |
00:02:52.680
That is impossible not to be fascinated with them.
link |
00:02:57.600
So what do you imagine when you think about a virus?
link |
00:03:00.040
Do you imagine the individual,
link |
00:03:02.840
sort of these 100 nanometer particle things?
link |
00:03:07.280
Or do you imagine the whole pandemic, like society level,
link |
00:03:11.960
when you say the efficiency at which they do their work,
link |
00:03:15.200
do you think of viruses as the millions
link |
00:03:18.960
that occupy human body or living organism,
link |
00:03:23.560
society level, like spreading as a pandemic,
link |
00:03:26.680
or do you think of the individual little guy?
link |
00:03:29.240
Yes, I think this is a unique concept
link |
00:03:34.680
that allows you to move from micro scale to the macro scale.
link |
00:03:40.000
So the virus itself, I mean, it's not a living organism.
link |
00:03:45.080
It's a machine to me, it's a machine.
link |
00:03:48.800
But it is perfected to the way
link |
00:03:51.720
that it essentially has a limited number of functions,
link |
00:03:57.000
it needs to do necessary functions.
link |
00:04:00.560
And it essentially has enough information
link |
00:04:05.840
just to do those functions,
link |
00:04:07.760
as well as the ability to modify itself.
link |
00:04:13.400
So it's a machine, it's an intelligent machine.
link |
00:04:18.280
So yeah, look, maybe on that point,
link |
00:04:20.240
you're in danger of reducing the power of this thing
link |
00:04:23.320
by calling it a machine, right?
link |
00:04:26.880
But you now mentioned that it's also possibly intelligent.
link |
00:04:30.520
It seems that there is these elements of brilliance
link |
00:04:34.360
that a virus has, of intelligence,
link |
00:04:37.520
of maximizing so many things about its behavior
link |
00:04:42.680
and to ensure its survival and its success.
link |
00:04:45.680
So do you see it as intelligent?
link |
00:04:48.680
So, you know, I think it's a different,
link |
00:04:53.200
I understand it differently than, you know,
link |
00:04:55.800
I think about, you know, intelligence of humankind
link |
00:05:00.000
or intelligence of the artificial intelligence mechanisms.
link |
00:05:10.560
I think the intelligence of a virus
link |
00:05:14.040
is in its simplicity.
link |
00:05:18.280
The ability to do so much
link |
00:05:23.800
with so little material and information.
link |
00:05:31.160
But also, I think it's interesting.
link |
00:05:33.680
It keeps me thinking, you know,
link |
00:05:35.720
it keeps me wondering whether or not it's also the,
link |
00:05:40.720
an example of the basic swarm intelligence
link |
00:05:48.200
where, you know, essentially, the viruses act as the whole
link |
00:05:54.800
and they're extremely efficient in that.
link |
00:05:59.320
So what do you attribute the incredible simplicity
link |
00:06:02.040
and the efficiency to?
link |
00:06:04.360
Is it the evolutionary process?
link |
00:06:06.560
So maybe another way to ask that question
link |
00:06:08.560
is if you look at the next hundred years,
link |
00:06:11.400
are you more worried about the natural pandemics
link |
00:06:15.360
or the engineered pandemics?
link |
00:06:17.400
So how hard is it to build a virus?
link |
00:06:20.240
Yes, it's a very, very interesting question
link |
00:06:23.240
because obviously there's a lot of conversations
link |
00:06:27.680
about the, you know, whether we are capable
link |
00:06:32.680
of engineering a, you know, an even worse virus.
link |
00:06:39.280
I personally expect and am mostly concerned
link |
00:06:44.280
with the naturally occurring viruses
link |
00:06:47.840
simply because we keep seeing that.
link |
00:06:51.680
We keep seeing new strains of influenza emerging,
link |
00:06:56.320
some of them becoming pandemic.
link |
00:06:58.720
We keep seeing new strains of coronaviruses emerging.
link |
00:07:04.400
This is a natural process
link |
00:07:06.360
and I think this is why it's so powerful.
link |
00:07:14.400
You know, if you ask me, you know,
link |
00:07:17.400
I've read papers about scientists trying to study
link |
00:07:22.400
the capacity of the modern, you know,
link |
00:07:27.400
biotechnology to alter the viruses.
link |
00:07:33.440
But I hope that, you know,
link |
00:07:36.440
it won't be our main concern in the nearest future.
link |
00:07:43.400
What do you mean by hope?
link |
00:07:46.400
Well, you know, if you look back and look at the history
link |
00:07:51.400
of the most dangerous viruses, right?
link |
00:07:55.400
So the first thing that comes into mind is a smallpox.
link |
00:08:02.400
So right now there is perhaps a handful of places
link |
00:08:08.400
where there is a smallpox.
link |
00:08:11.400
There is perhaps a handful of places where this,
link |
00:08:17.400
you know, the strains of this virus are stored, right?
link |
00:08:22.400
So this is essentially the effort of the whole society
link |
00:08:27.400
to limit the access to those viruses.
link |
00:08:32.400
You mean in a lab in a controlled environment
link |
00:08:34.400
in order to study? Correct.
link |
00:08:36.400
And then smallpox is one of the viruses
link |
00:08:38.400
that should be stated there's a vaccine is developed.
link |
00:08:43.400
Yes, yes.
link |
00:08:44.400
And that's, you know, it's until 70s,
link |
00:08:48.400
I mean, in my opinion, it was perhaps
link |
00:08:52.400
the most dangerous thing that was there.
link |
00:08:56.400
Is that a very different virus
link |
00:08:58.400
than the influenza and the coronaviruses?
link |
00:09:03.400
It is, it is different in several aspects.
link |
00:09:08.400
Biologically, it's a so called double stranded DNA virus,
link |
00:09:15.400
but also in the way that it is much more contagious.
link |
00:09:24.400
So the R naught for, so this is the...
link |
00:09:29.400
What's R naught?
link |
00:09:30.400
R naught is essentially an average number
link |
00:09:35.400
as person infected by the virus can spread to other people.
link |
00:09:42.400
So then the average number of people
link |
00:09:44.400
that he or she can, you know, spread it to.
link |
00:09:50.400
And, you know, there is still some, you know,
link |
00:09:54.400
discussion about the estimates of the current virus,
link |
00:09:59.400
you know, the estimations vary between, you know, 1.5 and 3.
link |
00:10:06.400
In case of smallpox, it was 5 to 7.
link |
00:10:13.400
And we're talking about the exponential growth, right?
link |
00:10:17.400
So that's a very big difference.
link |
00:10:23.400
It's not the most contagious one.
link |
00:10:25.400
Measles, for example, it's, I think, 15 and up.
link |
00:10:30.400
So it's, you know, but it's definitely more contagious
link |
00:10:38.400
that the seasonal flu than the current coronavirus
link |
00:10:45.400
or SARS for that matter.
link |
00:10:48.400
What makes a virus more contagious?
link |
00:10:52.400
I'm sure there's a lot of variables that come into play,
link |
00:10:54.400
but is it that whole discussion of aerosol
link |
00:10:58.400
and like the size of droplets if it's airborne,
link |
00:11:01.400
or is there some other stuff that's more biology centered?
link |
00:11:04.400
I mean, there are a lot of components
link |
00:11:06.400
and there are biological components
link |
00:11:09.400
that there are also, you know, social components.
link |
00:11:14.400
The ability of the virus to, you know,
link |
00:11:17.400
so the ways in which the virus is spread is definitely one.
link |
00:11:21.400
The ability of the virus to stay on the surfaces, to survive.
link |
00:11:27.400
The ability of the virus to replicate fast or so, you know.
link |
00:11:33.400
Or once it's in the cell or whatever.
link |
00:11:35.400
Once it's inside the host.
link |
00:11:38.400
And interestingly enough, something that I think
link |
00:11:42.400
we didn't pay that much attention to is the incubation period.
link |
00:11:49.400
The, where, you know, hosts are symptomatic.
link |
00:11:52.400
And now it turns out that another thing that we,
link |
00:11:55.400
one really needs to take into account,
link |
00:12:00.400
the percentage of the symptomatic population.
link |
00:12:04.400
Because those people still shed this virus
link |
00:12:08.400
and still are, you know, they still are contagious.
link |
00:12:12.400
So there's an, the Iceland study,
link |
00:12:14.400
which I think is probably the most impressive size wise,
link |
00:12:17.400
shows 50% asymptomatic for this virus.
link |
00:12:22.400
I also recently learned the swine flu is,
link |
00:12:28.400
like the, just the number of people who got infected
link |
00:12:33.400
was in the billions.
link |
00:12:35.400
It was some crazy number.
link |
00:12:37.400
It was like, it was like, like 20% of the pop,
link |
00:12:41.400
30% of the population, something crazy like that.
link |
00:12:44.400
So the lucky thing there is the fatality rate is low,
link |
00:12:49.400
but the fact that a virus can just take over
link |
00:12:52.400
an entire population so quickly, it's terrifying.
link |
00:12:56.400
I think, I mean, this is, you know,
link |
00:12:59.400
that's perhaps my favorite example of a butterfly effect
link |
00:13:04.400
because it's really, I mean, it's even tinier than a butterfly
link |
00:13:09.400
and look at, you know, and with, you know,
link |
00:13:11.400
if you think about it, right.
link |
00:13:13.400
It used to be in those bat species.
link |
00:13:19.400
And perhaps because of, you know,
link |
00:13:22.400
a couple of small changes in the viral genome,
link |
00:13:28.400
it first had, you know, become capable of jumping
link |
00:13:32.400
from bats to human, and then it became capable
link |
00:13:36.400
of jumping from human to human, right.
link |
00:13:39.400
So this is, I mean, it's not even the size of a virus.
link |
00:13:42.400
It's the size of several, you know, several atoms
link |
00:13:46.400
or a few atoms.
link |
00:13:50.400
And all of a sudden this change has such a major impact.
link |
00:13:57.400
So is that a mutation like on a single virus?
link |
00:14:01.400
Is that like, so if we talk about those,
link |
00:14:04.400
the flap of a butterfly wing, like what's the first flap?
link |
00:14:08.400
Well, I think this is the mutations that make,
link |
00:14:12.400
that made this virus capable of jumping
link |
00:14:17.400
from bat species to human.
link |
00:14:20.400
Of course there's, you know, the scientists are still trying
link |
00:14:23.400
to find, I mean, they're still even trying to find
link |
00:14:26.400
who was the first infected, right, the patient zero.
link |
00:14:30.400
The first human.
link |
00:14:31.400
The first human infected, right.
link |
00:14:34.400
I mean, the fact that there are coronaviruses,
link |
00:14:38.400
different strains of coronaviruses
link |
00:14:40.400
in various bat species, I mean, we know that.
link |
00:14:43.400
So we, you know, virologists observe them.
link |
00:14:47.400
They study them.
link |
00:14:48.400
They look at their genomic sequences.
link |
00:14:51.400
They're trying, of course, to understand what make
link |
00:14:55.400
these viruses to jump from bats to human.
link |
00:15:01.400
Because, you know, similar to that in influenza,
link |
00:15:05.400
there was, I think a few years ago, there was this,
link |
00:15:09.400
you know, interesting story where several groups
link |
00:15:16.400
of scientists studying influenza virus essentially,
link |
00:15:21.400
you know, made experiments to show that this virus
link |
00:15:25.400
can jump from one species to another,
link |
00:15:29.400
you know, by changing, I think, just a couple of residues.
link |
00:15:34.400
And, of course, it was very controversial.
link |
00:15:38.400
I think there was a moratorium on this study for a while.
link |
00:15:43.400
But then the study was released.
link |
00:15:45.400
It was published.
link |
00:15:47.400
So that, why was there a moratorium?
link |
00:15:49.400
Because it shows through engineering it,
link |
00:15:52.400
through modifying it, you can make it jump.
link |
00:15:55.400
Yes.
link |
00:15:57.400
I personally think it is important to study this.
link |
00:16:02.400
I mean, we should be informed.
link |
00:16:05.400
We should try to understand as much as possible
link |
00:16:08.400
in order to prevent it.
link |
00:16:10.400
But so then the engineering aspect there is,
link |
00:16:14.400
can't you then just start searching because there's
link |
00:16:18.400
so many strands of viruses out there.
link |
00:16:20.400
Can't you just search for the ones in bats that are
link |
00:16:24.400
the deadliest from the virologist perspective
link |
00:16:28.400
and then just try to engineer, try to see how to.
link |
00:16:33.400
But see, there's a nice aspect to it.
link |
00:16:37.400
The really nice thing about engineering viruses,
link |
00:16:41.400
it has the same problem as nuclear weapons.
link |
00:16:44.400
It's hard for it to not lead to mutual self destruction.
link |
00:16:49.400
So you can't control a virus.
link |
00:16:51.400
It can't be used as a weapon, right?
link |
00:16:53.400
Yeah, that's why in the beginning I said,
link |
00:16:56.400
I'm hopeful because there are definitely regulations
link |
00:17:02.400
needed to be introduced.
link |
00:17:05.400
And I mean, as the scientific society is,
link |
00:17:10.400
we are in charge of making the right actions,
link |
00:17:17.400
making the right decisions.
link |
00:17:19.400
But I think we will benefit tremendously by understanding
link |
00:17:25.400
the mechanisms by which the virus can jump,
link |
00:17:31.400
by which the virus can become more dangerous to humans
link |
00:17:40.400
because all these answers would eventually lead to designing
link |
00:17:47.400
better vaccines, hopefully universal vaccines, right?
link |
00:17:50.400
And that would be a triumph of science.
link |
00:17:56.400
So what's the universal vaccine?
link |
00:17:58.400
So is that something that, how universal is universal?
link |
00:18:02.400
Well, I mean, you know, so what's the dream, I guess,
link |
00:18:04.400
because you kind of mentioned the dream of this.
link |
00:18:06.400
I would be extremely happy if we designed the vaccine
link |
00:18:13.400
that is able, I mean, I'll give you an example.
link |
00:18:16.400
So every year we do a seasonal flu shot.
link |
00:18:21.400
The reason we do it is because, you know, we are in the arms race,
link |
00:18:25.400
you know, our vaccines are in the arms race
link |
00:18:28.400
with constantly changing virus, right?
link |
00:18:32.400
Now, if the next pandemic, influenza pandemic will occur,
link |
00:18:39.400
most likely this vaccine would not save us, right?
link |
00:18:43.400
Although it's, you know, it's the same virus,
link |
00:18:48.400
might be different strain.
link |
00:18:52.400
So if we're able to essentially design a vaccine against,
link |
00:18:58.400
you know, influenza A virus, no matter what's the strain,
link |
00:19:02.400
no matter which species did it jump from, that would be,
link |
00:19:08.400
I think that would be a huge, huge progress and advancement.
link |
00:19:13.400
You mentioned the smallpox until the 70s,
link |
00:19:16.400
might've been something that you would be worried the most about.
link |
00:19:20.400
What about these days?
link |
00:19:22.400
Well, we're sitting here in the middle of a COVID 19 pandemic,
link |
00:19:28.400
but these days, nevertheless, what is your biggest worry virus wise?
link |
00:19:33.400
What are you keeping your eye out on?
link |
00:19:37.400
It looks like, you know, based on the past several years
link |
00:19:43.400
of the new viruses emerging,
link |
00:19:47.400
I think we're still dealing with different types of influence.
link |
00:19:55.400
I mean, so the H7N9 avian flu that emerged,
link |
00:20:04.400
I think a couple of years ago in China,
link |
00:20:07.400
I think the mortality rate was incredible.
link |
00:20:13.400
I mean, it was, you know, I think about 30%, you know,
link |
00:20:18.400
so this is, this is huge.
link |
00:20:20.400
I mean, luckily for us, this strain was not pandemic, right?
link |
00:20:26.400
So it was jumping from birds to human,
link |
00:20:29.400
but I don't think it was actually transmittable between the humans.
link |
00:20:34.400
And, you know, this is actually a very interesting question,
link |
00:20:38.400
which scientists try to understand, right?
link |
00:20:42.400
So the balance, the delicate balance between the virus being very contagious,
link |
00:20:47.400
right, so efficient in spreading and virus to be very pathogenic,
link |
00:20:55.400
you know, causing, you know, harms, you know, and that's to the horse.
link |
00:21:04.400
So it looks like that the more pathogenic the virus is,
link |
00:21:10.400
the less contagious it is.
link |
00:21:13.400
Is that a property of biology or what is it?
link |
00:21:17.400
I don't have an answer to that.
link |
00:21:19.400
And I think this is still an open question.
link |
00:21:22.400
But, you know, if you look at, you know, with the coronavirus, for example,
link |
00:21:28.400
if you look at, you know, the deadlier relative MERS,
link |
00:21:34.400
MERS was never a pandemic virus.
link |
00:21:39.400
Right.
link |
00:21:40.400
But, you know, again, the mortality rate from MERS is far above,
link |
00:21:46.400
you know, I think 20 or 30%, so.
link |
00:21:52.400
So whatever is making this all happen doesn't want us dead
link |
00:21:57.400
because it's balancing out nicely.
link |
00:21:59.400
I mean, how do you explain that we're not dead yet?
link |
00:22:05.400
Like, because there's so many viruses and they're so good at what they do.
link |
00:22:11.400
Why do they keep us alive?
link |
00:22:14.400
I mean, we also have, you know, a lot of protection.
link |
00:22:18.400
Right.
link |
00:22:19.400
So we do the immune system.
link |
00:22:21.400
And so, I mean, we do have, you know, ways to fight against those viruses.
link |
00:22:31.400
And I think with the now we're much better equipped.
link |
00:22:35.400
Right.
link |
00:22:36.400
So with the discoveries of vaccines and, you know,
link |
00:22:39.400
there are vaccines against the viruses that maybe 200 years ago
link |
00:22:46.400
would wipe us out completely.
link |
00:22:50.400
But because of these vaccines, we are actually, we are capable of eradicating
link |
00:22:55.400
pretty much fully as is the case with smallpox.
link |
00:22:58.400
So if we could, can we go to the basics a little bit of the biology of the virus?
link |
00:23:04.400
How does a virus infect the body?
link |
00:23:07.400
So I think there are some key steps that the virus needs to perform.
link |
00:23:13.400
And of course, the first one, the viral particle needs to get attached to the host cell.
link |
00:23:21.400
In the case of coronavirus, there is a lot of evidence that it actually interacts
link |
00:23:28.400
in the same way as the SARS coronavirus.
link |
00:23:33.400
So it gets attached to AC2 human receptor.
link |
00:23:38.400
And so there is, I mean, as we speak, there is a growing number of papers suggesting it.
link |
00:23:45.400
Moreover, most recent, I think most recent results suggest that this virus
link |
00:23:53.400
attaches more efficiently to this human receptor than SARS.
link |
00:23:59.400
So just to sort of back off, so there is a family of viruses that are coronaviruses
link |
00:24:06.400
and SARS, whatever the heck for that, whatever that stands for.
link |
00:24:11.400
So SARS actually stands for the disease that you get is the syndrome of acute respiratory syndrome.
link |
00:24:20.400
So SARS is the first strand and then there's MERS.
link |
00:24:24.400
And there is, yes, scientists actually know more than three strands.
link |
00:24:32.400
I mean, so there is the MHV strain, which is considered to be a canonical disease model in mice.
link |
00:24:46.400
And so there is a lot of work done on this virus because it's...
link |
00:24:52.400
But it hasn't jumped to humans yet?
link |
00:24:53.400
No.
link |
00:24:54.400
Oh, interesting.
link |
00:24:55.400
Yes.
link |
00:24:56.400
That's fascinating.
link |
00:24:57.400
And then you mentioned AC2.
link |
00:25:00.400
So when you say attach, proteins are involved on both sides.
link |
00:25:06.400
Yes.
link |
00:25:07.400
So we have this infamous spike protein on the surface of the virion particle,
link |
00:25:14.400
and it does look like a spike.
link |
00:25:16.400
And I mean, that's essentially because of this protein, we call the coronavirus coronavirus.
link |
00:25:22.400
So that's what makes corona on top of the surface.
link |
00:25:27.400
So this protein, it actually it acts, so it doesn't act alone.
link |
00:25:35.400
It actually it makes three copies and it makes so called trimer.
link |
00:25:42.400
So this trimer is essentially a functional unit,
link |
00:25:45.400
a single functional unit that starts interacting with the AC2 receptor.
link |
00:25:54.400
So this is, again, another protein that now sits on the surface of a human cell or host cell, I would say.
link |
00:26:03.400
And that's essentially in that way the virus anchors itself to the host cell.
link |
00:26:14.400
Because then it needs to actually it needs to get inside.
link |
00:26:18.400
You know, it fuses its membrane with the host membrane.
link |
00:26:23.400
It releases the key components.
link |
00:26:27.400
It releases its, you know, RNA and then essentially hijacks the machinery of the cell
link |
00:26:37.400
because none of the viruses that we know of have ribosome,
link |
00:26:45.400
the machinery that allows us to print out proteins.
link |
00:26:50.400
So in order to print out proteins that are necessary for functioning of this virus,
link |
00:26:55.400
it actually needs to hijack the host ribosomes.
link |
00:26:59.400
So a virus is an RNA wrapped in a bunch of proteins,
link |
00:27:03.400
one of which is this functional mechanism of a spike protein that does the attachment.
link |
00:27:09.400
Yeah, so if you look at this virus, there are several basic components.
link |
00:27:15.400
So we start with the spike protein.
link |
00:27:18.400
This is not the only surface protein, the protein that lives on the surface of the viral particle.
link |
00:27:24.400
There is also perhaps the protein with the highest number of copies is the membrane protein.
link |
00:27:33.400
So it's essentially it forms the envelope of the protein of the viral particle
link |
00:27:43.400
and essentially, you know, helps to maintain a certain curvature, helps to make a certain curvature.
link |
00:27:54.400
Then there is another protein called envelope protein or E protein,
link |
00:28:00.400
and it actually occurs in far less quantities.
link |
00:28:05.400
And still there is ongoing research what exactly does this protein do?
link |
00:28:13.400
So these are sort of the three major surface proteins that, you know, make the viral envelope.
link |
00:28:21.400
And when we go inside, then we have another structural protein called nuclear protein.
link |
00:28:29.400
And the purpose of this protein is to protect the viral RNA.
link |
00:28:34.400
It actually binds to the viral RNA, creates a capsid.
link |
00:28:39.400
And so the rest of the virus viral information is inside of this RNA.
link |
00:28:46.400
And, you know, if you compare the amount of the genes or proteins that are made of these genes,
link |
00:28:58.400
it's significantly higher than of influenza virus, for example.
link |
00:29:04.400
Influenza virus has, I think, around eight or nine proteins where this one has at least 29.
link |
00:29:12.400
Wow. That has to do with the length of the RNA strand?
link |
00:29:17.400
So it affects the length of the RNA strand.
link |
00:29:21.400
So because you essentially need to have sort of the minimum amount of information to encode those genes.
link |
00:29:29.400
How many proteins did you say?
link |
00:29:31.400
29.
link |
00:29:32.400
29 proteins.
link |
00:29:34.400
Yes. So this is, you know, something definitely interesting because, you know, believe it or not,
link |
00:29:42.400
we've been studying, you know, coronaviruses for over two decades.
link |
00:29:47.400
We've yet to uncover all functionalities of these proteins.
link |
00:29:52.400
Could we maybe take a small attention and can you say how one would try to figure out what a function of a particular protein is?
link |
00:30:03.400
So you've mentioned people are still trying to figure out what the function of the envelope protein might be or what's the process?
link |
00:30:11.400
So this is where the research that computational scientists do might be of help because, you know,
link |
00:30:21.400
in the past several decades, we actually have collected a pretty decent amount of knowledge about different proteins in different viruses.
link |
00:30:34.400
So what we can actually try to do, and this is sort of could be sort of our first lead to a possible function is to see whether those, you know,
link |
00:30:46.400
say we have this genome of the coronavirus, of the novel coronavirus, and we identify the potential proteins.
link |
00:30:56.400
Then in order to infer the function, what we can do, we can actually see whether those proteins are similar to those ones that we already know.
link |
00:31:07.400
OK, in such a way, we can, you know, for example, clearly identify, you know, some critical components that RNA polymerase or different types of proteases.
link |
00:31:19.400
These are the proteins that essentially clip the protein sequences.
link |
00:31:27.400
And so this works in many cases. However, in some cases you have truly novel proteins.
link |
00:31:36.400
And this is a much more difficult task.
link |
00:31:40.400
Now, as a small pause, when you say similar, like what if some parts are different and some parts are similar?
link |
00:31:48.400
Like, how do you disentangle that?
link |
00:31:51.400
You know, it's a big question. Of course, you know, what bioinformatics does, it does predictions, right?
link |
00:31:59.400
So those predictions, they have to be validated by experiments.
link |
00:32:05.400
Functional or structural predictions?
link |
00:32:07.400
Both. I mean, we do structural predictions, we do functional predictions, we do interactions predictions.
link |
00:32:14.400
Oh, so this is interesting. So you just generate a lot of predictions, like reasonable predictions based on structural function, interaction, like you said.
link |
00:32:23.400
And then here you go. That's the power of bioinformatics is data grounded, good predictions of what should happen.
link |
00:32:32.400
So, you know, in a way I see it, we're helping experimental scientists to streamline the discovery process.
link |
00:32:42.400
And the experimental scientists, is that what a virologist is?
link |
00:32:47.400
So yeah, virology is one of the experimental sciences that, you know, focus on viruses.
link |
00:32:54.400
They often work with other experimental scientists, for example, the molecular imaging scientists, right?
link |
00:33:02.400
So the viruses often can be viewed and reconstructed through electron microscopy techniques.
link |
00:33:11.400
So but these are, you know, specialists that are not necessarily virologists.
link |
00:33:16.400
They work with small particles, whether it's viruses or it's an organelle of a human cell, whether it's a complex molecular machinery.
link |
00:33:33.400
So the techniques that are used are very similar in sort of in their essence.
link |
00:33:41.400
And so, yeah, so typically we see it now, the research on, you know, that is emerging and that is needed often involves the collaborations between virologists, you know, biochemists,
link |
00:34:06.400
people from pharmaceutical sciences, computational sciences.
link |
00:34:15.400
So we have to work together.
link |
00:34:18.400
So from my perspective, just to step back, sometimes I look at this stuff, just how much we understand about RNA and DNA, how much we understand about protein, like your work,
link |
00:34:29.400
the amount of proteins that you're exploring, is it surprising to you that we were able, we descendants of apes, were able to figure all of this out?
link |
00:34:41.400
Like how? So you're a computer scientist.
link |
00:34:46.400
So for me, from a computer science perspective, I know how to write a Python program, things are clear.
link |
00:34:51.400
But biology is a giant mess, it feels like to me from an outsider's perspective.
link |
00:34:58.400
How surprising is it, amazing is it that we were able to figure this stuff out?
link |
00:35:04.400
You know, if you look at the, you know, how computational science and computer science was evolving, right?
link |
00:35:12.400
I think it was just a matter of time that we would approach biology.
link |
00:35:16.400
So we started from, you know, applications to much more fundamental systems, physics, you know, and now we are, or, you know, small chemical compounds.
link |
00:35:32.400
So now we are approaching the more complex biological systems, and I think it's a natural evolution of, you know, of the computer science, of mathematics.
link |
00:35:48.400
So sure, that's the computer science side, I just meant even in higher level.
link |
00:35:52.400
So that to me is surprising that computer science can offer help in this messy world.
link |
00:35:57.400
But I just mean, it's incredible that the biologists and the chemists can figure all this out.
link |
00:36:02.400
Or does that just sound ridiculous to you, that of course they would.
link |
00:36:07.400
It just seems like a very complicated set of problems, like the variety of the kinds of things that could be produced in the body.
link |
00:36:15.400
Just like you said, 29 protein, I mean, just getting a hang of it so quickly, it just seems impossible to me.
link |
00:36:26.400
I agree. I mean, it's, and I have to say we are, you know, in the very, very beginning of this journey.
link |
00:36:34.400
I mean, we've yet to, I mean, we've yet to comprehend, not even try to understand and figure out all the details, but we've yet to comprehend the complexity of the cell.
link |
00:36:51.400
We know that neuroscience is not even at the beginning of understanding the human mind.
link |
00:36:59.400
So where's biology sit in terms of understanding the function, deeply understanding the function of viruses and cells?
link |
00:37:09.400
So there, sometimes it's easy to say when you talk about function, what you really refer to is perhaps not a deep understanding, but more of a understanding sufficient to be able to mess with it using a antivirus, like mess with it chemically to prevent some of its function.
link |
00:37:29.400
Or do you understand the function?
link |
00:37:31.400
Well, I think, I think we are much farther in terms of understanding of the complex genetic disorder, such as cancer, where you have layers of complexity.
link |
00:37:42.400
And we, you know, as in my laboratory, we're trying to contribute to that research, but we're also, you know, we're overwhelmed with how many different layers of complexity, different layers of mechanisms that can be hijacked by cancer simultaneously.
link |
00:38:00.400
And so, you know, I think biology in the past 20 years, again, from the perspective of the outsider, because I'm not a biologist, but I think it has advanced tremendously.
link |
00:38:18.400
And one thing that where computational scientists and data scientists are now becoming very, very helpful is in the fact, it's coming from the fact that we are now able to generate a lot of information about the cell.
link |
00:38:43.400
Whether it's next generation sequencing or transcriptomics, whether it's life imaging information, where it is, you know, complex interactions between proteins or between proteins and small molecules such as drugs.
link |
00:39:01.400
We are becoming very efficient in generating this information. And now the next step is to become equally efficient in processing this information and extracting the key knowledge from that.
link |
00:39:20.400
That could then be validated with experiment.
link |
00:39:23.400
Yes.
link |
00:39:24.400
So maybe then going all the way back, we were talking, you said the first step is seeing if we can match the new proteins you found in the virus against something we've seen before to figure out its function.
link |
00:39:37.400
And then you also mentioned that, but there could be cases where it's a totally new protein. Is there something bioinformatics can offer when it's a totally new protein?
link |
00:39:47.400
This is where many of the methods and you probably are aware of, you know, the case of machine learning, many of these methods rely on the previous knowledge.
link |
00:39:59.400
Right.
link |
00:40:00.400
Right. So things that where we try to do from scratch are incredibly difficult.
link |
00:40:07.400
You know, something that we call ab initio. And this is, I mean, it's not just the function. I mean, you know, we've yet to have a robust method to predict the structures of these proteins in ab initio, you know, by not using any templates of other related proteins.
link |
00:40:31.400
So protein is a chain of amino acids.
link |
00:40:35.400
It's residues.
link |
00:40:36.400
Residues. Yeah. And then somehow magically, maybe you can tell me, they seem to fold in incredibly weird and complicated 3D shapes.
link |
00:40:48.400
Yes.
link |
00:40:49.400
So, and that's where actually the idea of protein folding or just not the idea, but the problem of figuring out how the concept, how they fold into those weird shapes comes in.
link |
00:41:04.400
So that's another side of computational work. So can you describe what protein folding from the computational side is and maybe your thoughts on the folding at home efforts that a lot of people know that you can use your machine to do protein folding?
link |
00:41:22.400
So yeah, protein folding is, you know, one of those $1 million price challenges, right?
link |
00:41:30.400
So the reason for that is we've yet to understand precisely how the protein gets folded so efficiently to the point that in many cases where you, you know, where you try to unfold it due to the high temperature, it actually folds back into its original state.
link |
00:41:53.400
So we know a lot about the mechanisms, right? But putting those mechanisms together and making sense, it's a computationally very expensive task.
link |
00:42:10.400
In general, do proteins fold, can they fold in arbitrary large number of ways or do they usually fold in a very small number of ways?
link |
00:42:19.400
It's typically, I mean, we tend to think that, you know, there is a one sort of canonical fold for a protein, although there are many cases where the proteins, you know, upon destabilization, it can be folded into a different confirmation.
link |
00:42:36.400
And this is especially true when you look at sort of proteins that include more than one structural unit. So those structural units, we call them protein domains.
link |
00:42:48.400
Essentially, protein domain is a single unit that typically is evolutionary preserved, that typically carries out a single function and typically has a very distinct fold, right?
link |
00:43:04.400
The structure, 3D structure organization. But turns out that if you look at human, an average protein in a human cell would have a bit of two or three such subunits and how they are trying to fold into the sort of, you know, next level fold, right?
link |
00:43:30.400
So within subunit there's folding and then they fold into the larger 3D structure, right?
link |
00:43:38.400
And all of that, there's some understanding of the basic mechanisms, but not to put together to be able to fold it.
link |
00:43:44.400
We're still, I mean, we're still struggling. I mean, we're getting pretty good about folding relatively small proteins up to 100 residues. I mean, but we're still far away from folding, you know, larger proteins.
link |
00:44:02.400
And some of them are notoriously difficult. For example, transmembrane proteins, proteins that sit in the membranes of the cell, they're incredibly important, but they are incredibly difficult to solve.
link |
00:44:19.400
And so basically there's a lot of degrees of freedom, how it folds. And so it's a combinatorial problem where it just explodes. There's so many dimensions.
link |
00:44:28.400
Well, it is a combinatorial problem, but it doesn't mean that we cannot approach it from the, not from the brute force approach. And so the machine learning approaches, you know, have been emerged that try to tackle it.
link |
00:44:47.400
So folding at home, I don't know how familiar you are with it, but is that using machine learning or is it more brute force?
link |
00:44:55.400
So folding at home, it was originally, and I remember I was, I mean, it was a long time ago. I was a postdoc and we learned about this, you know, this game because it was originally designed as the game.
link |
00:45:10.400
And we, you know, I took a look at it and it's interesting because it's really, you know, it's very transparent, very intuitive. So, and from what I heard, I've yet to introduce it to my son, but you know, kids are actually getting very good at folding the proteins.
link |
00:45:32.400
And it was, you know, it came to me as the, not as a surprise, but actually as the sort of manifest of, you know, our capacity to do this kind of, to solve this kind of problems.
link |
00:45:52.400
When a paper was published in one of these top journals with the coauthors being the actual players of this game.
link |
00:46:07.400
So, and what happened was that they managed to get better structures than the scientists themselves.
link |
00:46:18.400
So that, you know, that was very, I mean, it was kind of profound, you know, revelation that problems that are so challenging for a computational science, maybe not that challenging for a human brain.
link |
00:46:38.400
That's a really good, that's a hopeful message always when there's a, the proof of existence, the existence proof that it's possible. That's really interesting, but it seems, what are the best ways to do protein folding now?
link |
00:46:58.400
So if you look at what DeepMind does with AlphaFold, so they kind of, that's a learning approach. What's your sense? I mean, your background is in machine learning, but is this a learnable problem? Is this still a brute force?
link |
00:47:14.400
Are we in the Gary Kasparov deep blue days or are we in the AlphaGo playing the game of Go days of folding?
link |
00:47:24.400
Well, I think we are, we are advancing towards this direction. I mean, if you look, so there is a sort of Olympic game for protein folders called CASP, and it's essentially, it's, you know, it's a competition where different teams are given exactly the same
link |
00:47:45.400
protein sequences and they try to predict their structures, right? And of course there are different sort of sub tasks, but in the recent competition, AlphaFold was among the top performing teams, if not the top performing team.
link |
00:48:04.400
So there is definitely a benefit from the data that have been generated, you know, in the past several decades, the structural data. And certainly, you know, we are now at the capacity to summarize this data, to generalize this data and to use those principles, you know, in order to predict protein structures.
link |
00:48:33.400
That's one of the really cool things here is there's, maybe you can comment on it. There seems to be these open data sets of protein. How did that?
link |
00:48:43.400
Protein Data Bank?
link |
00:48:45.400
Yeah, Protein Data Bank. I mean, that's crazy. Is this a recent thing for just the coronavirus?
link |
00:48:52.400
It's been for many, many years. I believe the first Protein Data Bank was designed on flashcards. So, yes, this is a great example of the community efforts of everyone contributing because every time you solve a protein or a protein complex,
link |
00:49:21.400
this is where you submit it. And, you know, the scientists get access to it, scientists get to test it. And we, bioinformaticians, use this information to, you know, to make predictions.
link |
00:49:41.400
So, there's no culture of like hoarding discoveries here. So, I mean, you've released a few or a bunch of proteins that were matching, whatever. We'll talk about details a little bit, but it's kind of amazing how open the culture here is.
link |
00:50:06.400
It is. And I think this pandemic actually demonstrated the ability of scientific community to, you know, to solve this challenge collaboratively. And this is, I think, if anything, it actually moved us to a brand new level of collaborations of the efficiency
link |
00:50:34.400
in which people establish new collaborations, in which people offer their help to each other, scientists offer their help to each other.
link |
00:50:44.400
And publish results too. It's very interesting. We're now trying to figure out, there's a few journals that are trying to sort of do the very accelerated review cycle, but so many preprints. So, just posting a paper going out, I think it's fundamentally changing the way we think about papers.
link |
00:51:03.400
Yes. I mean, the way we think about knowledge, I would say, yes. Because, yes, I completely agree. I think now the knowledge is becoming sort of the core value, not the paper or the journal where this knowledge is published.
link |
00:51:26.400
And I think this is, again, we are living in the times where it becomes really crystallized, the idea that the most important value is in the knowledge.
link |
00:51:43.400
So, maybe you can comment, like, what do you think the future of that knowledge sharing looks like? So, you have this paper that I hope we get a chance to talk about a little bit, but it has, like, a really nice abstract and introduction related, like, it has all the usual, I mean, probably took a long time to put together.
link |
00:52:00.400
So, but is that going to remain, like, you could have communicated a lot of fundamental ideas here in much shorter amount that's less traditionally acceptable by the journal context.
link |
00:52:15.400
So, well, you know, so the first version that we posted, not even on the bioarchive, because bioarchive back then, it was essentially, you know, overwhelmed with the number of submissions.
link |
00:52:33.400
So, our submission, I think it took five or six days to just for it to be screened and put online. So, we, you know, essentially we put the first preprint on our website, and, you know, it started getting access right away.
link |
00:52:55.400
So, and, you know, so this original preprint was in a much rougher shape than this paper.
link |
00:53:05.400
And, but we tried, I mean, we honestly tried to be as compact as possible with, you know, introducing the information that is necessary to explain our, you know, our results.
link |
00:53:26.400
So, maybe you can dive right in if it's okay. Sure. So, this is a paper called Structural Genomics of SARS Co, how do you even pronounce? SARS CoV2. CoV2? Yeah.
link |
00:53:38.400
By the way, CoVid is such a terrible name, but it stuck. Anyway, SARS CoV2 indicates evolutionary conserved functional regions of viral proteins.
link |
00:53:49.400
So, this is looking at all kinds of proteins that are part of this novel coronavirus and how they match up against the previous other kinds of coronaviruses.
link |
00:54:01.400
I mean, there's a lot of beautiful figures. I was wondering if you could, I mean, there's so many questions I could ask here, but maybe at the, how do you get started doing this paper?
link |
00:54:11.400
So, how do you start to figure out the 3D structure of a novel virus?
link |
00:54:15.400
Yes. So, there is actually a little story behind it. And so, the story actually dated back in September of 2019.
link |
00:54:27.400
And you probably remember that back then, we had another dangerous virus, triple E virus. It's a queen encephalitis virus.
link |
00:54:39.400
Can you maybe linger on it? I have to admit, I was sadly completely unaware.
link |
00:54:45.400
So, that was actually a virus outbreak that happened in New England only. The danger in this virus was that it actually targeted your brain.
link |
00:54:57.400
So, the word death from this virus, it was transferred, the main vector was mosquitoes.
link |
00:55:11.400
And obviously, fall time is the time where you have a lot of them in New England.
link |
00:55:18.400
And on one hand, people realize this is actually a very dangerous thing. So, it had an impact on the local economy.
link |
00:55:31.400
The schools were closed past six o clock, no activities outside for the kids because the kids were suffering quite tremendously when infected from this virus.
link |
00:55:47.400
How do I not know about this? Was universities impacted?
link |
00:55:51.400
It was in the news. I mean, it was not impacted to a high degree in Boston necessarily, but in the Metro West area and actually spread around, I think, all the way to New Hampshire, Connecticut.
link |
00:56:08.400
And you mentioned affecting the brain. That's one other comment we should make.
link |
00:56:13.400
So, you mentioned AC2 for the coronavirus. So, these viruses kind of attached to something in the body.
link |
00:56:23.400
So, it essentially attaches to these proteins in those cells in the body where those proteins are expressed, where they actually have them in abundance.
link |
00:56:35.400
So, sometimes that could be in the lungs, that could be in the brain, that could be in something.
link |
00:56:39.400
So, I think right now, from what I read, they have the epithelial cells inside.
link |
00:56:49.400
What does that mean?
link |
00:56:50.400
So, the cells that are covering the surface, so inside the nasal surfaces, the throat, the lung cells, and I believe liver as a couple of other organs where they are actually expressed in abundance.
link |
00:57:13.400
That's for the AC2 you said?
link |
00:57:14.400
For the AC2 receptors.
link |
00:57:16.400
So, okay. So, back to the story, the outbreak in the fall.
link |
00:57:20.400
So, now the impact of this virus is significant.
link |
00:57:29.400
However, it's a prelocal problem to the point that this is something that we would call a neglected disease
link |
00:57:38.400
because it's not big enough to make the drug design companies to design a new antiviral or a new vaccine.
link |
00:57:52.400
It's not big enough to generate a lot of grants from the national funding agencies.
link |
00:58:03.400
So, it doesn't mean we cannot do anything about it.
link |
00:58:08.400
And so, what I did is I taught a bioinformatics class in Worcester Polytechnic Institute, and we are very much a problem learning institution.
link |
00:58:25.400
So, I thought that that would be a perfect project for the class.
link |
00:58:31.400
It's an ongoing case study.
link |
00:58:34.400
So, we essentially designed a study where we tried to use bioinformatics to understand as much as possible about this virus.
link |
00:58:47.400
And a very substantial portion of the study was to understand the structures of the proteins,
link |
00:58:55.400
to understand how they interact with each other and with the host proteins, try to understand the evolution of this virus.
link |
00:59:08.400
So, obviously, a very important question, where it will evolve further, how it happened here.
link |
00:59:21.400
So, we did all these projects, and now I'm trying to put them into a paper where all these undergraduate students will be coauthors.
link |
00:59:32.400
But essentially, the projects were finished right about mid December.
link |
00:59:39.400
And a couple of weeks later, I heard about this mysterious new virus that was discovered and was reported in Wuhan province.
link |
00:59:50.400
And immediately I thought that, well, we just did that, can't we do the same thing with this virus?
link |
01:00:00.400
And so, we started waiting for the genome to be released, because that's essentially the first piece of information that is critical.
link |
01:00:08.400
Once you have the genome sequence, you can start doing a lot using bioinformatics.
link |
01:00:13.400
When you say genome sequence, that's referring to the sequence of letters that make up the RNA?
link |
01:00:20.400
Well, the sequence that make up the entire information encoded in the protein, right?
link |
01:00:28.400
So, that includes all 29 genes.
link |
01:00:34.400
What are genes? What's the encoding of information?
link |
01:00:38.400
So, genes is essentially a basic functional unit that we can consider.
link |
01:00:46.400
So, each gene in the virus would correspond to a protein.
link |
01:00:53.400
So, gene by itself doesn't do its function.
link |
01:00:56.400
It needs to be converted or translated into the protein that will become the actual functional unit.
link |
01:01:06.400
Yeah, like you said, the printer.
link |
01:01:09.400
So, we need the printer for that.
link |
01:01:11.400
We need the printer, okay.
link |
01:01:12.400
So, the first step is to figure out the genome, the sequence of things that could be then used for printing the protein.
link |
01:01:21.400
So, okay.
link |
01:01:22.400
So, then the next step, so once we have this and so we use the existing information about SARS because the SARS genomics has been done in abundance.
link |
01:01:37.400
So, we have different strains of SARS and actually other related coronaviruses, MERS, the bat coronavirus.
link |
01:01:48.400
And we started by identifying the potential genes because right now it's just a sequence, right?
link |
01:01:56.400
So, it's a sequence that is roughly, it's less than 30,000 nucleotide long.
link |
01:02:04.400
Just a raw sequence.
link |
01:02:06.400
It's a raw sequence.
link |
01:02:07.400
No other information really.
link |
01:02:08.400
And we now need to define the boundaries of the genes that would then be used to identify the protein and protein structures.
link |
01:02:22.400
How hard is that problem?
link |
01:02:23.400
It's not, I mean, it's pretty straightforward.
link |
01:02:27.400
So, you know, so because we use the existing information about SARS proteins and SARS genes.
link |
01:02:35.400
So, once again, you kind of, we are relying on the, yes.
link |
01:02:40.400
So, and then once we get there, this is where sort of the first more traditional bioinformatics step begins.
link |
01:02:54.400
We're trying to use this protein sequences and get the 3D information about those proteins.
link |
01:03:03.400
So, this is where we are relying heavily on the structure information specifically from the protein databank that we're talking about.
link |
01:03:15.400
And here you're looking for similar proteins.
link |
01:03:18.400
Yes.
link |
01:03:19.400
So, the concept that we are operating when we do this kind of modeling, it's called homology or template based modeling.
link |
01:03:27.400
So, essentially using the concept that if you have two sequences that are similar in terms of the letters, the structures of the sequences are expected to be similar as well.
link |
01:03:43.400
And this is at the micro, at the very local scale?
link |
01:03:48.400
At the scale of the whole protein.
link |
01:03:50.400
At the whole protein.
link |
01:03:51.400
So, actually, so, you know, so, of course the devil is in the details.
link |
01:03:57.400
And this is why we need actually pre sophisticated modeling tools to do so.
link |
01:04:11.400
Once we get the structures of the individual proteins, we try to see whether or not these proteins act alone or they have to be forming protein complexes in order to perform this function.
link |
01:04:31.400
And again, so, this is sort of the next level of the modeling because now you need to understand how proteins interact and it could be the case that the protein interacts with itself and makes sort of a multimeric complex.
link |
01:04:51.400
The same protein just repeated multiple times and we have quite a few such proteins in SARS CoV2, specifically spike protein needs three copies to function, envelope protein needs five copies to function.
link |
01:05:14.400
And there are some other multimeric complexes.
link |
01:05:18.400
That's what you mean by attracted with itself and you see multiple copies. So, how do you, how do you make a good guess whether something's going to interact?
link |
01:05:27.400
Well, again, so there are two approaches, right? So one is look at the previously solved complexes. Now we're looking not at the individual structures but the structures of the whole complex.
link |
01:05:41.400
Complex is a bunch of multiple proteins.
link |
01:05:43.400
Yeah, so it's a bunch of proteins essentially glued together.
link |
01:05:47.400
And when you say glued, that's the interaction.
link |
01:05:49.400
That's the interaction. So there are different forces, different sort of physical forces behind this.
link |
01:05:57.400
Sorry to keep asking dumb questions, but is the interaction fundamentally structural or is it functional? Like in the way you're thinking about it?
link |
01:06:10.400
That's actually a very good way to ask this question because it turns out that the interaction is structural, but in the way it forms the structure, it actually also carries out the function.
link |
01:06:27.400
So interaction is often needed to carry out very specific function of a protein.
link |
01:06:35.400
But in terms of on the other side, figuring out you're really starting at the structure before you figure out the function.
link |
01:06:43.400
So there's a beautiful figure too in the paper of all the different proteins that make up, able to figure out that make up the novel coronavirus.
link |
01:06:58.400
What are we looking at? So these are like, that's through the step two that you mentioned, when you try to guess at the possible proteins, that's what you're going to get is these blue cyan blobs.
link |
01:07:16.400
Yes. So those are the individual proteins for which we have at least some information from the previous studies.
link |
01:07:28.400
So there is advantage and disadvantage of using previous studies. The biggest, well, the disadvantage is that we may not necessarily have the coverage of all 29 proteins.
link |
01:07:40.400
However, the biggest advantage is that the accuracy in which we can model these proteins is very high, much higher compared to ab initio methods that do not use any template information.
link |
01:07:56.400
So, but nevertheless, this figure also has, it's such a beautiful and I love these pictures so much. It has like the pink parts, which are the parts that are different.
link |
01:08:10.400
So you're highlighting, so the difference you find is on the 2D sequence. And then you try to infer what that will look like on the 3D.
link |
01:08:18.400
Yeah. So the difference actually is on the 1D sequence.
link |
01:08:23.400
1D, sorry, 2D, right.
link |
01:08:26.400
So this is one of these first questions that we try to answer is that, well, if you take this new virus and you take the closest relatives, which are SARS and a couple of bad coronavirus strains, they are already the closest relatives that we are aware of.
link |
01:08:51.400
Now, what are the difference between this virus and its close relatives, right? And if you look, typically when you take a sequence, those differences could be quite far away from each other.
link |
01:09:07.400
So what make, what 3D structure makes those difference to do, very often they tend to cluster together.
link |
01:09:18.400
Interesting.
link |
01:09:19.400
And then all of a sudden the differences that may look completely unrelated actually relate to each other. And sometimes they are there because they correspond, they attack the functional site, right?
link |
01:09:36.400
So they are there because this is the functional site that is highly mutated.
link |
01:09:43.400
So that's a computational approach to figuring something out. And when it comes together like that, that's kind of a nice clean indication that there's something, this could be actually indicative of what's happening.
link |
01:09:58.400
Yes. I mean, so we need this information and the 3D structure gives us just a very intuitive way to look at this information and then start to ask, start asking questions such as, so this place of this protein that is highly mutated,
link |
01:10:23.400
does it, is it the functional part of the protein? So does this part of the protein interact with some other proteins or maybe with some other ligands, small molecules, right?
link |
01:10:42.400
So we will try now to functionally inform this 3D structure.
link |
01:10:50.400
So you have a bunch of these mutated parts, if like, I don't know, how many are there in the novel coronavirus when you compare to SARS? We're talking about hundreds, thousands, like these pink regions.
link |
01:11:08.400
No, no, much less than that. And it's very interesting that if you look at that, you know, so the first thing that you start seeing, right, you know, you look at patterns, right? And the first pattern that becomes obvious is that some of the proteins in the new coronavirus are pretty much intact.
link |
01:11:31.400
So they're pretty much exactly the same as SARS, as the bat coronavirus, whereas some others are heavily mutated.
link |
01:11:42.400
So it looks like that the, you know, the evolution is not occurring, you know, uniformly across the entire, you know, viral genome, but actually target very specific proteins.
link |
01:12:01.400
And what do you do with that? Like from the Sherlock Holmes perspective?
link |
01:12:05.400
Well, you know, so one of the most interesting findings we had was the fact that the viral, so the binding sites on the viral surfaces that get targeted by the known small molecules, they were pretty much not affected at all.
link |
01:12:34.400
And so that means that the same small drugs or small drug like compounds can be efficient for the new coronavirus.
link |
01:12:50.400
Ah, so this all actually maps to the drug compounds too. So you're actually mapping out what old stuff is going to work on this thing and then possibilities for new stuff to work by mapping out the things that have mutated.
link |
01:13:07.400
Yes. So we essentially know which parts behave differently and which parts are likely to behave similar. And again, you know, of course, all our predictions need to be validated by experiments.
link |
01:13:25.400
But hopefully that sort of helps us to delineate the regions of this virus that, you know, can be promising in terms of the drug discovery.
link |
01:13:38.400
You kind of, you kind of mentioned this already, but maybe you can elaborate. So how different from the structural and functional perspective does the new coronavirus appear to be relative to SARS?
link |
01:13:51.400
We now are trying to understand the overall structural characteristics of this virus because I mean, that's our next step, trying to model the viral particle of single viral particle of this virus.
link |
01:14:08.400
So that means you have the individual proteins, like you said, you have to figure out what their interaction is. So you have this, is that where this graph kind of interactome?
link |
01:14:19.400
So the interactome is essentially our prediction on the potential interactions, some of them that we already deciphered from the structural knowledge, but some of them that are essentially are deciphered from the knowledge of the existing interactions that people previously obtained for SARS, for MERS or other related viruses.
link |
01:14:48.400
Is there kind of interactomes, am I pronouncing that correctly by the way? Are those already converged towards for SARS?
link |
01:15:01.400
So I think there are a couple of papers that now investigate the sort of the large scale set of interactions between the new SARS and its host. And so I think that's an ongoing study.
link |
01:15:25.400
And the success of that, the result would be an interactome. Yes. And so when you say not trying to figure out the entire, the particle, the entire thing.
link |
01:15:37.400
So if you look, you know, so structure, right? So what this viral particle looks like, right? So as I said, it's, you know, the surface of it is an envelope, which is essentially a so called lipid bilayer with proteins integrated into the surface.
link |
01:15:58.400
So how, so an average particle is around 80 nanometers, right? So this particle can have about 50 to 100 spike proteins.
link |
01:16:20.400
So at least we suspect it and, you know, based on the micrographs images, it's very comparable to MHV virus in mice and SARS virus.
link |
01:16:32.400
Micrographs are actual pictures of the actual virus. Okay. So these are models. This is the actual images, right?
link |
01:16:40.400
What do they, sorry for the tangents, but what are these things? So when you look on the internet, the models and the pictures are kind of, and the models you have here are just gorgeous and beautiful.
link |
01:16:51.400
When you actually take pictures of them with a micrograph, like what, what do we look?
link |
01:16:55.400
Well, they typically are not perfect. Right? So, so the, most of the images that you see now is the, is the sphere with those spikes.
link |
01:17:08.400
You actually see the spikes? Yes, you do see the spikes. And now, you know, the, our collaborators for Texas A&M University, Benjamin Newman, he actually in the recent paper about SARS he proposed, and there's some actually evidence behind it,
link |
01:17:31.400
that the particle is not a sphere, but is actually as elongated ellipsoid like particles. So, so that's what we are trying to incorporate into our model.
link |
01:17:49.400
And the, I mean, you know, if you look at the actual micrographs, you see that those particles are, you know, are not symmetric.
link |
01:18:02.400
So the, the, the, some of them, and of course, you know, it could be due to the treatment of the, of the material. It could be due to the, some noise in the imaging.
link |
01:18:14.400
Right. So there's a lot of uncertainty in all this. So it's okay. So structurally figuring out the entire part. By the way, again, sorry for the tangents, but why the term particle? Or is it just something that's stuck?
link |
01:18:27.400
It's a, it's a, it's a single, you know, so we call, you know, we call it the virion. So virion particle, it's essentially a single virus.
link |
01:18:35.400
But it just feels like, because particle to me, from the physics perspective, feels like this, the most basic unit, because there seems to be so much going on inside the virus.
link |
01:18:48.400
Yeah.
link |
01:18:49.400
It doesn't feel like a particle to me.
link |
01:18:50.400
Yeah, well, yeah, it's probably, I think it's the, you know, virion is a good way to call it.
link |
01:18:57.400
So, okay, so trying to figure out, trying to figure out the entirety of the system.
link |
01:19:05.400
Yes. So, you know, so, you know, so this is, so the virion has 5200 spikes, trimer spikes. It has roughly 200 to 400 membrane protein dimers. And those are arranged in the very nice lattice.
link |
01:19:28.400
So you can actually see sort of the, it's like a carpet of...
link |
01:19:35.400
On the surface again.
link |
01:19:36.400
Exactly, on the surface. And occasionally you also see this envelope protein inside.
link |
01:19:43.400
Is that the one we don't know what it does?
link |
01:19:46.400
Exactly. Exactly. The one that forms the pentamer, this very nice pentameric ring. And so, you know, so this is what we're trying to, you know, we're trying to put now all our knowledge together and see whether we can actually generate this overall virion model.
link |
01:20:08.400
With an idea to understand, you know, well, first of all, to understand how it looks like, how far it is from those images that were generated. But I mean, the implications are, you know, there is a potential for the, you know, nanoparticle design that will mimic this virion particle.
link |
01:20:35.400
Is the process of nanoparticle design meaning artificially designing something that looks similar?
link |
01:20:41.400
Yes. And also the one that can potentially compete with the actual virion particles and therefore reduce the effect of the infection.
link |
01:20:54.400
So is this the idea of, like, what is a vaccine?
link |
01:20:58.400
So vaccine, so there are two ways of essentially treating and in the case of vaccine is preventing the infection.
link |
01:21:09.400
So vaccine is, you know, a way to train our immune system.
link |
01:21:18.400
So our immune system becomes aware of this new danger and therefore is capable of generating the antibodies then will essentially bind to the spike proteins because that's the main target for the, you know, for the vaccine's design and block its functioning.
link |
01:21:47.400
If you have the spike with the antibody on top, it can no longer interact with AC2 receptor.
link |
01:21:56.400
So the process of designing a vaccine then is you have to understand enough about the structure of the virus itself to be able to create an artificial, an artificial particle?
link |
01:22:09.400
Well, I mean, so the nanoparticle is a very exciting and new research. So there are already established ways to, you know, to make vaccines and there are several different ones, right?
link |
01:22:25.400
So there is one where essentially the virus gets through the cell culture multiple times, so it becomes essentially adjusted to the specific embryonic cell and as a result becomes less, you know, compatible with the host human cells.
link |
01:22:52.400
So therefore it's sort of the idea of the live vaccine where the particles are there, but they are not so efficient, you know, so they cannot replicate, you know, as rapidly as, you know, before the vaccine.
link |
01:23:12.400
They can be introduced to the immune system, the immune system will learn and the person who gets this vaccine won't get, you know, sick or, you know, will have mild, you know, mild symptoms.
link |
01:23:29.400
So then there is sort of different types of the way to introduce the nonfunctional parts of this virus or the virus where some of the information is stripped down.
link |
01:23:45.400
For example, the virus with no genetic material, so with no RNA genome, exactly. So it cannot replicate, it cannot essentially perform most of its functions.
link |
01:23:59.400
What is the biggest hurdle to design one of these, to arrive at one of these? Is it the work that you're doing in the fundamental understanding of this new virus or is it in the, from our perspective, well, complicated world of experimental validation and sort of showing that this, like going through the whole process of showing this is actually going to work with FDA approval, all that kind of stuff?
link |
01:24:24.400
I think it's both. I mean, you know, our understanding of the molecular mechanisms will allow us to, you know, to design, to have more efficient designs of the vaccines. However, once you design a vaccine, it needs to be tested.
link |
01:24:42.400
But when you look at the 18 months and the different projections, it seems like an exceptionally, historically speaking, maybe you can correct me, but it's even 18 months seems like a very accelerated timeline.
link |
01:24:54.400
It is. It is. I mean, I remember reading about, you know, in the book about some previous vaccines that it could take up to 10 years to design and, you know, properly test a vaccine before its mass production.
link |
01:25:14.400
So yeah, we, you know, everything is accelerated these days. I mean, for better, for worse, but, but, you know, we definitely need that.
link |
01:25:23.400
Well, especially with the coronavirus, I mean, the scientific community is really stepping up and working together. The collaborative aspect is really interesting. You mentioned, so the vaccine is one and then there's antivirals, antiviral drugs.
link |
01:25:35.400
So antiviral drugs. So where, you know, vaccines are typically needed to prevent the infection. Right. But once you have an infection, one, you know, so what we try to do, we try to stop it.
link |
01:25:47.400
So we try to stop virus from functioning. And so the antiviral drugs are designed to block some critical functioning of the proteins from the virus.
link |
01:26:06.400
So there are a number of interesting candidates. And I think, you know, if you ask me, I, you know, I think Remdesivir is perhaps the most promising.
link |
01:26:25.400
It's, it has been shown to be, you know, an efficient and effective antiviral for SARS.
link |
01:26:38.400
Originally, it was the antiviral drug developed for a completely different virus, I think, for Ebola and Marburg.
link |
01:26:49.400
At high levels, you know how it works?
link |
01:26:51.400
So it tries to mimic one of the nucleotides in RNA and essentially that stops the replication.
link |
01:27:04.400
So messes, I guess that's what, so antiviral drugs mess with some aspect of this process.
link |
01:27:11.400
So, you know, so essentially we try to stop certain functions of the virus. There are some other ones, you know, that are designed to inhibit the protease, the thing that clips protein sequences.
link |
01:27:31.400
There is one that was originally designed for malaria, which is a bacterial, you know, bacterial disease.
link |
01:27:42.400
This is so cool. So, but that's exactly where your work steps in is you're figuring out the functional and the structure of these different, so like providing candidates for where drugs can plug in.
link |
01:27:54.400
Well, yes, because, you know, one thing that we don't know is whether or not, so let's say we have a perfect drug candidate that is efficient against SARS and against MERS.
link |
01:28:08.400
Now, is it going to be efficient against new SARS COVID 2?
link |
01:28:14.400
We don't know that. And there are multiple aspects that can affect this efficiency.
link |
01:28:22.400
So, for instance, if the binding site, so the part of the protein where this ligand gets attached, if this site is mutated, then the ligand may not be attachable to this part any longer.
link |
01:28:40.400
And, you know, our work and the work of other bioinformatics groups, you know, essentially are trying to understand whether or not that will be the case.
link |
01:28:54.400
And it looks like for the ligands that we looked at, the ligand binding sites are pretty much intact, which is very promising.
link |
01:29:07.400
So, if we can just like zoom out for a second. Are you optimistic?
link |
01:29:15.400
So, there's two, well, there's three possible ends to the coronavirus pandemic.
link |
01:29:21.400
So, one is drugs or vaccines get figured out very quickly, probably drugs first.
link |
01:29:30.400
The other is the pandemic runs its course for this wave, at least.
link |
01:29:37.400
And then the third is, you know, things go much worse in some dark, bad, very bad direction.
link |
01:29:46.400
Do you see, let's focus on the first two.
link |
01:29:50.400
Do you see the anti drugs or the work you're doing being relevant for us right now in stopping the pandemic?
link |
01:30:03.400
Or do you hope that the pandemic will run its course?
link |
01:30:06.400
So, the social distancing, things like wearing masks, all those discussions that we're having will be the method with which we fight coronavirus in the short term.
link |
01:30:20.400
Or do you think that it will have to be antiviral drugs?
link |
01:30:25.400
I think antivirals would be, I would view that as at least the short term solution.
link |
01:30:36.400
I see more and more cases in the news of those new drug candidates being administered in hospitals.
link |
01:30:48.400
And I mean, this is right now the best what we have.
link |
01:30:55.400
But do we need it in order to reopen the economy?
link |
01:30:58.400
I mean, we definitely need it.
link |
01:31:01.400
I cannot sort of speculate on how that will affect reopening of the economy because we are, you know, we are kind of deep into the pandemic.
link |
01:31:16.400
And it's not just the states. It's also, you know, worldwide, you know.
link |
01:31:23.400
Of course, you know, there is also the possibility of the second wave, as we, you know, as you mentioned.
link |
01:31:34.400
And this is why, you know, we need to be super careful.
link |
01:31:41.400
We need to follow all the precautions that the doctors tell us to do.
link |
01:31:50.400
Are you worried about the mutation of the virus?
link |
01:31:54.400
It's, of course, a real possibility.
link |
01:31:58.400
Now, how, to what extent this virus can mutate, it's an open question.
link |
01:32:06.400
I mean, we know that it is able to mutate, to jump from one species to another and to become transmittable between humans.
link |
01:32:19.400
Right. So will it, you know, so let's imagine that we have the new antiviral.
link |
01:32:26.400
Will this virus become eventually resistant to this antiviral?
link |
01:32:33.400
We don't know. I mean, this is what needs to be studied.
link |
01:32:36.400
This is such a beautiful and terrifying process that a virus, some viruses may be able to mutate to respond to the, to mutate around the thing we've put before it.
link |
01:32:51.400
Can you explain that process? Like, how does that happen? Is that just the way of evolution?
link |
01:32:57.400
I would say so, yes. I mean, it's the evolutionary mechanisms.
link |
01:33:02.400
There is nothing imprinted into this virus that makes it, you know, it just the way it evolves.
link |
01:33:12.400
And actually, it's the way it core evolves with its host.
link |
01:33:18.400
It's just amazing, especially the evolution mechanisms, especially amazing given how simple the virus is.
link |
01:33:27.400
It's incredible that it's, I mean, it's beautiful.
link |
01:33:32.400
It's beautiful because it's one of the cleanest examples of evolution working.
link |
01:33:38.400
Well, I think, I mean, one of the sort of the reasons for its simplicity is because it does not require all the necessary functions to be stored.
link |
01:33:53.400
So it actually can hijack the majority of the necessary functions from the host cell.
link |
01:34:00.400
So the ability to do so, in my view, reduces the complexity of this machine drastically.
link |
01:34:11.400
Although if you look at the, you know, most recent discoveries.
link |
01:34:15.400
So the scientists discovered viruses that are as large as bacteria.
link |
01:34:21.400
Right. So this MIMI viruses and MAMA viruses.
link |
01:34:26.400
It actually, those discoveries made scientists to reconsider the origins of the virus.
link |
01:34:36.400
You know, and what are the mechanisms and how, you know, what are the mechanisms, the evolution mechanisms that leads to the appearance of the viruses.
link |
01:34:46.400
By the way, I mean, you did mention that viruses are, I think you mentioned that they're not living.
link |
01:34:52.400
Yes, they're not living organisms.
link |
01:34:54.400
So let me ask that question again.
link |
01:34:57.400
Why do you think they're not living organisms?
link |
01:35:00.400
Well, because they are dependent.
link |
01:35:04.400
The majority of the functions of the virus are dependent on the host.
link |
01:35:12.400
So let me do the devil's advocate, let me be the philosophical devil's advocate here and say,
link |
01:35:19.400
well, humans, which we would say are living, need our host planet to survive.
link |
01:35:27.400
So you can basically take every living organism that we think of as definitively living.
link |
01:35:34.400
It's always going to have some aspects of its host that it needs, of its environment.
link |
01:35:42.400
So is that really the key aspect of why a virus is that dependence?
link |
01:35:49.400
Because it seems to be very good at doing so many things that we consider to be intelligent.
link |
01:35:57.400
It's just that dependence part.
link |
01:36:00.400
Well, I mean, it's difficult to answer in this way.
link |
01:36:10.400
I mean, the way I think about the virus is, you know, in order for it to function,
link |
01:36:21.400
it needs to have the critical component, the critical tools that it doesn't have.
link |
01:36:31.400
So, I mean, in my way, it's not autonomous.
link |
01:36:42.400
That's how I separate the idea of the living organism on a very high level between the living organism
link |
01:36:50.400
and...
link |
01:36:51.400
And you have some, we have, I mean, these are just terms and perhaps they don't mean much,
link |
01:36:57.400
but we have some kind of sense of what autonomous means and that humans are autonomous.
link |
01:37:05.400
You've also done excellent work in the epidemiological modeling, the simulation of these things.
link |
01:37:15.400
So the zooming out outside of the body, doing the agent based simulation.
link |
01:37:19.400
So that's where you actually simulate individual human beings
link |
01:37:24.400
and then the spread of viruses from one to the other.
link |
01:37:28.400
How does at a high level agent based simulation work?
link |
01:37:33.400
All right.
link |
01:37:34.400
So it's also one of this irony of timing.
link |
01:37:40.400
Because, I mean, we've worked on this project for the past five years
link |
01:37:46.400
and the New Year's Eve, I got an email from my PhD student that the last experiments were completed.
link |
01:37:57.400
And three weeks after that, we get this Diamond Princess story
link |
01:38:03.400
and emailing each other with the same news saying like...
link |
01:38:09.400
So the Diamond Princess is a cruise ship.
link |
01:38:12.400
Yes.
link |
01:38:13.400
And what was the project that you worked on for five years?
link |
01:38:15.400
The project, I mean, the code name, it started with a bunch of undergraduates.
link |
01:38:23.400
The code name was Zombies on a Cruise Ship.
link |
01:38:27.400
So they wanted to essentially model the zombie apocalypse on a cruise ship.
link |
01:38:35.400
And after having some fun, we then thought about the fact that if you look at the cruise ships,
link |
01:38:44.400
the infectious outbreak has been one of the biggest threats to the cruise ship economy.
link |
01:38:53.400
So perhaps the most frequently occurring is the Norwalk virus.
link |
01:39:01.400
And this is essentially one of these stomach flus that you have.
link |
01:39:07.400
And it can be quite devastating.
link |
01:39:12.400
So occasionally there are cruise ships, they get canceled, they get returned back to the origin.
link |
01:39:24.400
And so we wanted to study, and this is very different from the traditional epidemiological studies
link |
01:39:31.400
where the scale is much larger.
link |
01:39:33.400
So we wanted to study this in a confined environment, which is a cruise ship, it could be a school,
link |
01:39:41.400
it could be other places such as this large company where people are in interaction.
link |
01:39:53.400
And the benefit of this model is we can actually track that in the real time.
link |
01:40:01.400
So we can actually see the whole course of the evolution, the whole course of the interaction between the infected host
link |
01:40:16.400
and the host and the pathogen, et cetera.
link |
01:40:21.400
So agent based system or multi agent system to be precisely is a good way to approach this problem
link |
01:40:37.400
because we can introduce the behavior of the passengers, of the crews.
link |
01:40:47.400
And what we did for the first time, that's where we introduced some novelty is we introduced a pathogen agent explicitly.
link |
01:40:59.400
So that allowed us to essentially model the behavior on the host side as well on the pathogen side.
link |
01:41:11.400
And all of a sudden we can have a flexible model that allows us to integrate all the key parameters about the infections.
link |
01:41:23.400
So for example, the virus, right?
link |
01:41:29.400
So the ways of transmitting the virus between the host.
link |
01:41:36.400
How long does virus survive on the surface, the fomite?
link |
01:41:44.400
What is, you know, how much of the viral particles does a host shed when he or she is asymptomatic versus symptomatic?
link |
01:42:02.400
And you can encode all of that into this pathogen. It's just for people who don't know.
link |
01:42:06.400
So agent based simulation, usually the agent represents a single human being.
link |
01:42:11.400
And then there's some graphs, like contact graphs that represent the interaction between those human beings.
link |
01:42:18.400
So, yes. So we, so essentially, you know, so agents are, you know, individual programs that are run in parallel.
link |
01:42:30.400
And we can provide instructions for these agents how to interact with each other, how to exchange information, in this case, exchange the infection.
link |
01:42:45.400
But in this case, in your case, you've added a pathogen as an agent. I mean, that's kind of fascinating.
link |
01:42:51.400
It's kind of a brilliant way to condense the parameters, to aggregate, to bring the parameters together that represent the pathogen, the virus.
link |
01:43:04.400
Yes. That's fascinating, actually.
link |
01:43:06.400
So, yeah, it was, you know, we realized that, you know, by bringing in the virus, we can actually start modeling.
link |
01:43:15.400
I mean, we are no longer bounded by very specific sort of aspects of the specific virus.
link |
01:43:24.400
So we end up, we started with, you know, Norwalk virus and of course, zombies.
link |
01:43:30.400
But we continued to modeling Ebola virus outbreak, flu, SARS, and because I felt that we need to add a little bit more sort of excitement for our undergraduate students.
link |
01:43:51.400
So we actually modeled the virus from the Contagion movie.
link |
01:43:56.400
So MEV1 and, you know, unfortunately, that virus and we tried to extract as much information.
link |
01:44:06.400
Luckily, the this movie was the scientific consultant was Ian Lipkin, a virologist from Columbia University, who is actually who provided.
link |
01:44:20.400
I think he designed this virus for this movie based on Nipah virus.
link |
01:44:26.400
And I think with some ideas behind SARS or flu like airborne viruses and, you know, the movie surprisingly contained enough details for us to extract and to model it.
link |
01:44:43.400
I was hoping you would like publish a paper of how this virus works.
link |
01:44:47.400
Yeah, we are planning to publish.
link |
01:44:49.400
I would love it if you did, but it would be nice if the, you know, if the the the origin of the virus.
link |
01:44:57.400
But you're now actually being a scientist and studying the virus from that perspective.
link |
01:45:01.400
But the origin of the virus, you know, you know, the first time I actually saw this movie is assignment number one in my bioinformatics class that they give.
link |
01:45:13.400
Because it it also tell it tells you that, you know, bioinformatics can be of use because if if I don't know you watched it.
link |
01:45:22.400
Have you watched it a long time ago?
link |
01:45:24.400
So so there is, you know, approximately a week from the virus detection.
link |
01:45:31.400
We see a screenshot of scientists looking at the structure of the surface protein.
link |
01:45:39.400
And this is where I tell my students that, you know, if you ask an experimental biologist, they will tell you that it's impossible because it takes months,
link |
01:45:49.400
maybe years to get the crystal structure of this, you know, the structure that is represented.
link |
01:45:55.400
If you ask a bioinformatician, they tell you, sure, why not just get it modeled.
link |
01:46:03.400
And and yes, but but it was very interesting to to see that there is actually, you know, and if you do it, do screenshots,
link |
01:46:17.400
you actually see the filogenetic tree, the evolutionary tree that relate this virus with other viruses.
link |
01:46:23.400
So it was a lot of scientific thought put into the movie.
link |
01:46:27.400
And one thing that I was actually, you know, it was interesting to learn is that the origin of this virus was there were two animals that led to the,
link |
01:46:41.400
you know, the the, you know, the zoonotic origin of this virus were fruit bat and a pig.
link |
01:46:51.400
So, you know, so this is this is this doesn't feel like we're this.
link |
01:46:57.400
This definitely feels like we're living in a simulation.
link |
01:47:00.400
OK, but maybe a big picture.
link |
01:47:05.400
Ageing based simulation now, larger scale, sort of not focused on inclusion, but larger scale are used now to drive some policy.
link |
01:47:14.400
So politicians use them to tell stories and narratives and try to figure out how how to move forward under so much, so much uncertainty.
link |
01:47:23.400
But in your sense, are ageing based simulation useful for actually predicting the future?
link |
01:47:31.400
Or are they useful mostly for comparing relative comparison of different intervention methods?
link |
01:47:37.400
Well, I think both because, you know, in the case of new coronavirus, we we essentially learning that the current intervention methods may not be efficient enough.
link |
01:47:53.400
One thing that one important aspect that I find to be so critical and yet something that was overlooked, you know, during the past pandemics is the effect of the asymptomatic period.
link |
01:48:18.400
This virus is different because it has such a long symptomatic period.
link |
01:48:25.400
And all of a sudden, that creates a completely new game when trying to contain this virus.
link |
01:48:33.400
In terms of the dynamics of the infection.
link |
01:48:36.400
Exactly.
link |
01:48:37.400
Do you also I don't know how close you're tracking this, but do you also think that there's a different rate of infection for when you're asymptomatic like that?
link |
01:48:52.400
That aspect or does a virus not care?
link |
01:48:55.400
So there were a couple of works.
link |
01:48:59.400
So one important parameter that tells us how contagious the the person was asymptomatic versus asymptomatic is looking at the number of viral particles this person sheds.
link |
01:49:18.400
You know, as a function of time.
link |
01:49:22.400
So, so far, what I saw is the study that tells us that the, you know, the person during the asymptomatic period is already contagious and it sheds the person sheds enough viruses to infect another host.
link |
01:49:47.400
And I think there's so many excellent papers coming out, but I think I just saw some maybe a nature paper that said the first week is when you're symptomatic or asymptomatic, you're the most contagious.
link |
01:50:00.400
So the highest level of the like the plot sort of in the 14 day period that collected a bunch of subjects.
link |
01:50:08.400
And I think the first week is when it's the most.
link |
01:50:11.400
Yeah, I think, I mean, I'm waiting, I'm waiting to see sort of more, more populated studies with higher numbers.
link |
01:50:24.400
My one of my favorite studies was, again, very recent one where scientists determined that
link |
01:50:35.400
tears are not contagious. So, so there is, you know, so there is no viral shedding done through, through tears.
link |
01:50:46.400
So they found one moist thing that's not contagious. And I mean, there's a lot of, I've personally been, because I'm on a survey paper, somehow that's looking at masks.
link |
01:51:01.400
And there's been so much interesting debates on the efficacy of masks.
link |
01:51:05.400
And there's a lot of work and there's a lot of interesting work on whether this virus is airborne.
link |
01:51:13.400
I mean, it's a totally open question. It's leaning one way right now, but it's a totally open question whether it can travel in aerosols long distances.
link |
01:51:22.400
I mean, do you have a, do you think about this stuff? Do you track this stuff? Are you focused on the, the bioinformatics of it?
link |
01:51:28.400
I mean, this is, this is a very important aspect for our epidemiology study.
link |
01:51:36.400
I think the, I mean, and it's sort of a very simple sort of idea, but I agree with people who say that the mask, the masks work in both ways.
link |
01:51:55.400
So it not only protects you from the, you know, incoming viral particles, but also, you know, it, it, you know, makes the potentially contagious person not to spread the viral particles.
link |
01:52:11.400
Who is, when they're asymptomatic may not even know that they're, in fact, it seems to be, there's evidence that they don't, surgical and certainly homemade masks,
link |
01:52:21.400
which is what's needed now actually, because there's a huge shortage of, they don't work as to protect you that well.
link |
01:52:29.400
They work much better to protect others. So it's, it's, it's a motivation for us to all wear one.
link |
01:52:36.400
Exactly. Cause I mean, you know, you don't know where, you know, about 30%, as far as I remember, at least 30% of the asymptomatic cases are completely asymptomatic.
link |
01:52:50.400
Right. So you don't really cough. You don't, I mean, you don't have any symptoms, yet you shed viruses.
link |
01:52:58.400
Do you think it's possible that we'll all wear masks? I wore a mask at a grocery store and you just, you get looks.
link |
01:53:05.400
I mean, this was like a week ago. Maybe it's already changed because I think CDC or somebody, I think the CDC has said that we should be wearing masks, like LA, they starting to happen.
link |
01:53:17.400
But do you, it just seems like something that this country will really struggle doing or no?
link |
01:53:24.400
I hope not. I mean, you know, it, it was interesting. I was looking through the, through the old pictures during the Spanish flu and you could see that the, you know,
link |
01:53:40.400
pretty much everyone was wearing masks with some exceptions and they were like, you know, sort of iconic photograph of the, I think it was San Francisco, this tram who was refusing to let in a, you know, someone without the mask.
link |
01:53:58.400
So I think, well, you know, it's also, you know, it's related to the fact of how much we are scared. Right. So how much do we treat this problem seriously?
link |
01:54:16.400
And, you know, my take on it is we should, because it is very serious.
link |
01:54:28.400
Yeah, I, from a psychology perspective, just worry about the entirety, the entire big mess of a psychology experiment that this is, whether mask will help it or hurt it. You know, masks have a way of distancing us from others by removing the emotional expression and all that kind of stuff.
link |
01:54:51.400
But at the same time, mask also signal that I care about your wellbeing. Exactly. So it's a really interesting trade off. That's just, yeah, it's, it's interesting, right? About distancing. Aren't we distanced enough?
link |
01:55:07.400
Right. Exactly. And when we try to come closer together, when they do reopen the economy, that's going to be a long road of rebuilding trust and not all being huge germaphobes.
link |
01:55:24.400
Let me ask sort of, you have a bit of a Russian accent, Russian or no Russian accent? Were you born in Russia? Yes. And you're too kind. I have a pretty thick Russian accent.
link |
01:55:41.400
What are your favorite memories of Russia?
link |
01:55:44.400
So I moved first to Canada and then to the United States back in 99. So by that time I was 22. So, you know, whatever Russian accent I got back then, you know, it stuck with me for the rest of my life.
link |
01:56:07.400
You know, it's, yeah, so I, you know, by the time the Soviet Union collapsed, I was, you know, I was a kid, but sort of, you know, old enough to realize that there are changes.
link |
01:56:28.400
Did you want to be a scientist back then?
link |
01:56:30.400
Oh, yes. Oh, yeah. I mean, my first, the first sort of 10 years of my sort of, you know, juvenile life, I wanted to be a pilot of a passenger jet plane.
link |
01:56:50.400
Wow.
link |
01:56:51.400
So yes, it was like, you know, I was getting ready, you know, to go to a college to get the degree, but I've been always fascinated by science.
link |
01:57:06.400
And, you know, so not just by math, of course, math was one of my favorite subjects, but, you know, biology, chemistry, physics, somehow I, you know, I liked those four subjects together.
link |
01:57:22.400
And yes, so essentially after a certain period of time, I wanted to actually, back then it was a very popular sort of area of science called cybernetics.
link |
01:57:42.400
So it's sort of, it's not really computer science, but it was like, you know, computational robotics in this sense.
link |
01:57:50.400
And so I really wanted to do that. And but then, you know, I, you know, I realized that, you know, my biggest passion was in mathematics.
link |
01:58:06.400
And later I, you know, when, you know, studying in Moscow State University, I also realized that I really want to apply the knowledge.
link |
01:58:20.400
So I really wanted to mix, you know, the mathematical knowledge that I get with real life problems.
link |
01:58:31.400
And that could be, you mentioned chemistry and now biology. And I sort of, does it make you sad?
link |
01:58:41.400
Maybe I'm wrong on this, but it seems like it's difficult to be in collaboration, to do open, big science in Russia.
link |
01:58:54.400
From my distant perspective in computer science, I don't, I'm not, I can go to conferences in Russia.
link |
01:59:01.400
I sadly don't have many collaborators in Russia. I don't know many people doing great AI work in Russia.
link |
01:59:09.400
Does it make, does that make you sad? Am I wrong in seeing it this way?
link |
01:59:14.400
Well, I mean, I am, I have to tell you, I am privileged to have collaborators in bioinformatics in Russia. And I think this is the bioinformatics school in Russia is very strong.
link |
01:59:29.400
In Moscow?
link |
01:59:30.400
In Moscow, in Novosibirsk, in St. Petersburg, have great collaborators in Kazan. And so at least, you know, in terms of, you know, my area of research.
link |
01:59:51.400
There's strong people there.
link |
01:59:53.400
Yeah, strong people, a lot of great ideas, very open to collaborations. So I, perhaps, you know, it's my luck, but, you know, I haven't experienced, you know, any difficulties in establishing collaborations.
link |
02:00:13.400
That's bioinformatics though.
link |
02:00:14.400
It could be bioinformatics too. And it could, yeah, it could be person by person related, but I just don't feel the warmth and love that I would, you know, you talk about the Seminole people who are French in artificial intelligence.
link |
02:00:29.400
France welcomes them with open arms in so many ways. I just don't feel the love from Russia. I do on the human beings, like people in general, like friends and just cool, interesting people. But from the scientific community, no conferences, no big conferences.
link |
02:00:49.400
Yeah, it's actually, you know, I'm trying to think. Yeah, I cannot recall any big AI conferences in Russia.
link |
02:01:00.400
It has an effect on, for me, I haven't sadly been back to Russia. But my problem is it's very difficult. So now I have to renounce the citizenship.
link |
02:01:13.400
Oh, is that right?
link |
02:01:14.400
I mean, I'm a citizen in the United States and it makes it very difficult. There's a mess now, right? So, I want to be able to travel like, you know, legitimately.
link |
02:01:25.400
Yeah.
link |
02:01:26.400
And it's not an obvious process that will make it super easy. I mean, that's part of that, like, you know, it should be super easy for me to travel there.
link |
02:01:34.400
Well, you know, hopefully this unfortunate circumstances that we're in will actually promote the remote collaborations.
link |
02:01:47.400
Yes.
link |
02:01:48.400
And I think what we are experiencing right now is that you still can do science, you know, being quarantined in your own homes, especially when it comes. I mean, you know, I certainly understand there is a very challenging time for experimental scientists.
link |
02:02:06.400
I mean, I have many collaborators who are, you know, who are affected by that. But for computational scientists.
link |
02:02:13.400
Yeah, we're really leaning into the remote communication. Nevertheless, I had to force you to talk to you in person because there's something that you just can't do in terms of conversation like this.
link |
02:02:25.400
I don't know why, but in person is very much needed. So I really appreciate you doing it.
link |
02:02:31.400
You have a collection of science bobbleheads.
link |
02:02:34.400
Yes.
link |
02:02:35.400
Which look amazing. Which bobblehead is your favorite and which real world version, which scientist is your favorite?
link |
02:02:46.400
So yeah, by the way, I was trying to bring it in, but they are quarantined now. In my office, they sort of demonstrate the social distance so they're nicely spaced away from each other.
link |
02:03:01.400
But so, you know, it's interesting. So I've been collecting those bobbleheads for the past maybe 12 or 13 years. And it, you know, interestingly enough, it started with the two bobbleheads of Watson and Crick.
link |
02:03:19.400
And interestingly enough, my last bobblehead in this collection for now, and my favorite one, because I felt so good when I got it, was the Rosalind Franklin.
link |
02:03:35.400
Who is the full group? So I have Watson, Crick, Newton, Einstein, Marie Curie, Tesla, of course, Charles Darwin, and Rosalind Franklin.
link |
02:03:58.400
I am definitely missing quite a few of my favorite scientists. And but so, you know, if I were to add to this collection, so I would add, of course, Kolmogorov.
link |
02:04:16.400
That's, you know, I've been always fascinated by his, well, his dedication to science, but also his dedication to educating young people, the next generation. So it's very inspiring.
link |
02:04:36.400
He's one of the, okay, yeah, he's one of the Russia's greats. Yes. Yeah. So he also, you know, the school, the high school that I attended was named after him, and he was great.
link |
02:04:51.400
You know, so he founded the school, and he actually taught there.
link |
02:04:58.400
Is this in Moscow? Yes. So, but then, I mean, you know, other people that I would definitely like to see in my collections was, would be Alan Turing, would be John von Neumann.
link |
02:05:18.400
Yeah, you're a little bit late on the computer scientists. Yes. Well, I mean, they don't, they don't make them, you know, I still am amazed that they haven't made Alan Turing yet.
link |
02:05:29.400
Yes. And I would also add Linus Pauling. Linus Pauling. Who is Linus Pauling?
link |
02:05:40.400
So this is, this is, to me is one of the greatest chemists. And the person who actually discovered the secondary structure of proteins, who was very close to solving the DNA structure.
link |
02:06:00.400
And, you know, people argue, but some of them were pretty sure that if not for this, you know, photograph 51 by Rosalind Franklin that, you know, Watson and Crick got access to, he would be, he would be the one who would solve it.
link |
02:06:26.400
Science is a funny race. It is. Let me ask the biggest and the most ridiculous question. So you've kind of studied the human body and its defenses and these enemies that are about from a biological perspective, bioinformatics perspective, a computer scientists perspective.
link |
02:06:51.400
How has that made you see your own life, sort of the meaning of it, or just even seeing your, what it means to be human?
link |
02:07:04.400
Well, it certainly makes me realizing how fragile the human life is. If you think about this little tiny thing can impact the life of the whole human kind to such extent.
link |
02:07:25.400
So, you know, it's, it's something to appreciate and to remember that, that, you know, we are fragile, we have to bond together as a society.
link |
02:07:51.400
And, you know, it also gives me sort of hope that what we do as scientists is useful.
link |
02:08:05.400
Well, I don't think there's a better way to end it. Dmitry, thank you so much for talking today. It was an honor.
link |
02:08:09.400
Thank you very much.
link |
02:08:11.400
Thanks for listening to this conversation with Dmitry Korkin. And thank you to our presenting sponsor, Cash App. Please consider supporting the podcast by downloading Cash App and using code LexPodcast.
link |
02:08:22.400
If you enjoy this podcast, subscribe on YouTube, review it with five stars on Apple Podcast, support it on Patreon, or simply connect with me on Twitter at LexFriedman.
link |
02:08:33.400
And now, let me leave you with some words from Edward Osborne Wilson, E.O. Wilson, the variety of genes on the planet and viruses exceeds or is likely to exceed that in all of the rest of life combined.
link |
02:08:49.400
Thank you for listening and hope to see you next time.