Ryan Hisner Profile picture
Oct 1 48 tweets 16 min read Twitter logo Read on Twitter
I think this preprint, by @Stuartturville, is potentially one of the more important recent SARS-CoV-2 papers. It promises to unwrap some of the most mystifying aspects of Omicron surrounding cell entry, TMPRSS2 use, & reduced lung invasion/pneumonia. 1/48
When BA.1 emerged, it was apparent it did not cause the same degree of pneumonia or disease severity as Delta (though unprecedented levels of infection resulted in similarly large numbers of hospitalizations & deaths in many areas). h/t @mike_famulare 2/48
Many studies showed this was attributable to Omicron’s impaired ability to infect lung cells & reduced cell-cell fusion (syncytia formation). But what accounted for these attributes of Omicron?
(Image 1 from paper by @GuptaR_lab, @veeslerlab @sigallab @SystemsVirology) 3/48
Image
Image
A little background first: The SARS-CoV-2 spike must undergo two cleavages to efficiently infect a cell. The first cleavage comes at the S1/S2 boundary around amino acid 685 of the 1273-residue spike protein. 4/48 Image
S1/S2 cleavage is performed by the enzyme furin in the Golgi apparatus of an infected cell. S1 & S2 remain connected but are held together by non-covalent bonds rather than much stronger covalent bonds. This optimally primes spike for later S2’ cleavage & cell entry. 5/48 Image
The 2nd spike cleavage essential for SARS-CoV-2 cell entry occurs around 816, called S2’. There are two major ways S2’ cleavage could happen. First, 2’ can be cleaved by TMPRSS2, leading to cell membrane fusion entry. 6/48 Image
Importantly, TMPRSS2—a protein-cutting enzyme embedded in cell membranes—is especially common in lung cells, so the ability of a variant to undergo efficient S2’ cleavage by TMPRSS2 is essential for lower-respiratory tract (LRT) tropism. 7/48 Image
Alternatively, the virus could undergo endosomal entry, in which it enters the cell enveloped in an endosome. In this case, enzymes called cathepsins cleave S2’, allowing the virus to fuse with the endosomal membrane and enter the cytoplasm, where it replicates. 8/48 Image
After BA.1 emerged, most studies seemed to agree that its impaired ability to infect the lower respiratory tract (LRT) & fuse cells was connected to its inability to utilize TMPRSS2 for efficient spike cleavage and subsequent cell membrane fusion. 9/48


Image
Image
Image
Image
Instead, in an abrupt change from previous lineages, all of which primarily used TMPRSS2/cell membrane fusion for entry, it seemed that Omicron favored the endosomal pathway and subsequent S2’ cleavage within the endosome by cathepsins. 10/48 Image
The endosomal-entry story neatly explained the two most striking changes in Omicron compared to previous lineages: its preference for the upper-respiratory tract (URT) & its markedly reduced cell-cell fusion capabilities.

There’s only one problem: it’s wrong.
11/48
Many studies have now shown that, in human airway cells & other models, Omicron not only utilizes TMPRSS2 but does so hyper-efficiently. See paper by @EnyaQing & Tom Gallagher that ingeniously uses viral-like particles (VLPs) & extracellular vesicles (EVs) to show… 12/48 Image
…that compared to pre-Omicron lineages, Omicron is hypersensitive to S2’ cleavage by TMPRSS2, which activates spike for fusion & cell entry. (This paper is a trove of surprising & intriguing findings, particularly the bits on S2. Deserves its own thread, really.) 13/48 Image
Also see the earlier observation by @PeacockFlu & @wendybarclay11 that Omicron very efficiently used TMPRSS2 in human nasal cells, even as TMPRSS2 seemed almost entirely incapable of Omicron S2’ cleavage in traditional lab cell lines—now a familiar pattern. 14/48 Image
Most recently, separate studies by @bart_haagmans & @GartnerM94 both found that in human airway organoid cells, all SARS-CoV-2 lineages, including Omicron, required TMPRSS2—but not cathepsins (used in endosomal entry) for cell entry. 15/48
As @bart_haagmans put it in his recent study in @JVirology,
“The data indicate that the preferred SARS-CoV-2 entry route depends on the type of cell line being used.” 16/48 Image
It’s now clear that in human cells, Omicron utilizes TMPRSS2 very efficiently & indeed apparently *requires* TMPRSS2 for successful infection. Endosomal entry seems an artifact confined to lab cell lines, many of which overexpress TMPRSS2 & ACE2. 17/48 Image
But if Omicron S2’ cleavage by TMPRSS2 is ultra-efficient, leading to successful cell membrane-fusion entry—strongly implying that endosomal entry plays no major role—why have Omicron lineages been so unsuccessful at infecting lung cells, which have TMPRSS2 in spades? 18/48
It was all an utter mystery to me.

Until I read the new paper by @StuartTurville & @milogiannakis. In it, they provide a compelling explanation for the contradictory findings regarding Omicron & TMPRSS2 & for Omicron’s reduced LRT tropism. 19/48
When BA.1 emerged, labs struggled to grow it in cell lines they’d successfully used to grow pre-Omicron variants: “In many cases… the ACCE2-TMPRSS2 pathway…appeared no longer functional for Omicron lineages.” So Turville engineered a cell line—VeroE6-T2 (E6-T2)— 20/48 Image
—in which all variants, including Omicrons, replicated well. Puzzlingly, E6-T2 highly expressed TMPRSS2, which in previous studies (& all other cell lines Turville tested) potently inhibited Omicron replication. Somehow, in contrast to other engineered cell lines… 21/48 Image
…rich in TMPRSS2, Omicron S2’ cleavage successfully takes place in E6-T2 cells. Turville proved Omicron utilized TMPRSS2 by showing that a TMPRSS2 inhibitor (Nafamostat) stifled Omicron replication while a compound that prevents endosomal cell entry (E64D) had no effect. 22/48 Image
Exactly why Omicron succeeds in E6-T2 cells but not other high-TMPRSS2 cell lines remains a mystery (as far as I can tell). Their next experiment, however, was even more confounding. VeroE6-T2 cells have low levels of ACE2… 23/48 Image
Since ACE2 is the essential cell-membrane receptor used by all SARS-CoV-2, higher ACE2 expression should increase viral cell entry & replication, & this is what previous studies have found. So Turville created an E6-T2 cell line with high levels of ACE2, (E6-T2-ACE2). 24/48 Image
As expected, increased ACE2 levels enhanced replication—in pre-Omicron variants. But for Omicron, it potently *inhibited* replication. Even within Omicron & pre-Omicron lineages, there’s a distinct downward trend in viral titers in high-ACE2+TMPRSS2 cells—note log scale. 25/48 Image
What’s going on here? Why in the world would increasing the density of a virus’s receptor reduce its ability to infect a cell? Quite the riddle. 26/48
To understand the (partial) answer, we need to know that TMPRSS2 can cleave ACE2. (I felt rather stupid for not knowing this.) It sounds as if this might destroy its usefulness as a receptor, but ACE2 actually *requires* this cleavage to perform its function in the… 27/48
…renin-angiotensin-aldosterone system (RAAS), which regulates blood pressure & electrolyte balance. TMPRSS2 cleavage preserves ACE2 in the cell membrane by competing with a different enzyme (ADAM17) that cleaves ACE2 in a way that chucks it off the membrane altogether. 28/48 Image
Two great papers, one by Tom Gallagher & Stanley Perlman & one by @snpoehlm, showed TMPRSS2-mediated ACE2 cleavage potently enhanced SARS-CoV-1 replication—30-fold increase in cell entry, 9-fold increase in viral RNA—much like in pre-Omi SARS-2 variants. 29/48
Image
Image
But ACE2 also plays an entirely different role in the body: pairing up with one of two solute-carrier proteins—B0AT1 (also called SLC6A19) or SIT1 (SLC6A20) to chaperone certain amino acids across the plasma membrane & into cells. 30/48 Image
In fact, that vast majority of ACE2 in the body acts in this chaperone role rather than its more well-known RAAS role. How is this relevant? ACE2 in its chaperone role, attaches to B0AT1 or SIT1 in a way that conceals its cleavage site, preventing TMPRSS2 from cutting it. 31/48 Image
In a 2020 paper, the man who discovered & first described the function of B0AT1, Bruce R. Stevens, wrote a fantastic preprint describing how the B0AT1-ACE2 chaperone complex hides the ACE2 cleavage site from TMPRSS2 in all conformations. 32/48 Image
Now we finally seem to have the beginnings of an explanation for why Omicron’s strikingly reduced ability to invade the lungs: the high LRT levels of TMPRSS2 efficiently cleave ACE2, which improves pre-Omi cell entry & replication but potently inhibits Omicron variants. 33/48
Turville provided further support for this hypothesis by putting an uncleavable version of ACE2 in the high-ACE2+TMPRSS2 E6-T2-ACE2 cells that Omicron previously failed in. Result: Omicron replication completely rescued & now equal to pre-Omicron variants. 34/48 Image
Since intestinal ACE2 overwhelmingly functions in its amino acid-transport chaperone role (which cannot be cleaved by TMPRSS2), one would expect intestinal replication of Omicron to be as efficient as pre-Omicron variants, in contrast to the attenuation seen in lungs. 35/48
Wastewater SARS-CoV-2 levels, which presumably reflect intestinal replication, seem to confirm this. WW levels at the peak the Omicron and Delta waves correspond well to estimated case prevalence based on both seroprevalence & official case numbers. 36/48 Image
This paper casts a clarifying light on the crucial & confounding mystery of why Omicron fails to succeed in lung cells despite its demonstrated efficiency at utilizing TMPRS2. It appears a real breakthrough to me that deserves more attention than it seems to have received. 37/48
Many mysteries remain. Why exactly does ACE2 cleavage by TMPRSS2 enhance pre-Omicron lineage infection? And how does this cleavage inhibit Omicron? The most obvious explanation is that Omicron’s extremely divergent spike cannot bind to cleaved ACE2. But this can’t be right. 38/48
Omicron successfully infected Vero-E6-T2 cells, which had high levels of TMPRSS2 & low levels of ACE2 was drastically inhibited in E6-T2 cells with high levels of both TMPRSS2 & ACE2. If Omicron’s failure was due to an inability to utilize cleaved ACE2… 39/48
… then it ought to have done better in the high-ACE2+TMPRSS2 cells, where a small amount of uncleaved ACE2 might have slipped through. But the opposite happened. There seems to be something about the absolute amount of ACE2 cleavage itself that stymies Omicron. 40/48
High ACE2 affinity seems to result in greater inhibition in TMPRSS2 + cleavable ACE2 cells compared to the same cells w/non-cleavable ACE2. Is this a clue that could lead us to an explanation?
A million things are going on in cells, so the explanation is likely complex. 41/48
Another major mystery to me is why Omicron is able to replicate so well in human nasal epithelial cells (hNECs). ACE2 acts in its RAAS role there, as in the lungs, so it ought to be cleaved by TMPRSS2. Yet Omicron seems proficient at using TMPRSS2 in hNECs. 42/48 Image
@GartnerM94’s study is suggestive on this question. First, note that Omicron is actually outcompeted by WT & Delta in nasal cells (& other respiratory cell tested) 2 days post-infection (dpi). But Omicron somehow gets a jump start on the others—as in @PeacockFlu’s study. 43/48
Image
Image
Furthermore, while the TMPRSS2 inhibitor Camostat annihilated infection by WT & Delta, Omicron replication was only mildly reduced. But a compound inhibiting endosomal entry (CA-074 Me) had ~zero effect on all variants, so this can’t explain the discrepancy. 44/48 Image
Gartner suggests Omicron may be utilizing a different protease for S2’ cleavage in hNECs. If so, in addition to explaining Omicron’s success in the face of TMPRSS2 inhibition, might this somehow also explain its remarkably rapid replication in the first hours of infection? 45/48 Image
Omicron doesn’t fuse cells (form syncytia) as much as previous variants. One merit of the endosomal-entry hypothesis was that it readily explained this. In endosomal entry, membrane fusion & occurs in endosomes within the cell, not on the cell membrane. 46/48 Image
But now that endosomal entry seems disproven and especially given findings that Omicron may actually be much *more* primed for fusion than previous variants, its reduced syncytia formation again begs for an explanation. (@EnyaQing & Tom Gallagher again below.) 47/48 Image
I’m sure others have far more insight on these questions than I do.
In any case, it’s exciting to see some light being shed on the various Omicron enigmas, & I hope others build on Turville’s insights & illuminate more of Omicron’s dark secrets. 48/48

• • •

Missing some Tweet in this thread? You can try to force a refresh
 

Keep Current with Ryan Hisner

Ryan Hisner Profile picture

Stay in touch and get notified when new unrolls are available from this author!

Read all threads

This Thread may be Removed Anytime!

PDF

Twitter may remove this content at anytime! Save it as PDF for later use!

Try unrolling a thread yourself!

how to unroll video
  1. Follow @ThreadReaderApp to mention us!

  2. From a Twitter thread mention us with a keyword "unroll"
@threadreaderapp unroll

Practice here first or read more on our help page!

More from @LongDesertTrain

Sep 28
Charles Chaplin, ultra-influential author and public health official in the early 1900s, said almost the exact same thing. If disease is airborne, people will despair & not be properly sanitary. Therefore, we must tell them disease is not airborne. 1/3
thebaffler.com/salvos/airborn…

Image
Chaplin's view was influential—not because the evidence favored droplet transmission but because his views led to politically acceptable conclusions. With droplet transmission no public action or business regulation was required. @BenEhrenreich: 2/3 thebaffler.com/salvos/airborn…
Image
It's hard to put it any better than @jljcolorado: "Droplets & surfaces are very convenient for people in power—all the responsibility is on the individual. ...if you admit it is airborne, institutions, governments, & companies have to do something." 3/3
(Credit to @mdc_martinus) Image
Read 4 tweets
Sep 26
The main database for SARS-CoV-2 genomes—GISAID—has been operating so slowly for the past 4 days that it is essentially non-functional. We know GISAID is actively throttling accounts, possibly through some sort of algorithm.
Is this accidental or intentional? No one knows. 1/4
Every mouse click or any other action results in a 5-20-second delay. Is this necessary because there is some sort of long-lasting technical problem at GISAID? The result of a faulty algorithm? Something else? No one knows. 2/4
Is the network on the verge of collapse? Or is everything operating perfectly & the current uselessness of GISAID just a technical glitch? If the latter, why is it taking so long to fix? When will GISAID be operational again?
Again, no one knows. 3/4
Read 4 tweets
Sep 17
In the estimable @GuptaR_lab's hands, BA.2.86 has the highest fusogenicity of all Omicron lineages, though it remains far lower than Delta.
Fusogenicity is generally associated with more severe disease.
OTOH, the only other study I know of measuring BA.2.86 fusogenicity... 1/3
...showed precisely opposite results. We've seen similarly contradictory results regarding BA.2.86 infectivity.
It seems the only thing for certain about BA.2.86 is that it's the most difficult variant to study yet.

Just to be clear... 2/3
...these results are from the best scientists on the planet. Conflicting results are normal part of science, especially biology, where even the simplest systems are incomprehensibly complex. This is just an indication of how incredibly difficult research is. 3/3
Read 4 tweets
Sep 12
TL;DR—Among mutations common in chronic infections are two in PLpro, which combats innate immunity. Both mutations change the SARS-2 to a SARS-1-type AA & have clear effects—remarkable intrahost functional convergence. Then questions + wild speculation at thread's end. 1/58
If you follow SARS-CoV-2 evolution, you notice certain mutations patterns repeatedly appearing. This is even more true for chronic-infection sequences. The hallmark mutations pop up over & over. In some cases, we even have a decent idea of what they do. 2/
Thanks to the amazing work of @jbloom_lab & @tylernstarr, we can inspect almost any mutation in the spike RBD (receptor binding domain) and get a very good idea of how it affects ACE2 binding & spike stability. 3/58 tstarrlab.github.io/SARS-CoV-2-RBD…
Read 58 tweets
Sep 9
@LooftSabine @EckerleIsabella @PeacockFlu What's fascinating is that despite the clear evolutionary pressure that led to zero or near-zero ORF8 expression in XBB.1*, BA.5, and XBC (and previously Alpha), XBB.2.3 has steadfastly maintained its intact ORF8.

One other XBB.2.3 peculiarity is, I think, connected...
@LooftSabine @EckerleIsabella @PeacockFlu Or at least might be. And that's the fact that while ORF9b:I5T has proven clearly advantageous in XBB.1* (as well as most other lineages), XBB.2.3 has virtually never acquired it.
What could the connection be? ORF9b & ORF8 both have immune-evasion functions of some sort...
@LooftSabine @EckerleIsabella @PeacockFlu ORF9b's mechanism of innate immune evasion (one of them, anyway) is fairly clear—suppression of type-1 interferon, achieved through K63-linked deubiquitination. ORF8's functions & mechanism are much more nebulous (at least it seems so to me), but the fact that...
Read 9 tweets
Sep 5
Went to check the nuc context & expected to see an ideal context for APOBEC-induced C->T mutation... but found the opposite. C26681 is followed by a C, easily the worst nuc there. Of 16 possibilities, TCC is the 2nd-worst for C->T. Shay's onto something here.
Translation... 1/
Image
...even though C->T nucleotide mutations are the most common type, this is not a site you'd expect a C->T mutation to frequently occur at. The fact that C26681T has been found in 7 variants likely indicates it has a real effect—despite not changing the amino acid sequence. 2/
What does C26681T do? I doubt anyone knows. There's something called "secondary RNA structure," which refers to the way SARS-CoV-2's RNA genome twists around, matching nucleotides bonding to each other. (G-C, A-T, but also G-T). Perhaps C26881T alters this structure. 3/
Read 8 tweets

Did Thread Reader help you today?

Support us! We are indie developers!


This site is made by just two indie developers on a laptop doing marketing, support and development! Read more about the story.

Become a Premium Member ($3/month or $30/year) and get exclusive features!

Become Premium

Don't want to be a Premium member but still want to support us?

Make a small donation by buying us coffee ($5) or help with server cost ($10)

Donate via Paypal

Or Donate anonymously using crypto!

Ethereum

0xfe58350B80634f60Fa6Dc149a72b4DFbc17D341E copy

Bitcoin

3ATGMxNzCUFzxpMCHL5sWSt4DVtS8UqXpi copy

Thank you for your support!

Follow Us on Twitter!

:(