Sunday, December 19, 2021

COVID-19 vaccine: What’s RNA                                      research got to do with it?        

December 14, 2020
Illustration of coronavirus protein binding to receptor on human cell.A coronavirus uses a protein on its membrane—shown here in red in a molecular model—to bind to a receptor—                                                                                                   shown in blue—on a human cell to enter the cell. Once inside, the virus uses the cells' machinery to make more                                                                             copies of itself. (Juan Gaertner / Science Source)               
Rochester research into RNA structure and             function provides key information for developing coronavirus treatments.

The US Food and Drug Administration recently approved emergency use                                                                authorization for a COVID-19 vaccine developed by Pfizer and the                                                                        German pharmaceutical company BioNTech.

The vaccine made history not only because it reported a 95 percent efficacy rate                                                                        at preventing COVID-19 in clinical trials, but because it is the first vaccine                                                                    ever approved by the FDA for human use that is based on RNA technology.

“The development of RNA vaccines is a great boon to the future of treating                                                                   infectious diseases,” says Lynne Maquat, the J. Lowell Orbison Distinguished                                                          Service Alumni Professor in biochemistry and biophysics, oncology, and                                                                pediatrics at Rochester and the director of Rochester’s Center for RNA Biology.

COVID-19, short for “coronavirus disease 2019,” is caused by the novel                                                              coronavirus SARS-CoV-2. Like many other viruses, SARS-CoV-2 is an RNA virus.                                                               This means that, unlike in humans and other mammals, the genetic material for                                                           SARS-CoV-2 is encoded in ribonucleic acid (RNA).                                                                                                        The viral RNA is sneaky: its features cause the protein synthesis machinery in                                                         humans to mistake it for RNA produced by our own DNA.

For that reason, several of the leading COVID-19 vaccines and treatments                                                                                      are based on RNA technology.

A contingent of researchers at the University of Rochester study the RNA of                                                           viruses to better understand how RNAs work and how they are involved in                                                               diseases. This RNA research provides an important foundation for developing                                                         vaccines and other drugs and therapeutics to disrupt the virus and stop infections.

“Understanding RNA structure and function helps us understand how to throw a                                                              therapeutic wrench into what the COVID-19 RNA does—make new virus that can                                                             infect more of our cells and also the cells of other human beings,” Maquat says.


In the past few decades, as scientists came to realize that genetic material is largely regulated by the RNA it encodes, that most of our DNA produces RNA,       and that RNA is not only a target but also a tool for disease therapies, “the RNA research world has exploded,” Maquat says. “The University of Rochester                                                        understood this.”

In 2007, Maquat founded The Center for RNA Biology as a means of                                                                 conducting interdisciplinary research in the function, structure, and processing                                                                  of RNAs. The Center involves researchers from both the River Campus and the                                                                     Medical Center, combining expertise in biology, chemistry, engineering, neurology,                                                       and pharmacology.

“Our strength as a university is our diversity of research expertise, combined                                                                 with our highly collaborative nature,” says Dragony Fu, an associate professor                                                                of biology on the River Campus and a member of the Center for RNA Biology.                                                             “We are surrounded by outstanding researchers who enhance our understanding                                                           of RNA biology, and a medical center that provides a translational aspect where                                                                        the knowledge gained from RNA biology can be applied for therapeutics.”

How does RNA relate to disease?

graphic created by the New York Times illustrates how the coronavirus that                                                            causes COVID-19 enters the body through the nose, mouth, or eyes and attaches                                                                        to our cells. Once the virus is inside our cells, it releases its RNA.                                                                                Our hijacked cells serve as virus factories, reading the virus’s RNA and making                                                                     long viral proteins to compromise the immune system.                                                                                                        The virus assembles new copies of itself and spreads to more parts of the body                                                                   and—by way of saliva, sweat, and other bodily fluids—to other humans.

“Once the virus is in our cells, the entire process of infection and re-infection                                                          depends on the viral RNA,” Maquat says.

One of the reasons viruses are such a challenge is that they change and                                                                  mutate in response to drugs.

That means novel virus treatments and vaccines have to be created each time                                                                a new strain of virus presents itself. Armed with innovative research on the                                                                        fundamentals of RNA, scientists are better able to develop and test therapeutics                                                                  that directly target the RNAs and processes critical to a virus’s life cycle.

How do RNA vaccines work?

Traditional vaccines against viruses like influenza inject inactivated virus proteins                                                                                  called antigens. The antigens stimulate the body’s immune system to recognize                                                                      the specific virus and produce antibodies in response, with the hope                                                                                   that these antibodies will fight against future virus infection.

RNA-based vaccines—such as those developed by Pfizer/BioNTech and American                                               biotechnology company Moderna—do not introduce an antigen, but instead                                                                                          inject a short sequence of synthetic messenger RNA (mRNA) that is enclosed in a                                                    specially engineered lipid nanoparticle.                                                                                                                          This mRNA provides cells with instructions to produce the virus antigen                                                                themselves.

Once the mRNA from a vaccine is in our body, for example,                                                                                               it “instructs” the protein synthesis machinery in our cells, which normally                                                                           generates proteins from the mRNAs that derive from our genes, to produce                                                                        a piece of the SARS-CoV-2 virus spike protein.                                                                                                               Since the SARS-CoV-2 virus spike protein is foreign to our bodies,                                                                                   our bodies will then make antibodies that inactivate the protein.

“Should the virus enter our body from an infected person,                                                                                              these antibodies will bind to and inactivate the virus by binding to its                                                                                spike proteins, which coat the outside of the viral capsule,” Maquat says.

An RNA-based vaccine therefore acts as a code to instruct our human cells to                                                                                            make many copies of the virus protein which, as a consequence, creates                                                           antibodies resulting in an immune response.

Unlike more traditional vaccines, RNA-based vaccines are also beneficial in                                                                      that they eliminate the need to work with the actual virus.

“Working with a live virus is costly and very involved, requiring that researchers                                                              use special biosafety laboratories and wear bulky personal protective                                                                         equipment so that the virus is ‘biocontained,’ and no one gets infected,”                                                                         Maquat says.

Developing a vaccine from a live virus additionally takes much longer than                                                                     generating an mRNA-based vaccine, but “no one should think the process is                                                                simple,”  Maquat says of the Pfizer/BioNTech vaccine.                                                                                                 “Since it is the first of its kind, a lot had to be worked out.”

What does RNA stand for?

RNA stands for ribonucleic acid.

What is RNA?

RNA delivers the genetic instructions contained in DNA to the rest of the cell.

What does Covid stand for?

Covid-19 stands for “coronavirus disease 2019.”

How is Rochester’s RNA research applicable to COVID-19? 

Horizontal portraits of Doug Anderson, Dragony Fu, and Lynne Maquat, scientists who study RNA of viruses.






Researchers Douglas Anderson, Dragony Fu, and Lynne Maquat are among the scientists at the University of Rochester                                                                                                                who study the RNA of viruses to better understand how RNAs work and how they are involved in diseases.                                                                                                              (University of Rochester photos / Matt Wittmeyer / J. Adam Fenster)                                                                                                                                                                                               

Maquat has been studying RNA since 1972 and was part of the earliest wave of                                                     scientists to realize the important role RNA plays in human health and disease.

Our cells have a number of ways to combat viruses in what can be viewed as                                                                 an “arms race” between host and virus.                                                                                                                            One of the weapons in our cells’ arsenal is an RNA surveillance mechanism                                                                     Maquat discovered called nonsense-mediated mRNA decay (NMD).

“Nonsense-mediated mRNA decay protects us from many genetic mutations that                                                           could cause disease if NMD were not active to destroy the RNA harbouring the                                                                mutation,” she says.

Maquat’s discovery has contributed to the development of drug therapies for                                                                 genetic disorders such as cystic fibrosis, and may be useful in developing                                                                        treatments for coronavirus.

“NMD also helps us combat viral infections, which is why many viruses either                                                               inhibit or evade NMD,” she adds. “The genome of the virus COVID-19 is                                                                           a positive-sense, single-stranded RNA. It is well known that other positive-sense,                                                          single-stranded RNA viruses evade NMD by having RNA structures that prevent                                                             NMD from degrading viral RNAs.”

Maquat’s lab has been collaborating with a lab at Harvard University to test how                                                                 viral proteins can inhibit the NMD machinery.

Their recent work is focused on the SARS-CoV-2 structural protein called N. Lab                                                   experiments and data sets from infected human cells indicate this virus is                                                                 unusual because it does not inhibit the NMD pathway that regulates many of our                                                          genes and some of the virus’s genes. Instead, the virus N protein seems to                                                              promote the pathway.

“SARS-CoV-2 reproduces its RNA genome with much higher efficiency than                                                                               other pathogenic human viruses,” Maquat says. “Maybe there is a connection                                                            there; time will tell.”

In the Department of Biology, Fu and Jack Werren, the Nathaniel and                                                                                Helen Wisch Professor of Biology, received expedited funding awards from the                                                            National Science Foundation to apply their expertise in cellular and evolutionary                                                         biology to research proteins involved in infections from COVID-19.                                                                                  The funding was part of the NSF’s Rapid Response Research (RAPID) program                                                               to mobilize funding for high priority projects.

Werren’s research will be important in ameliorating some of the potential side                                                               effects of COVID-19 infections, including blood clots and heart diseases,                                                                                           while Fu’s research will provide insight into the potential effects of viral infection                                                             on human cell metabolism.

“Our research will provide insight into the potential effects of viral infection on                                                                         host cellular processes,” Fu says. “Identifying which cell functions are affected                                                                 by the virus could help lessen some of the negative effects caused by COVID-19.”                                                                  

Douglas Anderson, an assistant professor of medicine in the                                                                                                Aab Cardiovascular Research Institute and a member of the Center for                                                                         RNA Biology, studies how RNA mutations can give rise to human disease                                                                      and has found that alternative therapeutics, such as the gene-editing                                                                 technology CRISPR, may additionally “usher in a new approach to how we                                                                target and combat infectious diseases,” he says.

For the past few years, Anderson’s lab has developed tools and delivery systems                                                                          that use the RNA-targeting CRISPR-Cas13 to treat human genetic diseases that                                                         affect muscle function. CRISPR-Cas13 is like a molecular pair of scissors that                                                               can target specific RNAs for degradation, using small, programmable guide RNAs.

When the health crisis first became apparent in Wuhan, China, researchers in                                                          Anderson’s lab turned their focus toward developing a                                                                                                       CRISPR-Cas13 therapeutic aimed at SARS-CoV-2.                                                                                                                   Applying the knowledge already available about coronavirus RNA replication,                                                                 they designed single CRISPR guide RNAs capable of targeting every                                                                                  viral RNA that is made within a SARS-CoV-2 infected cell. Using a novel                                                                                cloning method developed in Anderson’s lab, multiple CRISPR guide-RNAs                                                                    could be packaged into a single therapeutic vector (a genetically engineered                                                                                    carrier) to target numerous viral RNA sites simultaneously.                                                                                              The multi-pronged targeting strategy could be used as a therapy to                                                                          safeguard against virus-induced cell toxicity and prevent ‘escape’ of viruses                                                                  which may have undergone mutation.

“Infectious viruses and pandemics seemingly come out of nowhere,                                                                             which has made it hard to rapidly develop and screen traditional small molecule                                                       therapeutics or vaccines,” Anderson says.                                                                                                                               “There is a clear need to develop alternative targeted therapeutics,                                                                                                        such as CRISPR-Cas13, which have the ability to be rapidly reprogrammed                                                                         to target new emerging pandemics.”

bat with wings spread.                    While many new treatments for the novel coronavirus are being considered,                                                                  there is one thing that is certain, Maquat says: “Targeting RNA,                                                                                                  or the proteins it produces, is essential for therapeutically combatting this disease.”

What role will RNA play in the future of vaccines                                                                          and disease treatments?

Most people living in the United States today have only read about the                                                                           1918 flu pandemic and the relatively recent RNA viruses,                                                                                                such as Ebola or Zika, that are seen largely in other countries.

“RNA treatments will most likely be a wave of the future for these and other                                                                            emerging diseases,” Maquat says. “Epidemiologists know new infectious                                                                  pathogens are coming given how small the world has become with international                                                           travel, including to and from places where humans and animals are in close                                                             contact.”

Bats, in particular, are reservoirs for viruses.                                                                                                            Many bat species are able to live with viruses without experiencing ill effects,                                                                       given the bats’ unusual physiology.                                                                                                                                          If these bat viruses mutate so they become capable of infecting humans,                                                                         however, there will be new diseases, Maquat says.

“It is just a matter of when this will happen and what the virus will be.                                                                               The hope is that we will be ready and able to develop vaccines against these                                                                     new viruses with the new pipelines that have been put in place for COVID-19.”

This story was originally published on April 28, 2020, and updated on                                                                       December 14, 2020.


Read more

health care worker gives someone a vaccineFDA votes to approve emergency use of Pfizer coronavirus vaccine
Researchers and volunteers in Rochester have been involved in the testing of the Pfizer/BioNTech vaccine since May, and technologies used in the development of the vaccine can trace their origins to decades of infectious disease research conducted at Rochester.

Bats offer clues to treating COVID-19
Bats carry many viruses, including the one behind COVID-19, without becoming ill. University of Rochester biologists are studying the immune system of bats to find potential ways to “mimic” that system in humans.
illustration of cellular enzymeRochester biologists selected for ‘rapid research’ on COVID-19
Rochester biologists are exploring how coronavirus interacts with cellular proteins to cause COVID-19 under a priority NSF program.

 

Tags: ,                                                                        ,                                                                                                   ,                                                                                   

CategoryScience & Technology

Wednesday, December 15, 2021

Overview 

  • On 26 November 2021, the World Health Organization ( W.H.O.) designated the
  •  variant B.1.1.529 a variant of concern (VOC), on the basis of advice from WHO’s 
  • Technical Advisory Group on Virus Evolution. 
  • The variant has been given the name Omicron. 
  • Omicron variant is a highly divergent variant with a high number of mutations, including 
  • 26-32 in the spike protein, some of which are concerning and may be associated with
  •  immune escape potential and higher transmissibility.
  •  However, there are still considerable uncertainties.
  •  As of 9 December 2021, cases of human infections with this variant have been identified
  •  in 63 countries across all six WHO regions.
  •  Current understanding of the Omicron variant from recent data are likely to evolve as 
  • more data becomes available.
  • The overall threat posed by Omicron largely depends on three key questions,
  •  including: (1) how transmissible the variant is; 
  • (2) how well vaccines and prior infection protect against infection, transmission, 
  • clinical disease and death; and (3) how virulent the variant is compared to other variants.                                                      Public health advice is based on current information and will be tailored as more evidence
  •  emerges around those key questions.
  • Based on current limited evidence Omicron appears to have a growth advantage over 
  • Delta. It is spreading faster than the Delta variant in South Africa where Delta circulation 
  • was low, but also appears to spread more quickly than the Delta variant in other countries
  •  where the incidence of Delta is high, such as in the United Kingdom. 
  • Whether Omicron’s observed rapid growth rate in countries with high levels of population 
  • immunity is related to immune evasion, intrinsic increased transmissibility, 
  • or a combination of both remains uncertain. However, given the current available data,
  •  it is likely that Omicron will outpace the Delta variant where community transmission occurs.
  • There are still limited data on the clinical severity of Omicron. 
  • While preliminary findings from South Africa suggest it may be less severe than Delta,
  •  and all cases reported in the EU/EEA to date have been mild or asymptomatic, 
  • it remains unclear to what extent Omicron may be inherently less virulent.
  •  More data are needed to understand the severity profile.
  • There are limited available data, and no peer-reviewed evidence, on vaccine efficacy or
  •  effectiveness to date for Omicron. Preliminary evidence, and the considerably altered 
  • antigenic profile of the Omicron spike protein, suggests a reduction in vaccine efficacy
  •  against infection and transmission associated with Omicron. 
  • There is some preliminary evidence that the incidence of reinfection has increased 
  • in South Africa, which may be associated with humoral (antibody-mediated) immune
  •  evasion. In addition, preliminary evidence from a few studies of limited sample size 
  • have shown that sera obtained from vaccinated and previously infected individuals had 
  • lower neutralization activity (the size of the reduction ranges considerably) than with
  •  any other circulating VOCs of SARS-CoV-2 and the ancestral strain.
  • The diagnostic accuracy of routinely used PCR and antigen-based rapid diagnostic test
  •  (Ag-RDT) assays does not appear to be influenced by Omicron. 
  • Most Omicron variant sequences reported include a deletion in the S gene, causing 
  • some S gene targeting PCR assays to appear negative. 
  • Although some publicly shared sequences lack this deletion, this remains a minority of 
  • currently available sequences, and S gene target failure (SGTF) can therefore be used
  •  as a useful proxy marker of Omicron, for surveillance purposes. 
  • However, confirmation should be obtained by sequencing, as this
  •  deletion can also be found in other VOCs (e.g., Alpha and subsets of Gamma and Delta).
  • Therapeutic interventions for the management of patients with severe or critical
  •  COVID-19 associated with the Omicron variant that target host responses
  •  (such as corticosteroids, and interleukin 6 receptor blockers and prophylaxis with
  •  anticoagulation) are expected to remain effective.  
  • However, monoclonal antibodies will need to be tested individually, for their 
  • antigen binding and virus neutralization and these studies should be prioritized.

    WHO TEAM
    WHO Headquarters (HQ),  WHO Health Emergencies Programme
    EDITORS
    World Health Emergencies Programme
    NUMBER OF PAGES
    2

    Sunday, December 12, 2021

            

    Forward to a Friend

    GLADSTONE-UCSF INSTITUTE OF GENOMIC IMMUNOLOGY

    Innovators in Genomic Immunology

    November 3, 2021
    9am–5pm PDT • Online

    Join us tomorrow to meet the researchers from the Gladstone-UCSF Institute of Genomic Immunology, who are using their diverse expertise in a rapidly advancing field to design and produce tailored immune cell therapies to combat a broad array of diseases.

    REGISTER

    Know someone who’d like to attend? Forward to a friend.

    The Gladstone-UCSF Institute of Genomic Immunology was launched in 2020 to bring experts in diverse, rapidly advancing fields together around the shared goal of understanding how to genetically control human immune cells and using this knowledge to develop innovative cell‑based immunotherapies.

    View in Your Browser

    © 2021 Gladstone Institutes. All rights reserved.

    Want to change how you receive these emails?
    Update your preferences or unsubscribe from this list.

    Gladstone Institutes
    1650 Owens Street
    San Francisco, CA  94158

    Add us to your address book

    Monday, December 6, 2021

     Gentle People:

     I am waiting for the third dose of vaccine to help keep me protected against the Covid 19 virus but this brings up an interesting question...will the third dose be effective against the Covid 19 Omicron variant?  

     I've read how a third dose of vaccine protects up to 80% of people already vaccinated but is this third dose aimed at the original virus or at the new and mutated Omicron virus?

     I have also read that Phizer and Moderna are already creating a vaccine aimed specifically at the Omicron variant but will that be included in the Third dose or will that be a Fourth dose?

     The game of catch-up to the mutating virus leads me back to Leor Weinberger and his colleagues at the U.S. Gladstone Institute. Years ago, Leor talked about playing catch-up with mutating viruses.

    In 1917 Leor claimed to have developed an anti-virus virus that worked against HIV.  On Ted.com, Leor claimed that he was going to try his vaccine first in Africa to see if it worked. HIV is a Corona type virus and if his anti-virus virus works against HIV in Africa, maybe it will work to help eradicate the other Corona viruses in North America and the rest of the world.  I am hoping that Leor and his gang not only catch up but beat the crap out of deadly viruses once and for all!                          N J R

    =================================

    IF IT WORKS AGAINST THE H.I.V. VIRUS, WHY CAN'T "ADAPTIVE TRANSMISSIBLE THERAPIES" BE ADOPTED AND ACTIVATED BY WORLD GOVERNMENTS AND THEIR GIANT PHARMACEUTICAL COMPANIES, TO STOP THE SPREAD OF THE COVID-19 VARIANT PANDEMIC?

    ======================

    ADAPTIVE TRANSMISSIBLE THERAPIES: A NEW CONCEPT FOR DISEASE CONTROL

    Existing measures for infectious disease control face three ‘universal’ barriers:

    (i)  Deployment (e.g. reaching the highest-risk, infectious ‘superspreaders’ who drive disease circulation)

    (ii)  Pathogen persistence & behavioral barriers (e.g. adherence)

    (iii)  Evolution (e.g. resistance and escape)

    These barriers exist because pathogens are dynamic—they mutate and transmit—while existing therapies are static, neither mutating nor transmitting.  To surmount these barriers, we have proposed a radical shift in therapeutic paradigm toward developing adaptive, dynamic therapies (Metzger et al. 2011).  Building off data-driven epidemiological models, we show that engineered molecular parasites, designed to piggyback on HIV-1, could circumvent each barrier and dramatically lower HIV/AIDS in sub-Saharan Africa as compared to established interventions. 

    Above: A representative model for how a small 'core groups' of high-risk 'superspreaders' (e.g. commercial sex workers and their clients) drove the HIV-1 


    Wednesday, November 17, 2021

    Barbed and ready

    It starts with the spikes. Each SARS-CoV-2 virion (virus particle) has an outer surface peppered with 24–40 haphazardly arranged spike proteins that are its key to fusing with human cells2. For other types of virus, such as influenza, external fusion proteins are relatively rigid. SARS-CoV-2 spikes, however, are wildly flexible and hinge at three points, according to work published in August 2020 by biochemist Martin Beck at the Max Planck Institute of Biophysics in Frankfurt, Germany, and his colleagues3.

    That allows the spikes to flop around, sway and rotate, which could make it easier for them to scan the cell surface and for multiple spikes to bind to a human cell. There are no similar experimental data for other coronaviruses, but because spike-protein sequences are highly evolutionarily conserved, it is fair to assume the trait is shared, says Beck.

    Slices through tomographic reconstructions of SARS-CoV-2 virions. Scale bars, 30 nm.

    Cryo-electron tomography images of SARS-CoV-2 virions. (Scale bar: 30 nanometres.)Credit: B. Turoňová et al./Science

    Early in the pandemic, researchers confirmed that the RBDs of SARS-CoV-2 spike proteins attach to a familiar protein called the ACE2 receptor, which adorns the outside of most human throat and lung cells. This receptor is also the docking point for SARS-CoV, the virus that causes severe acute respiratory syndrome (SARS). But compared with SARS-CoV, SARS-CoV-2 binds to ACE2 an estimated 2–4 times more strongly4, because several changes in the RBD stabilize its virus-binding hotspots5.

    Worrying variants of SARS-CoV-2 tend to have mutations in the S1 subunit of the spike protein, which hosts the RBDs and is responsible for binding to the ACE2 receptor. (A second spike subunit, S2, prompts viral fusion with the host cell’s membrane.)

    The Alpha variant, for example, includes ten changes in the spike-protein sequence, which result in RBDs being more likely to stay in the ‘up’ position6. “It is helping the virus along by making it easier to enter into cells,” says Priyamvada Acharya, a structural biologist at the Duke Human Vaccine Institute in Durham, North Carolina, who is studying the spike mutations.

    The Delta variant, which is now spreading around the world, hosts multiple mutations in the S1 subunit, including three in the RBD that seem to improve the RBD’s ability to bind to ACE2 and evade the immune system7.

    Restricted entry

    Once the viral spikes bind to ACE2, other proteins on the host cell’s surface initiate a process that leads to the merging of viral and cell membranes (see ‘Viral entry up close’).

    Viral entry up close: A graphic that shows the interaction between viral spike proteins and host receptors before viral entry.

    Source: Janet Iwasa, Univ. Utah; Graphic: Nik Spencer/Nature

    The virus that causes SARS, SARS-CoV, uses either of two host protease enzymes to break in: TMPRSS2 (pronounced ‘tempress two’) or cathepsin L. TMPRSS2 is the faster route in, but SARS-CoV often enters instead through an endosome — a lipid-surrounded bubble — which relies on cathepsin L. When virions enter cells by this route, however, antiviral proteins can trap them.

    SARS-CoV-2 differs from SARS-CoV because it efficiently uses TMPRSS2, an enzyme found in high amounts on the outside of respiratory cells. First, TMPRSS2 cuts a site on the spike’s S2 subunit8. That cut exposes a run of hydrophobic amino acids that rapidly buries itself in the closest membrane — that of the host cell. Next, the extended spike folds back onto itself, like a zipper, forcing the viral and cell membranes to fuse.

    Animated sequence of the SARS-CoV-2 virus merging its membrane with a cell.

    An animation of the way SARS-CoV-2 fuses with cells.Credit: Janet Iwasa, University of Utah

    The virus then ejects its genome directly into the cell. By invading in this spring-loaded manner, SARS-CoV-2 infects faster than SARS-CoV and avoids being trapped in endosomes, according to work published in April by Barclay and her colleagues at Imperial College London9.

    The virus’s speedy entry using TMPRSS2 explains why the malaria drug chloroquine didn’t work in clinical trials as a COVID-19 treatment, despite early promising studies in the lab10. Those turned out to have used cells that rely exclusively on cathepsins for endosomal entry. “When the virus transmits and replicates in the human airway, it doesn’t use endosomes, so chloroquine, which is an endosomal disrupting drug, is not effective in real life,” says Barclay.

    The discovery also points to protease inhibitors as a promising therapeutic option to prevent a virus from using TMPRSS2, cathepsin L or other proteases to enter host cells. One TMPRSS2 inhibitor, camostat mesylate, which is approved in Japan to treat pancreatitis, blocked viral entry into lung cells8, but the drug did not improve patients’ outcomes in an initial clinical trial11.

    “From my perspective, we should have such protease inhibitors as broad antivirals available to fight new disease outbreaks and prevent future pandemics at the very beginning,” says Stefan Pöhlmann, director of the Infection Biology Unit at the German Primate Center in Göttingen, who has led research on ACE2 binding and the TMPRSS2 pathway.

    A hidden spike: A graphic showing the structure of the SARS-CoV-2 spike protein and its coating of glycans.

    Source: Structural image from Lorenzo Casalino, Univ. California, San Diego (Ref. 1); Graphic: Nik Spencer/Nature

    In Amaro’s simulation, when the RBD lifted up above the glycan cloud, two glycans swooped in to lock it into place, like a kickstand on a bicycle. When Amaro mutated the glycans in the computer model, the RBD collapsed. McLellan’s team built a way to try the same experiment in the lab, and by June 2020, the collaborators had reported that mutating the two glycans reduced the ability of the spike protein to bind to a human cell receptor1 — a role that no one has previously recognized in coronaviruses, McLellan says. It’s possible that snipping out those two sugars could reduce the virus’s infectivity, says Amaro, although researchers don’t yet have a way to do this.

    Since the start of the COVID-19 pandemic, scientists have been developing a detailed understanding of how SARS-CoV-2 infects cells. By picking apart the infection process, they hope to find better ways to interrupt it through improved treatments and vaccines, and learn why the latest strains, such as the Delta variant, are more transmissible.

    What has emerged from 19 months of work, backed by decades of coronavirus research, is a blow-by-blow account of how SARS-CoV-2 invades human cells (see ‘Life cycle of the pandemic coronavirus’). Scientists have discovered key adaptations that help the virus to grab on to human cells with surprising strength and then hide itself once inside. Later, as it leaves cells, SARS-CoV-2 executes a crucial processing step to prepare its particles for infecting even more human cells. These are some of the tools that have enabled the virus to spread so quickly and claim millions of lives. “That’s why it’s so difficult to control,” says Wendy Barclay, a virologist at Imperial College London.

    Life cycle of the pandemic coronavirus: Infographic showing how the virus enters, adapts and exits from host cells.

    Source: Hui (Ann) Liu, Univ. Utah; Graphic: Nik Spencer/Nature 


    • NEWS FEATURE

    How the coronavirus infects cells — and why Delta is so dangerous

    Animated sequence of the SARS-CoV-2 virus.

    A computer simulation of the structure of the coronavirus SARS-CoV-2.Credit: Janet Iwasa, University of Utah

    The coronavirus sports a luxurious sugar coat. “It’s striking,” thought Rommie Amaro, staring at her computer simulation of one of the trademark spike proteins of SARS-CoV-2, which stick out from the virus’s surface. It was swathed in sugar molecules, known as glycans.

    “When you see it with all the glycans, it’s almost unrecognizable,” says Amaro, a computational biophysical chemist at the University of California, San Diego.

    Many viruses have glycans covering their outer proteins, camouflaging them from the human immune system like a wolf in sheep’s clothing. But last year, Amaro’s laboratory group and collaborators created the most detailed visualization yet of this coat, based on structural and genetic data and rendered atom-by-atom by a supercomputer. On 22 March 2020, she posted the simulation to Twitter. Within an hour, one researcher asked in a comment: what was the naked, uncoated loop sticking out of the top of the protein?

    Amaro had no idea. But ten minutes later, structural biologist Jason McLellan at the University of Texas at Austin chimed in: the uncoated loop was a receptor binding domain (RBD), one of three sections of the spike that bind to receptors on human cells (see ‘A hidden spike’).


      Hello my good friend Valdemar Oliveira! I am happy to hear you had a successfull heart operation.  I hope you live to be 110. I may not be...