Watching coronavirus multiply – the quest for structures of SARS-CoV-2 RNA polymerase

April 15, 2021

A guest entry by Hauke Hillen

In order for the novel coronavirus SARS-CoV-2 to replicate, it has to achieve two basic tasks: It needs to make copies of its genome that can be packaged into new virus particles, and it needs to activate viral genes to produce the proteins that actually form new virus particles, such as spike or nucleocapsid. Both tasks are carried out by a specialized molecular copying machine called the replication and transcription complex, or short RTC. The RTC is made up of a number of viral non-structural proteins (nsps) which act together to produce copies of the viral RNA genome. Some of the RTC components have been discussed in previous posts, for example the exonuclease nsp14, which can correct errors that occur during RNA copying (this is called proofreading),or the methyltransferases nsp14 and nsp16 that add chemical modifications to the RNA that help stabilize and hide it from the immune system (this is called capping).

In this post, we will have a closer look at the enzyme that carries out RNA replication, the RNA-dependent RNA polymerase (RdRp) nsp12, and how it interacts with the other components to form the RTC.

RNA polymera…what?

First off, let’s briefly discuss what a “RNA-dependent RNA polymerase” is, and why it is important for the virus. Polymerases are enzymes found in every living cell carrying out one of the most fundamental tasks in biology: they replicate genetic information. While most cells store their genetic information in form of DNA (deoxyribonucleic acid) and use RNA (ribonucleic acid) only as transient messenger molecules (mRNAs), some viruses rely on RNA for both information storage and transmission and are hence called “RNA viruses”. To replicate their genetic information, they need a polymerase that uses RNA as template to copy the encoded information into a new RNA molecule – a RNA-dependent RNA polymerase. Chemically speaking, RNA is a polymer (a long string of nearly-identical individual building blocks) composed of the four nucleotides adenosine (A), guanosine (G), cytosine (C) and uracil (U), the sequence of which defines the genetic information.

The job of the RNA polymerase is to read the sequence of nucleotides in a template RNA and synthesize new RNA with the same sequence (technically with a complementary sequence) using individual nucleotides as building blocks. To do this, the RNA polymerase has to achieve three basic steps. First, it needs to bind the template RNA. Second, it has to read the sequence of nucleotides in the RNA. Third, it has to incorporate the correct matching nucleotide building blocks to polymerize a new RNA strand.

Coronaviruses are RNA viruses and therefore have an RNA-dependent RNA polymerase (RdRp, or nsp12). However, they are exceptional in several ways. First, their RNA genomes are almost 30.000 nucleotides in length, which are the largest RNA virus genomes known to date. Second, their RNA polymerase nsp12 requires the additional proteins nsp7 and nsp8 in order to form the active “core” RdRp. Third, this core RdRp assembles with further viral proteins to form the RTC, which can carry out additional functions such as proofreading to remove copying errors - a highly unusual capability for RNA viruses.

Since the RNA polymerase has such a fundamental job during virus replication, it is an attractive drug target to combat viral infections. Indeed, many successful anti-viral drugs against Hepatitis C virus, HIV or Herpes virus act by inhibiting viral polymerases. Strikingly, viral RdRp enzymes are remarkably similar in their overall structure even between unrelated viruses, indicating that they share a common evolutionary ancestor and their function is so essential that it does not allow for drastic changes. Therefore, some known anti-virals developed to treat other viral diseases have also been tested for their activity against the polymerase of SARS-CoV-2 and even approved for clinical use against COVID-19 [1]. However, these repurposed compounds are generally not as effective as many had hoped, because even rather subtle structural differences between the polymerase enzymes of different viruses can have strong effects on the action of anti-viral drugs. Thus, more specific and efficient drugs against SARS-CoV-2 are therefore badly needed. In order to discover and develop such compounds, detailed knowledge of the structure and function of the RdRp is necessary.

Structure of the coronavirus RNA-dependent RNA polymerase (RdRp)

The first structure of a coronavirus polymerase was determined shortly before the outbreak of the current COVID-19 pandemic when Kirchdoerfer and Ward reported the cryo-electron microscopy (cryo-EM) structure of SARS-CoV-1 RdRp [2]. Since SARS-CoV-2 has emerged, scientists all over the world have been racing to determine the structures of its RdRp. As of February 2021, this has led to more than 20 structures of SARS-CoV-2 polymerase-complexes published in the PDB, and often several groups of scientists reported similar structures around the same time.

These structures show that the RNA polymerase nsp12 resembles a right hand with individual domains called palm, fingers and thumb (Figure 1) [3–7]. This “hand” shape is typical for viral RNA polymerases and holds a tight grip on the double-stranded RNA helix that forms between template and product strand during RNA synthesis. Within the palm lies the “active center” of the enzyme, where nucleotides are added to the growing product chain. The active center is accessible from the surface of the enzyme through a special tunnel, so that nucleotides can enter the substrate-binding site. As the template strand is opposite to the substrate-binding site, each nucleotide entering is “sampled” for whether it can form base-pairing interactions with the template base. If this is the case, the nucleotide remains bound, and it is added to the 3’ end of the product strand by forming a chemical bond. After that, the RdRp enzyme must slide ahead on the template strand by one nucleotide, which moves the newly produced 3’ end of the product RNA from the substrate-binding site (which is sometimes also referred as position +1) to the position where the previous 3’ end of the product was located prior to addition (position -1). This “translocation” completes the nucleotide addition cycle, as it positions the next templating base and frees up the substrate-binding site for the next matching building block.

Watching coronavirus multiply – the quest for structures of SARS-CoV-2 RNA polymerase 1
Figure 1 – Structure of SARS-CoV-2 RdRp

Left: Cryo-EM structure of SARS-CoV-2 RdRp (PDB 6YYT). Right: Enlarged active site with template strand in blue and nascent chain in red..

In addition to its polymerase domain, RdRp also contains a part that is only found in nidoviruses (the virus family that coronaviruses belong to) called “nidovirus RdRp-associated nucleotidyltransferase domain”, or short NiRAN-domain. Scientists believe that this domain has the capability to transfer nucleotidyl-residues, which means that it can form chemical bonds between nucleotides and other molecules. This hints that it may be involved in modification of the RNA (so-called “capping”) or in helping the enzyme initially kickstart RNA synthesis, but its precise role during coronavirus replication is still being studied by scientists.

In order to efficiently copy RNA, nsp12 requires two additional viral proteins, nsp7 and nsp8. The structures of coronavirus RdRp show that two molecules of nsp8 and one molecule of nsp7 bind on top of the hand-shaped nsp12. Interestingly, even though identical in amino acid sequence, the two nsp8 molecules adopt slightly different shapes. While one of them interacts with the finger domain of nsp12 directly, the interaction of the other one is mediated by nsp7. Both nsp8 molecules have long “arms” that protrude away from the polymerase and touch the RNA duplex as it emerges from the polymerase during replication. These “sliding poles” are unique to coronaviruses and most likely stabilize the RdRp on the RNA, which may help to make sure it doesn’t fall off during replication of the very large genome.

Seeing is believing - visualizing how anti-viral compounds block RdRp

So how exactly can this structural knowledge help to find new drugs against COVID-19? In many ways, enzymes are like tiny molecular machines. By studying their structure, one can analyze in detail how they work biochemically, and this in turn allows us to come up with ways to block their function. Most known anti-viral drugs that target RNA polymerases are so-called nucleoside analogs, which means they are molecules that structurally resemble the natural building blocks of RNA. These compounds can “trick” the RdRp by binding to the active site, but due to their chemical nature, they either cannot be incorporated into the product or lead to mutations that end the viral life cycle. The structures of SARS-CoV-2 RdRp reveal the exact architecture of the active site and show how the chemical environment that the enzyme creates around the product RNA and the substrate nucleotides, facilitates polymerization (Figure 1). This knowledge can help to rationally design or improve compounds in such a way that they bind more efficiently.

This is exemplified by recent studies analyzing how repurposed anti-virals inhibit SARS-CoV-2 RdRp. One such drug is Remdesivir, a compound originally developed against Ebola and other viruses that has also been approved by the FDA and European agencies for treatment of COVID-19 (see also this previous post). Remdesivir chemically resembles adenosine triphosphate (ATP), but has an additional bulky chemical residue called a cyano-group attached to the C1-atom of its ribose moiety. In contrast to most nucleoside analogs, Remdesivir does not block the RdRp immediately, but only after another three nucleotides are added, a process called “delayed stalling” [8–10]. Initial structures of SARS-CoV-2 RdRp in the presence of Remdesivir showed how it can act as an adenosine analog and how it can be incorporated at the 3’ end of the nascent product RNA strand and translocated to the -1 position (Figure 2a,b) [5,6]. However, this could not explain how it would lead to inhibition of RNA synthesis.

To pinpoint why Remdesivir interferes with RNA synthesis exactly after three subsequent nucleotides are added, the authors of a recent study used a combination of synthetic chemistry and structural biology [11]. They systematically determined structures of SARS-CoV-2 RdRp bound to a template-product RNA duplex which contained Remdesivir and either two or three additional nucleotides at the 3’ end. In the first case, the structure showed that the RdRp was in the post-translocated state, with Remdesivir at position -3 and  an empty substrate binding site, as expected (Figure 2c). In contrast, the structure of the RdRp with an RNA containing Remdesivir and three additional nucleotides was not in the post-translocated state and Remdesivir was not located at position -4. Instead, it remained at position -3, and the third additional nucleotide at the 3’ end of the product was stuck in the substrate-binding site (Figure 2d). This state resembles the situation directly after addition of a new nucleotide but before translocation and is hence called the pre-translocated state. This suggests that remdesivir inhibits the SARS-CoV-2 RNA polymerase by posing a translocation barrier, and the structures provide a molecular and chemical explanation for this: Initially, Remdesivir can be added to the growing RNA just like adenosine triphosphate and also translocated to add another two nucleotides. However, after binding and addition of a third nucleotide, Remdesivir can not be translocated to the -4 position, because its bulky cyano group would clash with a serine residue (Ser861) in the thumb domain of nsp12 (Figure 2c). Therefore, the polymerase gets stuck in the pre-translocated state, which explains why exactly three nucleotides can be added after Remdesivir incorporation – addition of a fourth nucleotide would first require translocation. This proposed mechanism is in agreement with previous modelling [5,10] and was shortly after independently confirmed by another structural study, in which the authors managed to trap an identical pre-translocated, stalled intermediate with Remdesivir in position -3 [12].

Watching coronavirus multiply – the quest for structures of SARS-CoV-2 RNA polymerase 2
Figure 2 – How remdesivir inhibits SARS-CoV-2 RdRp

Structural snapshots of remdesivir (purple) moving through the active site of SARS-CoV-2 RdRp. When it reaches the third position after its incorporation to the RNA (-3), its further movement is blocked because the cyano group would bump into Ser861. A) Remdesivir at position +1 (PDB 7BV2) B) Remdesivir at position -1 PDB 7C2K C) Remdesivir at position -3 (PDB 7B3B) D) Remdesivir at position -3 with the nucleotide at the 3’ end stuck in the substrate binding site (PDB 7B3C).

This mechanism also suggests how Remdesivir may at least partially escape the coronavirus proofreading enzyme nsp14, which removes misincorporated nucleotides at the 3’ end of the RNA and thus counteracts anti-virals that target RdRp. Since Remdesivir can be translocated until it reaches position -3 before it causes stalling, it may leave the active site of the enzyme before it can be recognized by the proofreading machinery.

Importantly, these studies also provide clues as to why Remdesivir has had limited success in fighting COVID-19. The structures show that the steric block between Ser861 and the cyano group of Remdesivir is not severe and can therefore be overcome by the enzyme, for example at high concentrations of substrate NTPs [8] Consistent with this, substitution of Ser861 with residues that clash even less (Alanine or Glycine) make the RdRp less sensitive or even resistant to Remdesivir [5,13]. This suggests that the translocation barrier could potentially be enhanced by a compound that leads to more severe clashes. One way to achieve this could be to modify Remdesivir to contain more bulky chemical moieties than the cyano group. Thus, the detailed molecular insights into the mechanism of Remdesivir also provide a rational basis for designing more potent anti-virals and test their effect on the SARS-CoV-2 RdRp.

Similar structure-function studies are now being undertaken also for other promising anti-viral compounds, such as Favipiravir. Like Remdesivir, it is a nucleoside analogue that was initially developed against other viruses, but showed some promising results against SARS-CoV-2. Structures of the SARS-CoV-2 RNA polymerase with Favipiravir show how it mimics both guanosine or adenosine in the active site of the enzyme by forming unusual base-pairing interactions with cytosine and uracil, respectively, and this leads to errors during RNA copying that eventually kill the virus [14,15]. Another study recently reported the structure of Suramin bound to SARS-CoV-2 RdRp [16]. In contrast to Remdesivir and Favipiravir, this compound is not a nucleoside analog and hence does not get incorporated into the RNA. Instead, two Suramin molecules can apparently bind to the RdRp and thereby prevent its association with template and product RNA, rendering it inactive.

These studies are good examples of how structural biology can visualize complicated chemical reactions in an intuitive way. Based on these results, drugs like Remdesivir or Favipiravir can be rationally optimized to more effectively combat COVID-19 and may cause less side effects.

Dissecting the SARS-CoV-2 RTC structure by structure

In addition to provide detailed snapshots of how anti-viral compounds act to inhibit SARS-CoV-2 RdRp, structural studies are also helping scientists to understand how the unique coronavirus RTC combines different functions such as RNA synthesis, proofreading and modification. After the structure of the “core” SARS-CoV-2 RdRp was determined within a few months after the outbreak of COVID-19, scientists quickly moved to studying how additional non-structural proteins bind to it to form the RTC. One of these is nsp13, which belongs to a protein class called “helicases”. These are enzymes that bind to DNA or RNA and, with the help of chemical energy in the form of ATP, move along them or unwind helices. Cryo-EM structures of nsp13 bound to the SARS-CoV-2 RdRp show that two molecules of nsp13 can bind to the RdRp, and suggest that it may allow the polymerase to move backwards on the RNA (Figure 3) [17,18]. This finding seems unexpected at first, but experts think this may be required for the proofreading enzyme nsp14 to remove errors that the polymerase makes during copying or to produce the mRNAs for certain viral proteins. Another recent cryo-EM structure shows that the small protein nsp9 interacts with the NiRAN domain in the SARS-CoV-2 polymerase [19]. In the accompanying paper, the authors suggest that the NiRAN domain may be involved in RNA capping, and nsp9 seems to block its activity. However, others have proposed that nsp9 is in fact a substrate of nucleotidylation by the NiRAN domain, and that it may be involved in priming the RNA polymerase for initial RNA synthesis [20]. Thus, further studies are necessary to determine whether the NiRAN is involved in capping, priming or even both.

Watching coronavirus multiply – the quest for structures of SARS-CoV-2 RNA polymerase 3
Figure 3 – Structures help uncover the roles of the different RTC components

Structure of the SARS-CoV-2 RdRp complex with nsp13 (salmon) and nsp9 (cyan) (PDB 7CYQ).

What’s next?

Over the past year, scientists have uncovered the structure of the SARS-CoV-2 RdRp and associated proteins at a record-breaking pace. While these structures provide impressive first glimpses at RdRp-complexes, researchers are already working to determine how the remaining nsp proteins interact with the RdRp to form the complete RTC complex. This may ultimately aid the quest to find new treatment options for COVID-19 that not only target the polymerase itself, but also proofreading or RNA capping. Such drugs are not only desperately needed for the current pandemic, but may also prove useful for future emerging coronaviruses, because the RNA polymerase is typically very similar even between different virus strains.

1. Ledford H: Hopes rise for coronavirus drug remdesivir. Nature 2020, doi:10.1038/d41586-020-01295-8.

2. Kirchdoerfer RN, Ward AB: Structure of the SARS-CoV nsp12 polymerase bound to nsp7 and nsp8 co-factors. Nat Commun 2019, 10:2342.

3. Gao Y, Yan L, Huang Y, Liu F, Zhao Y, Cao L, Wang T, Sun Q, Ming Z, Zhang L, et al.: Structure of the RNA-dependent RNA polymerase from COVID-19 virus. Science 2020, 368:779–782.

4. Hillen HS, Kokic G, Farnung L, Dienemann C, Tegunov D, Cramer P: Structure of replicating SARS-CoV-2 polymerase. Nature 2020, 584:154–156.

5. Wang Q, Wu J, Wang H, Gao Y, Liu Q, Mu A, Ji W, Yan L, Zhu Y, Zhu C, et al.: Structural Basis for RNA Replication by the SARS-CoV-2 Polymerase. Cell 2020, 182:417-428.e13.

6. Yin W, Mao C, Luan X, Shen D-D, Shen Q, Su H, Wang X, Zhou F, Zhao W, Gao M, et al.: Structural basis for inhibition of the RNA-dependent RNA polymerase from SARS-CoV-2 by remdesivir. Science 2020, 368:1499–1504.

7. Peng Q, Peng R, Yuan B, Zhao J, Wang M, Wang X, Wang Q, Sun Y, Fan Z, Qi J, et al.: Structural and Biochemical Characterization of the nsp12-nsp7-nsp8 Core Polymerase Complex from SARS-CoV-2. Cell Reports 2020, 31:107774.

8. Gordon CJ, Tchesnokov EP, Woolner E, Perry JK, Feng JY, Porter DP, Götte M: Remdesivir is a direct-acting antiviral that inhibits RNA-dependent RNA polymerase from severe acute respiratory syndrome coronavirus 2 with high potency. J Biol Chem 2020, 295:6785–6797.

9. Tchesnokov EP, Feng JY, Porter DP, Götte M: Mechanism of Inhibition of Ebola Virus RNA-Dependent RNA Polymerase by Remdesivir. Viruses 2019, 11:326.

10. Gordon CJ, Tchesnokov EP, Feng JY, Porter DP, Gotte M: The antiviral compound remdesivir potently inhibits RNA-dependent RNA polymerase from Middle East respiratory syndrome coronavirus. The Journal of biological chemistry 2020, doi:10.1074/jbc.ac120.013056.

11. Kokic G, Hillen HS, Tegunov D, Dienemann C, Seitz F, Schmitzova J, Farnung L, Siewert A, Höbartner C, Cramer P: Mechanism of SARS-CoV-2 polymerase stalling by remdesivir. Nat Commun 2021, 12:279.

12. Bravo JPK, Dangerfield TL, Taylor DW, Johnson KA: Remdesivir is a delayed translocation inhibitor of SARS CoV-2 replication in vitro. Biorxiv 2020, doi:10.1101/2020.12.14.422718.

13. Tchesnokov EP, Gordon CJ, Woolner E, Kocinkova D, Perry JK, Feng JY, Porter DP, Götte M: Template-dependent inhibition of coronavirus RNA-dependent RNA polymerase by remdesivir reveals a second mechanism of action. J Biol Chem 2020, 295:16156–16165.

14. Naydenova K, Muir KW, Wu L-F, Zhang Z, Coscia F, Peet MJ, Castro-Hartmann P, Qian P, Sader K, Dent K, et al.: Structural basis for the inhibition of the SARS-CoV-2 RNA-dependent RNA polymerase by favipiravir-RTP. Biorxiv 2020, doi:10.1101/2020.10.21.347690.

15. Peng Q, Peng R, Yuan B, Wang M, Zhao J, Fu L, Qi J, Shi Y: Structural basis of SARS-CoV-2 polymerase inhibition by Favipiravir. Innovation 2021, doi:10.1016/j.xinn.2021.100080.

16. Yin W, Luan X, Li Z, Zhou Z, Wang Q, Gao M, Wang X, Zhou F, Shi J, You E, et al.: Structural basis for inhibition of the SARS-CoV-2 RNA polymerase by suramin. Nat Struct Mol Biol 2021, 28:319–325.

17. Chen J, Malone B, Llewellyn E, Grasso M, Shelton PMM, Olinares PDB, Maruthi K, Eng ET, Vatandaslar H, Chait BT, et al.: Structural basis for helicase-polymerase coupling in the SARS-CoV-2 replication-transcription complex. Cell 2020, doi:10.1016/j.cell.2020.07.033.

18. Yan L, Zhang Y, Ge J, Zheng L, Gao Y, Wang T, Jia Z, Wang H, Huang Y, Li M, et al.: Architecture of a SARS-CoV-2 mini replication and transcription complex. Nat Commun 2020, 11:5874.

19. Yan L, Ge J, Zheng L, Zhang Y, Gao Y, Wang T, Huang Y, Yang Y, Gao S, Li M, et al.: Cryo-EM Structure of an Extended SARS-CoV-2 Replication and Transcription Complex Reveals an Intermediate State in Cap Synthesis. Cell 2021, 184:184-193.e10.

20. Slanina H, Madhugiri R, Bylapudi G, Schultheiß K, Karl N, Gulyaeva A, Gorbalenya AE, Linne U, Ziebuhr J: Coronavirus replication–transcription complex: Vital and selective NMPylation of a conserved site in nsp9 by the NiRAN-RdRp subunit. Proc National Acad Sci 2021, 118:e2022310118.

Corinna the Corona Cactus

@
Corinna works as an outreach person for all plant-related business and as a mascot. She gathered previous experience in the garden center, and even though she can be a bit spiky, she likes to cuddle and lie in the sun.
More about this author

Helen Ginn

Senior Research Scientist @ Diamond Light Source, Oxfordshire, UK
Dr Helen Ginn is a senior research scientist at Diamond Light Source in the UK and a computational methods developer in structural biology. She is currently working on Representation of Protein Entities (RoPE) for structural biologists to interpret subtle conformational changes in dynamic protein systems. She has developed Vagabond for torsion angle-driven model refinement and […]
More about this author

Nick Pearce

Assistant Professor @ SciLifeLab DDLS Fellow
Nick obtained his undergraduate degree in Physics from the University of Oxford in 2012, and then his PhD in Systems Approaches to Biomedical Sciences in 2016. He moved to Utrecht in the Netherlands in 2017 to work with Piet Gros, where he obtained an EMBO long-term fellowship and worked on analysing disorder in macromolecular structures. […]
More about this author

Mathias Schmidt

Molecular Life Sciences M.Sc. Student @ Hamburg University
Mathias is currently doing his Master's degree in Molecular Life Sciences at the University of Hamburg and has been an auxiliary scientist in the Corona Structural Taskforce since March 2022. There he is working on the question of the origin of SARS-CoV-2. His undergraduate research focuses on the development of synthetic molecular mechanisms to regulate […]
More about this author

David Briggs

Principal Laboratory Research Scientist @ Francis Crick Institute in London, UK
David Briggs is a Principal Laboratory Research Scientist in the Signalling and Structural Biology lab at the Francis Crick Institute in London, UK. A crystallographer by training, his work focuses on the biophysical and structural characterisation of human extracellular proteins involved in the synapse, which have important ramifications in both psychiatric and neurodegenerative disorders. He […]
More about this author

Lisa Schmidt

Web Developer and Illustrator @ Mullana
Lisa Schmidt is a freelance illustrator who studied Multimedia and Communication (BA) in Ansbach, Germany. Her work is focused on visualising topics around science and technology. She joined the Coronavirus Structural Task Force as media designer, where she does web design, 3D rendering for scientific illustrations and outreach work.
More about this author

Philip Wehling

Nanosciences M.Sc. Student @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Philip has long had an enthusiasm for biological processes which is paired with an analytical understanding of the world. After having worked for a long time as a registered nurse in various fields, he first studied mathematics and finally nanosciences. During a lecture series in preparation for a bachelor's thesis, he came into contact with […]
More about this author

Binisha Karki

Postdoctoral Research Associate @ BioNTech SE
Binisha works as a research associate at BioNTech where she works on the development of COVID-19 vaccine and cancer immunotherapies. She graduated as a Molecular Biology major from Southeastern Louisiana University in May 2019. Post-graduation she worked as a research technician in the Chodera Lab performing biophysical measurements of model protein-ligand systems for computational chemistry […]
More about this author

Binisha Karki

Wissenschaftliche Mitarbeiterin @ BioNTech SE
Binisha ist als wissenschaftliche Mitarbeiterin bei BioNTech angestellt und arbeitet an der Entwicklung von Impfstoffen gegen COVID-19 sowie Krebsimmuntherapien. Sie beendete ihr Studium der Molekularbiologie an der Southeastern Louisiana University im Mai 2019. Anschließend arbeitete sie als Forschungstechnikerin im Chodera-Lab, wo sie biophysikalische Messungen an Modellen von Protein-Liganden-Systeme für computerchemische Benchmarks durchführte.
More about this author

Hauke Hillen

Assistant Professor at the University Medical Center Göttingen & Group Leader at the MPI for Biophysical Chemistry @ University Medical Center Göttingen
Hauke ist Biochemiker und Strukturbiologe. Mit seinem Forschungsteam untersucht er mittels Röntgenkristallografie und Kryo-Elektronenmikroskopie die Struktur und Funktion von molekularen Maschinen, die für die Genexpression in eukaryotischen Zellen verantwortlich sind. Er interessiert sich dabei besonders dafür wie genetisches Material außerhalb des Zellkerns exprimiert wird, zum Beispiel in menschlichen Mitochondrien oder durch Viren im Zytoplasma.
More about this author

Richardson Lab

Richardson Lab @ Duke University, Durham, North Carolina, USA
The long-term goal of the Richardson lab is to contribute to a deeper understanding of the 3D structures of proteins and RNA, including their description, determinants, folding, evolution, and control. Their approaches include structural bioinformatics, macromolecular crystallography, molecular graphics, analysis of structures, and methods development, currently focussed on the improvement of structural accuracy. In this […]
More about this author

Holger Theymann

Agile Leadership Coach @ mehr-Freu.de GmbH
Holger keeps websites running. He makes data from scientific databases appear in nice tables. He also has an eye on keeping the sites fast, safe and reliable. His experience as a software developer, systems architect, agile project manager and coach enabled the Task Force to get the whole process well organized and he even taught […]
More about this author

Florens Fischer

Biology M.Sc. Student @ Rudolf Virchow Center, Würzburg University
Florens is studying biology (M.Sc.) and worked in the Task Force as a student assistant. He has focused on bioinformatics and supports the work on automation of scripts and structuralization of big data with machine learning. He also supported the team in other areas, such as scientific research.
More about this author

Ezika Joshua Onyeka

Public Health M.Sc. student @ Hamburg University of Applied Sciences
Joshua joined Thorn Lab as a student assistant. He is a Public Health practitioner, holds a bachelor's degree in Public Health and is currently enrolled at Hamburg University of Applied Sciences for his MPH. He has helped in implementing some vaccination programmes to improve immunisation coverage and training of immunisation frontline health workers. For the […]
More about this author

Katharina Hoffmann

Molecular Biology M.Sc. student @ Institut für Nanostruktur und Festkörperphysik, Universität Hamburg
Katharina worked as a student assistant at Thorn Lab. Normally, she studies molecular biology at the University of Hamburg. In her master's thesis, which was put on hold by Corona, she is working on the interruption of bacterial communication. Since the lockdown, she has been digging around in databases and analyzing sequences. She never thought […]
More about this author

Nicole Dörfel

Media Designer @
Nicole Dörfel ensures that we and our work are looking good! She is the illustrator, media designer and the artistic soul of the Task Force. She works her magic both in print and digitally—her focus is general media design. In the Task Force, she is mainly responsible for graphics, photo editing, design of all our […]
More about this author

Pairoh Seeliger

Administration Assistant @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Pairoh Seeliger is the admin wizard of the Task Force. She takes care of media requests, handles any logistical issues that come up and makes sure our science doesn’t sound too complicated in our German outreach efforts. She self-describes as "a jack of all trades with a University education in German studies and business administration, […]
More about this author

Oliver Kippes

Biochemistry B.Sc. Student @ Rudolf Virchow Center, Würzburg University
Oli is studying biochemistry (B.Sc) and has completed a training as an IT specialist prior to his studies. With the combined knowledge of his studies and training, he helps maintaining the structural database, programs applications for it and supports the team in literature research. In spite of his study, structural biology was still a new […]
More about this author

Luise Kandler

Biochemistry B.Sc. Student @ Rudolf-Virchow Center, Würzburg University
Luise is a B.Sc. student in biochemistry at the University of Würzburg and joined the Task Force during the first Corona lockdown. She did her bachelor's thesis with the Thorn Lab, where she learned programming with Python and worked on the implementation of a GUI for our machine learning tool HARUSPEX in Coot. In the […]
More about this author

Ferdinand Kirsten

Biochemistry B.Sc. Student @ Rudolf Virchow Center, Würzburg University
Ferdinand did his bachelor's thesis at Thorn Lab on solvent exchange and interactions in macromolecular crystallography. Still new to the world of crystallography and structural refinement, he tries to help wherever he can, with a main focus on literature and genome research as well as structural refinement with Coot. Even if he's more of the […]
More about this author

Kristopher Nolte

Biochemistry B.Sc. Student @ Rudolf-Virchow Center, Würzburg University
Kristopher joined Thorn Lab as part of his bachelor thesis. In this thesis he refined aspects of the diagnostic tool for graphical X-Ray data analysis (AUSPEX) with the help of machine learning. But since the corona crisis halted all our lives, he contributes to the Task Force by using his knowledge of bioinformatics and programming […]
More about this author

Erik Nebelung

Nanoscience M.Sc. Student @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Erik is studying nanoscience with a focus on biochemical methods and applications. From August 2020 till January 2021 he pursued his studies at the iNano institute in Aarhus, before starting his master's thesis back in Hamburg. He had his first taste of protein crystallization during his bachelor's thesis work and this sparked his interest in […]
More about this author

Toyin Akinselure

Nanoscience M.Sc. Student @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Toyin ist a microbiologist and presently an M.Sc. student in nanoscience with a focus on nanobiology and nanochemistry. She is interested in scientific research especially in protein chemistry and drug discovery. In the previous autumn and winter, she interned with two research projects, one in drug discovery and the other in protein structure. She found […]
More about this author

Lea von Soosten

Physics M.Sc. Student @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Lea is a M.Sc. physics student with a great interest in everything related to biology. Even though she comes from a different field, she joined the team to expand her knowledge in biochemistry and help the Task Force with a main focus on literature research. Also, she loves drawing!
More about this author

Sabrina Stäb

Biotechnology M.Sc. Student @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Sabrina is studying biochemistry (M.Sc.) and works as a research assistant for the Thorn Lab and the CSTF. During her bachelor thesis on "Crystallization and Structure Solution of High-Quality Structures for MAD Experiments", she was able to gain a lot of experience in the field of crystallography and now brings this experience to the project. […]
More about this author

Alexander Matthew Payne

Chemical Biology Ph.D. Student @ Chodera Lab, Memorial Sloan Kettering Center for Cancer Research, New York, U.S.
Alex is a Ph.D. student interested in understanding how proteins move! He has recently joined the labs of John Chodera and Richard Hite to work on a joint project involving molecular dynamics and Cryo-EM. His goal is to generate conformational ensembles from Cryo-EM data and simulate the ensemble using massive scale molecular dynamics via Folding@Home. […]
More about this author

Maximilian Edich

Bioinformatics Ph.D. Student @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Max studied bioinformatics and genome research in Bielefeld and joined the CSTF as a Ph.D. student in 2021. Previously, his focus was on molecular modeling. Now, he works on the so-called R-factor gap. He already learned what it is like to be part of a young, scientific team as a member of the iGEM contest […]
More about this author

Agnel Praveen Joseph

Computational Scientist @ Science and Technology Facilities Council, UK
Dr. Agnel Praveen works as a computational scientist in the CCP-EM team at the Science and Technology Facilities Council, UK. He is interested in approaches to interpret and validate maps and atomic models derived from Cryo-EM data and looks also into computational methods for the interpretation of Cryo-ET data. In collaboration with five other sites […]
More about this author

Dale Tronrud

Research Scientist @
Dale Tronrud has both solved protein crystal structures and developed methods and software for the optimization of macromolecular models against X-ray data and known chemical structural information. He has had a long-standing interest in enzyme:inhibitor complexes and photosynthetic proteins, focusing on the Fenna-Matthews-Olson protein. In addition, he has also been involved in the validation and […]
More about this author

Sam Horrell

Beamline Scientist @ Diamond Light Source, Oxfordshire, UK
Sam is a structural biologist working on method development around structural biology at Diamond Light Source, in particular for ways of better understanding how enzymes function through the production of structural movies. Sam is working through deposited structures related to SARS-CoV and SARS-CoV-2 with a view to providing the most accurate protein structures possible for […]
More about this author

Cameron Fyfe

Postdoctoral Research Associate @ Micalis Institute, INRAE, Paris, France
Cameron is a structural biologist who has worked extensively on proteins from microorganisms. With many years of experience in the pharmaceutical industry and in structural biology research, he joined the Task Force to contribute his skills to improve existing models for drug development. He is currently researching Radical SAM enzymes at INRAE. When not in […]
More about this author

Tristan Croll

Postdoctoral Research Associate @ Cambridge Institute for Medical Research, University of Cambridge
Tristan is a specialist in the modelling of atomic structures into low-resolution crystallographic and cryo-EM density, and developer of the model-building package ISOLDE. His focus in the project is on correcting the various errors in geometry and/or chemical identity that tend to occur in less well-resolved regions, with the overall aim of bringing the standards […]
More about this author

Gianluca Santoni

Serial Crystallography Data Scientist @ European Synchrotron Radiation Facility, Grenoble, France
Gianluca is an expert in protein crystallography data collection and analysis. After a PhD in structure-based drug design, he has worked as a postdoc on the beamline ID23-1 at the European Synchrotron Radiation Facility (ESRF) and has developed the SSX data analysis software ccCluster. His current interests are the optimization of data collection strategies for […]
More about this author

Yunyun Gao

Postdoctoral Research Associate in the AUSPEX Project @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Yunyun is a method developer for strategies of analysing data from biomacromolecules. Before joining the Thorn group, he had been working on SAXS/WAXS of polymers and proteins. He is interested in improving objectivity and reliability of data analysis. Yunyun is currently extending the functionality of AUSPEX. He is the repository manager and AUSPEX handler for […]
More about this author

Johannes Kaub

Scientific Coordinator @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Johannes Kaub studied chemistry at RWTH Aachen, with a focus on solid-state physical chemistry, before serving as a scientific employee at the Max Planck Instiute for the Structure and Dynamics of Matter. He supports the Coronavirus Structural Task Force as a scientific coordinator with his organizing ability and his talent for solving problems. Other than […]
More about this author

Andrea Thorn

Group Leader @ Institute for Nanostructure and Solid-State Physics, Hamburg University
Andrea is a specialist for crystallography and Cryo-EM structure solution, having contributed to programs like SHELX, ANODE and (a little bit) to PHASER in the past. Her group develops the diffraction diagnostics tool AUSPEX, a neural network for secondary structure annotation of Cryo-EM maps (HARUSPEX) and enables other scientists to solve problem structures. Andrea is […]
More about this author

Leave a Reply

Your email address will not be published. Required fields are marked *

cross