Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add filters

Database
Language
Document Type
Year range
1.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.03.15.435326

ABSTRACT

The global COVID-19 pandemic is caused by the SARS-CoV-2 virus and has infected over 100 million and caused over 2 million fatalities worldwide at the point of writing. There is currently a lack of effective drugs to treat people infected with SARS-CoV-2. The SARS-CoV-2 Non-structural protein 13 (NSP13) is a superfamily1B helicase that has been identified as a possible target for anti-viral drugs due to its high sequence conservation and essential role in viral replication. In this study we present crystal structures of SARS-CoV-2 NSP13 solved in the APO form and in the presence of both phosphate and the non-hydrolysable ATP analogue (AMP-PNP). Comparisons of these structures reveal details of global and local conformational changes that are induced by nucleotide binding and hydrolysis and provide insights into the helicase mechanism and possible modes of inhibition. Structural analysis reveals two pockets on NSP13 that are classified as "druggable" and include one of the most conserved sites in the entire SARS-CoV-2 proteome. To identify possible starting points for anti-viral drug development we have performed a crystallographic fragment screen against SARS-CoV-2 NSP13 helicase. The fragment screen reveals 65 fragment hits across 52 datasets, with hot spots in pockets predicted to be of functional importance, including the druggable nucleotide and nucleic acid binding sites, opening the way to structure guided development of novel antiviral agents.


Subject(s)
Severe Acute Respiratory Syndrome , Hallucinations , COVID-19
2.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.11.24.393405

ABSTRACT

The SARS-CoV-2 macrodomain (Mac1) within the non-structural protein 3 (Nsp3) counteracts host-mediated antiviral ADP-ribosylation signalling. This enzyme is a promising antiviral target because catalytic mutations render viruses non-pathogenic. Here, we report a massive crystallographic screening and computational docking effort, identifying new chemical matter primarily targeting the active site of the macrodomain. Crystallographic screening of diverse fragment libraries resulted in 214 unique macrodomain-binding fragments, out of 2,683 screened. An additional 60 molecules were selected from docking over 20 million fragments, of which 20 were crystallographically confirmed. X-ray data collection to ultra-high resolution and at physiological temperature enabled assessment of the conformational heterogeneity around the active site. Several crystallographic and docking fragment hits were validated for solution binding using three biophysical techniques (DSF, HTRF, ITC). Overall, the 234 fragment structures presented explore a wide range of chemotypes and provide starting points for development of potent SARS-CoV-2 macrodomain inhibitors.

3.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.11.24.390039

ABSTRACT

In order to produce proteins essential for their propagation, many pathogenic human viruses, including SARS-CoV-2 the causative agent of COVID-19 respiratory disease, commandeer host biosynthetic machineries and mechanisms. Three major structural proteins, the spike, envelope and membrane proteins, are amongst several SARS-CoV-2 components synthesised at the endoplasmic reticulum (ER) of infected human cells prior to the assembly of new viral particles. Hence, the inhibition of membrane protein synthesis at the ER is an attractive strategy for reducing the pathogenicity of SARS-CoV-2 and other obligate viral pathogens. Using an in vitro system, we demonstrate that the small molecule inhibitor ipomoeassin F (Ipom-F) potently blocks the Sec61-mediated ER membrane translocation/insertion of three therapeutic protein targets for SARS-CoV-2 infection; the viral spike and ORF8 proteins together with angiotensin-converting enzyme 2, the host cell plasma membrane receptor. Our findings highlight the potential for using ER protein translocation inhibitors such as Ipom-F as host-targeting, broad-spectrum, antiviral agents.


Subject(s)
Respiratory Tract Diseases , COVID-19
4.
- The COVID Moonshot Consortium; Hagit Achdout; Anthony Aimon; Elad Bar-David; Haim Barr; Amir Ben-Shmuel; James Bennett; Melissa L Bobby; Juliane Brun; Sarma BVNBS; Mark Calmiano; Anna Carbery; Emma Cattermole; John D. Chodera; Austin Clyde; Joseph E. Coffland; Galit Cohen; Jason Cole; Alessandro Contini; Lisa Cox; Milan Cvitkovic; Alex Dias; Alice Douangamath; Shirly Duberstein; Tim Dudgeon; Louise Dunnett; Peter K. Eastman; Noam Erez; Michael Fairhead; Daren Fearon; Oleg Fedorov; Matteo Ferla; Holly Foster; Richard Foster; Ronen Gabizon; Paul Gehrtz; Carina Gileadi; Charline Giroud; William G. Glass; Robert Glen; Itai Glinert; Marian Gorichko; Tyler Gorrie-Stone; Edward J Griffen; Jag Heer; Michelle Hill; Sam Horrell; Matthew F.D. Hurley; Tomer Israely; Andrew Jajack; Eric Jnoff; Tobias John; Anastassia L. Kantsadi; Peter W. Kenny; John L. Kiappes; Lizbe Koekemoer; Boris Kovar; Tobias Krojer; Alpha Albert Lee; Bruce A. Lefker; Haim Levy; Nir London; Petra Lukacik; Hannah Bruce Macdonald; Beth MacLean; Tika R. Malla; Tatiana Matviiuk; Willam McCorkindale; Sharon Melamed; Oleg Michurin; Halina Mikolajek; Aaron Morris; Garrett M. Morris; Melody Jane Morwitzer; Demetri Moustakas; Jose Brandao Neto; Vladas Oleinikovas; Gijs J. Overheul; David Owen; Ruby Pai; Jin Pan; Nir Paran; Benjamin Perry; Maneesh Pingle; Jakir Pinjari; Boaz Politi; Ailsa Powell; Vladimir Psenak; Reut Puni; Victor L. Rangel; Rambabu N. Reddi; St Patrick Reid; Efrat Resnick; Matthew C. Robinson; Ralph P. Robinson; Dominic Rufa; Christopher Schofield; Aarif Shaikh; Jiye Shi; Khriesto Shurrush; Assa Sittner; Rachael Skyner; Adam Smalley; Mihaela D. Smilova; John Spencer; Claire Strain-Damerell; Vishwanath Swamy; Hadas Tamir; Rachael Tennant; Andrew Thompson; Warren Thompson; Susana Tomasio; Anthony Tumber; Ioannis Vakonakis; Ronald P. van Rij; Finny S. Varghese; Mariana Vaschetto; Einat B. Vitner; Vincent Voelz; Annette von Delft; Frank von Delft; Martin Walsh; Walter Ward; Charlie Weatherall; Shay Weiss; Conor Francis Wild; Matthew Wittmann; Nathan Wright; Yfat Yahalom-Ronen; Daniel Zaidmann; Hadeer Zidane; Nicole Zitzmann.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.29.339317

ABSTRACT

Herein we provide a living summary of the data generated during the COVID Moonshot project focused on the development of SARS-CoV-2 main protease (Mpro) inhibitors. Our approach uniquely combines crowdsourced medicinal chemistry insights with high throughput crystallography, exascale computational chemistry infrastructure for simulations, and machine learning in triaging designs and predicting synthetic routes. This manuscript describes our methodologies leading to both covalent and non-covalent inhibitors displaying protease IC50 values under 150 nM and viral inhibition under 5 uM in multiple different viral replication assays. Furthermore, we provide over 200 crystal structures of fragment-like and lead-like molecules in complex with the main protease. Over 1000 synthesized and ordered compounds are also reported with the corresponding activity in Mpro enzymatic assays using two different experimental setups. The data referenced in this document will be continually updated to reflect the current experimental progress of the COVID Moonshot project, and serves as a citable reference for ensuing publications. All of the generated data is open to other researchers who may find it of use.

5.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.05.27.118117

ABSTRACT

COVID-19, caused by SARS-CoV-2, lacks effective therapeutics. Additionally, no antiviral drugs or vaccines were developed against the closely related coronavirus, SARS-CoV-1 or MERS-CoV, despite previous zoonotic outbreaks. To identify starting points for such therapeutics, we performed a large-scale screen of electrophile and non-covalent fragments through a combined mass spectrometry and X-ray approach against the SARS-CoV-2 main protease, one of two cysteine viral proteases essential for viral replication. Our crystallographic screen identified 71 hits that span the entire active site, as well as 3 hits at the dimer interface. These structures reveal routes to rapidly develop more potent inhibitors through merging of covalent and non-covalent fragment hits; one series of low-reactivity, tractable covalent fragments was progressed to discover improved binders. These combined hits offer unprecedented structural and reactivity information for on-going structure-based drug design against SARS-CoV-2 main protease.


Subject(s)
COVID-19 , Severe Acute Respiratory Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL