Conference Agenda

Overview and details of the sessions of this conference. Please select a date or location to show only sessions at that day or location. Please select a single session for detailed view (with abstracts and downloads if available).

Please note that all times are shown in the time zone of the conference. The current conference time is: 20th Apr 2024, 01:56:07am CEST

 
Filter by Area or Type of Session 
Only Sessions at Location/Venue 
 
 
Session Overview
Location: Panorama
400 1st floor
Date: Tuesday, 17/Aug/2021
2:50pm - 3:40pmPL-1: How data have revealed the structural universe
Location: Panorama
Session Chair: John Richard Helliwell

Helen Berman

 

The data universe of structural biology

Helen Berman1,2

1Rutgers University, United States of America; 2University of Southern California, United States of America

The Protein Data Bank (PDB) has grown from a small data resource for crystallographers to a worldwide resource a very broad community of researchers and educators. In this talk I will describe the history of the growth of the PDB and the role that the community has played in developing standards and policies. I also present examples of how other biophysics communities are collaborating with the worldwide PDB to create a network of interoperating data resources. This network will expand the capabilities of structural biology and enable the determination of increasingly complex structures.

External Resource:
Video Link
 
3:45pm - 4:40pmBragg: W.H. and W.L. Bragg Prize
Location: Panorama
Session Chair: Elena Boldyreva
 

Structure-guided design of next-generation malaria vaccine

Jean-Philippe Julien

The Hospital for Sick Children, Toronto, Canada

Half of the world’s population is at risk of contracting malaria, which is caused by Plasmodium parasites. Despite extensive public health and biomedical measures, the incidence of malaria continues to rise, with over 200 million cases each year. A highly effective vaccine will likely be required to eradicate malaria; however, current vaccine candidates against Plasmodium falciparum have fallen short in great part because of a lack of understanding of immunity to the parasite at the molecular level. Our recent X-ray crystallography and cryoEM work has revealed structural details of antibody immunity against the malaria parasite. These molecular blueprints of parasite inhibition by antibodies are being leveraged as guides for the design of next-generation subunit vaccines.

External Resource:
Video Link


Targeting COVID-19 Viral Enzymes in an Evolving Landscape of Publishing and Peer Review

James Fraser

UCSF, San Francisco, United States of America

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) macrodomain within the nonstructural protein 3 counteracts host-mediated antiviral adenosine diphosphate-ribosylation signaling. This enzyme is a promising antiviral target because catalytic mutations render viruses nonpathogenic. We conducted a massive crystallographic screening and computational docking effort, identifying new chemical matter primarily targeting the active site of the macrodomain. X-ray data collection to ultra-high resolution and at physiological temperature enabled assessment of the conformational heterogeneity around the active site. Neutron diffraction data is guiding hydrogen placement to improve docking calculations. Several hits have promising activity in solution and provide starting points for development of potent SARS-CoV-2 macrodomain inhibitors. The role of entropy in modulating binding affinity will also be discussed.

External Resource:
Video Link
 
Date: Saturday, 21/Aug/2021
2:45pm - 5:10pmMS-97: New methods to fight the pandemic
Location: Panorama
Session Chair: Jane Shelby Richardson
Session Chair: Nicholas Mark Pearce

Invited: Jasmine Young (USA), Tristan Croll (UK)

 
2:45pm - 2:50pm

Introduction to session

Jane Shelby Richardson, Nicholas Mark Pearce



2:50pm - 3:20pm

"Enhanced peer review" - rebuilding early SARS-CoV-2 structures in ISOLDE

Tristan Ian Croll

University of Cambridge, Cambridge, United Kingdom

The first few months of the SARS-CoV-2 pandemic illustrated, in many ways, the level of maturity and essential nature of modern structural biology. The outbreak was given official pandemic status on 11 Feb 2020 - six days after the release of the first crystal structure of the main protease; the first cryo-EM structure of the spike protein was released just two weeks later. These were the first of a flood of new structures - most, in a strong break with tradition, released well before the associated manuscripts. This, combined with the recent decision by the worldwide Protein Data Bank to allow re-versioning of submitted structures by the authors, allowed for an almost unprecedented scenario: while the experimentalists worked to get these critically important structures solved as quickly as possible, specialists in model building and refinement could check and (where necessary) improve their models, returning the results to the original authors often before their papers were ever published.

In this talk I will discuss some of my observations arising from inspecting and rebuilding some three dozen early SARS-CoV-2 and SARS-CoV-1 structures. In a great many respects, the remarkable improvement in the rate of modelling errors between SARS-CoV-1 and -2 structures shows just how far the field has come. However, the devil is in the details, and various classes of repeated errors in the modern structures point to the need for further improvement in model-building and validation methods.

External Resource:
Video Link


3:20pm - 3:50pm

Improving the quality of 3D structure data in the Protein Data Bank with coordinate versioning supported by OneDep

Jasmine Y. Young

RCSB Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey,Piscataway, United States of America

The Protein Data Bank (PDB) [1] was established as the first open-access digital data resource in biology in 1971 with just seven X-ray crystallographic structures of proteins. Today, the single global archive houses more than 177,000 experimentally determined 3D structures of biological macromolecules that are made freely available to millions of users worldwide with no limitations on usage. This information facilitates basic and applied research and education across the sciences, impacting fundamental biology, biomedicine, biotechnology, bioengineering, and energy sciences. The PDB archive is managed jointly by the Worldwide Protein Data Bank [2-3] (wwPDB, wwpdb.org) which is committed to making PDB data Findable-Accessible-Interoperable–Reusable (FAIR) [4].

To ensure the highest quality structure data, the wwPDB OneDep system for structure deposition [5], validation [6], and biocuration [7] (deposit.wwpdb.org) provides enhanced validation reports. During 2019-2020, wwPDB implemented depositor-initiated coordinate versioning that enables the Depositor of Record (or Principal Investigator) to replace previously released x,y,z atomic coordinates without obsoleting the original PDB entry or changing the PDB ID. This feature was developed in response to feedback from PDB depositors, who were reluctant to update their structures because the newly issued PDB ID would differ from that reported in original structure publication. We thank all of many PDB depositors, who have proactively corrected their structures using the new versioning feature within OneDep. To date, more than 200 PDB structures have been updated with newly versioned atomic coordinates.

Since the early days of the COVID-19 pandemic, PDB data have informed our understanding of SARS-CoV-2 protein structure, function, and evolution, and facilitated structure-guided discovery and development of anti-coronaviral drugs, vaccines, and neutralizing monoclonal antibodies. More than 1,000 SARS-CoV-2 related protein structures are now freely available from the PDB, reflecting enormous efforts made by the structural biology community in fighting the pandemic. Occasionally, rapid PDB data deposition and publication of coronavirus structural studies driven by an understandable sense of urgency has resulted in public release of PDB structures containing minor errors. The wwPDB coordinate versioning feature described above has enabled rapid correction of SARS-CoV-2 related structures archived in the PDB.

The wwPDB strongly encourages PDB depositors to update structures as needed using OneDep. Doing so will improve the quality of data stored in the archive, while preserving original PDB IDs and maintaining connections to the scientific literature.

[1] Protein Data Bank. (1971). Crystallography: Protein Data Bank. Nature (London), New Biol. 233:223-223.

[2] Berman, H., Henrick, K., Nakamura, H. (2003). Announcing the worldwide protein data bank. Nat Struct Biol. 10:980.

[3] wwPDB consortium. (2019). Protein Data Bank: The single global archive for 3d macromolecular structure data. Nucleic Acids Res. 47:D520-D528.

[4] Wilkinson, MD, Dumontier, M, Aalbersberg, IJ, Appleton, G, Axton, M, Baak, A, Blomberg, N, Boiten, JW, da Silva Santos, LB, Bourne, PE, et al. (2016). The FAIR guiding principles for scientific data management and stewardship. Sci Data. 3:1-9

[5] Young, J. Y., Westbrook, J. D., Feng, Z., Sala, R., Peisach, E., Oldfield, T. J., Sen, S., Gutmanas, A., Armstrong, D. R., Berrisford, J. M., et al. (2017). OneDep: Unified wwpdb system for deposition, biocuration, and validation of macromolecular structures in the pdb archive. Structure. 25:536-545.

[6] Gore, S., Sanz Garcia, E., Hendrickx, P. M. S., Gutmanas, A., Westbrook, J. D., Yang, H., Feng, Z., Baskaran, K., Berrisford, J. M., Hudson, B. P., et al. (2017). Validation of structures in the protein data bank. Structure. 25:1916-1927.

[7] Young, J. Y., Westbrook, J. D., Feng, Z., Peisach, E., Persikova, I., Sala, R., Sen, S., Berrisford, J. M., Swaminathan, G. J., Oldfield, T. J., et al. (2018). Worldwide protein data bank biocuration supporting open access to high-quality 3d structural biology data. Database. 2018:bay002.

RCSB PDB is funded by the National Science Foundation (DBI-1832184), the US Department of Energy (DE-SC0019749), and the National Cancer Institute, National Institute of Allergy and Infectious Diseases, and National Institute of General Medical Sciences of the National Institutes of Health under grant R01GM133198.

External Resource:
Video Link


3:50pm - 4:15pm

Neutron crystallography for drug design targeting SARS-CoV-2 viral proteins

Andrey Kovalevsky, Daniel Kneller, Leighton Coates

Oak Ridge National Lab, Oak Ridge, United States of America

COVID-19, caused by SARS-CoV-2, is a global health and economic catastrophe. The viral main protease (Mpro) is indispensable for SARS-CoV-2 replication and thus is an important target for small-molecule antivirals. Computer-assisted and structure-guided drug design strategies rely on atomic scale understanding of the target biomacromolecule traditionally derived from X-ray crystallographic data collected at cryogenic temperatures. Conventional protein X-ray crystallography is limited by possible cryo-artifacts and its inability to locate the functional hydrogen atoms crucial for understanding chemistry occurring in enzyme active sites. Neutrons are an ideal probe to observe the protonation states of ionizable amino acids at near-physiological temperature, directly determining their electric charges – crucial information for drug design. Our X-ray crystal structures of Mpro collected at near-physiological temperatures brought rapid insights into the reactivity of the catalytic cysteine, malleability of the active site, and binding modes with clinical protease inhibitors. The neutron crystal structures of ligand-free and inhibitor-bound Mpro were determined allowing the direct observation of protonation states of all residues in a coronavirus protein for the first time. At rest, the catalytic Cys-His dyad exists in the reactive zwitterionic state, with both Cys145 and His41 charged, instead of the anticipated neutral state. Covalent inhibitor binding results in modulation of the protonation states, retaining the overall electric charge of the Mpro active site cavity. In addition, high-throughput virtual screening in conjunction with in vitro assays identified a lead non-covalent compound with micromolar affinity, which is being used to design novel Mpro inhibitors. Our research is providing real-time data for atomistic design and discovery of Mpro inhibitors to combat the COVID-19 pandemic and prepare for future threats from pathogenic coronaviruses.

External Resource:
Video Link


4:15pm - 4:40pm

The temperature-dependent conformational ensemble of SARS-CoV-2 main protease (Mpro)

Ali Ebrahim1,2, Blake T. Riley2, Desigan Kumaran3, Babak Andi4,5, Martin R. Fuchs4, Sean McSweeney4,5, Daniel A. Keedy2,6,7

1Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, England; 2Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY 10031; 3Biology Department, Brookhaven National Laboratory, Upton, NY 11973; 4National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY 11973; 5National Virtual Biotechnology Laboratory (NVBL), US Department of Energy, Washington, DC, United States; 6Department of Chemistry and Biochemistry, City College of New York, New York, NY 10031; 7Ph.D. Programs in Biochemistry, Biology, and Chemistry, The Graduate Center – City University of New York, New York, NY 10016

The COVID-19 pandemic, instigated by the SARS-CoV-2 coronavirus, continues to plague the globe. The SARS-CoV-2 main protease, or Mpro, is a promising target for development of novel antiviral therapeutics. Previous X-ray crystal structures of Mpro were obtained at cryogenic temperature or room temperature only. Here we report a series of high-resolution crystal structures of unliganded Mpro across multiple temperatures from cryogenic to physiological, and another at high humidity. We interrogate these datasets with parsimonious multiconformer models, multi-copy ensemble models, and isomorphous difference density maps. Our analysis reveals a temperature-dependent conformational landscape for Mpro, including a mobile water interleaved between the catalytic dyad, mercurial conformational heterogeneity in a key substrate-binding loop, and a far-reaching intramolecular network bridging the active site and dimer interface. Our results may inspire new strategies for antiviral drug development to counter-punch COVID-19 and combat future coronavirus pandemics.

External Resource:
Video Link


4:40pm - 5:05pm

Validation of cryo-EM structures of SARS-CoV-2 and mapping genomic mutations

Agnel Praveen Joseph, Sony Malhotra, Martyn Winn

STFC, Scientific Computing Department, Harwell, Didcot, United Kingdom

Technological developments and growing interests in the study of cellular assemblies have led cryo-EM as a powerful technique to solve the three-dimensional structures of macromolecular complexes. More than 450 structures of molecular complexes from SARS-CoV-2 have been solved using cryo-EM and the structures have been crucial to understand molecular details behind the viral infection and development of drug molecules and vaccines. Given that majority of the cryo-EM reconstructions are solved at resolutions worse than 2.5Å and often the local resolution within the map varies considerably, atomic model validation is crucial to identify errors and less reliable areas of the model.

Here, we present the database CoVal, which is a repository of amino acid replacement mutations identified in the SARS-CoV-2 genome sequences, mapped onto protein structures from cryo-EM and X-ray crystallography. We provide information on the demographic distribution of these mutations, along with details on co-occuring mutations. CoVal gives easy access to mutation sites mapped to known structures with multiple metrics on the quality of the structure and agreement with experimental data. We also provide validation scores for the local quality of mutation site(s) and their structural neighbors. The database is freely accessible at: https://coval.ccpem.ac.uk. We also discuss tools for atomic model validation in the CCP-EM software suite [1] and results from the validation analysis of cryo-EM structures from SARS-CoV-2.

External Resource:
Video Link
 
6:10pm - 7:00pmPL-3: The structural biology of pathogenic amyloid fibrils
Location: Panorama

David Eisenberg

 

The Expanding Amyloid Family: Structure, Stability, Function, and Pathogenesis

David Eisenberg

UCLA, Los Angeles, United States of America

The hidden world of amyloid biology has suddenly snapped into atomic level focus revealing over 80 amyloid protein fibrils, both pathogenic and functional. Many of the most prevalent degenerative diseases, including Alzheimer’s, Parkinson’s, ALS, and type 2 diabetes are associated with particular proteins in amyloid fibril form. Fibrils structures determined X-ray and electron crystallography, as well as particle averaging by cryoEM, and solid-state NMR have contributed to deepened understanding of the formation, stability, and pathology of structures have led to design of compounds that inhibit fibril formation as well as some compounds that disaggregated fibrils. A subclass of functional amyloid-like fibrils are formed by reversible interaction of low complexity domains, having underrepresented members of the 20 coded amino acids. When mutated or at high concentration reversible amyloid fibrils can transition to irreversible pathogenic form. Unlike globular proteins, amyloid proteins flatten and stack into unbranched fibrils. Also unlike globular proteins, a single protein sequence can adopt wildly different two-dimensional conformations, yielding distinct amyloid fibril polymorphs. Hence, an amyloid protein may define distinct diseases depending on its conformation.

I will describe the energetic basis for the great stability of pathogenic amyloid, the structural differences found in reversible amyloid, and chemical methods for inhibiting and disaggregating amyloid. Our database of amyloid structure and energy is available at https://people.mbi.ucla.edu/sawaya/amyloidatlas/

Reference: The Expanding Amyloid Family: Structure, Stability, Function, and Pathogenesis. Michael R. Sawaya, Michael P. Hughes, Jose A. Rodriguez, Roland Riek, David S. Eisenberg. Cell, in press.

External Resource:
Video Link
 
7:00pm - 7:45pmClosing ceremony: Poster prizes, closing
Location: Panorama

 
Contact and Legal Notice · Contact Address:
Privacy Statement · Conference: IUCr 2021 | August 14 - 22, 2021 | Prague, Czech Republic
Conference Software: ConfTool Pro 2.8.101+TC+CC
© 2001–2024 by Dr. H. Weinreich, Hamburg, Germany