Benchmarking AlphaFold for protein complex modeling reveals accuracy determinants

dc.contributor.authorYin, Rui
dc.contributor.authorYeng, Brandon F.
dc.contributor.authorVarshney, Amitabh
dc.contributor.authorPierce, Brian G.
dc.date.accessioned2023-09-27T18:00:57Z
dc.date.available2023-09-27T18:00:57Z
dc.date.issued2022-07-13
dc.description.abstractHigh-resolution experimental structural determination of protein–protein interactions has led to valuable mechanistic insights, yet due to the massive number of interactions and experimental limitations there is a need for computational methods that can accurately model their structures. Here we explore the use of the recently developed deep learning method, AlphaFold, to predict structures of protein complexes from sequence. With a benchmark of 152 diverse heterodimeric protein complexes, multiple implementations and parameters of AlphaFold were tested for accuracy. Remarkably, many cases (43%) had near-native models (medium or high critical assessment of predicted interactions accuracy) generated as top-ranked predictions by AlphaFold, greatly surpassing the performance of unbound protein–protein docking (9% success rate for near-native top-ranked models), however AlphaFold modeling of antibody–antigen complexes within our set was unsuccessful. We identified sequence and structural features associated with lack of AlphaFold success, and we also investigated the impact of multiple sequence alignment input. Benchmarking of a multimer-optimized version of AlphaFold (AlphaFold-Multimer) with a set of recently released antibody–antigen structures confirmed a low rate of success for antibody–antigen complexes (11% success), and we found that T cell receptor–antigen complexes are likewise not accurately modeled by that algorithm, showing that adaptive immune recognition poses a challenge for the current AlphaFold algorithm and model. Overall, our study demonstrates that end-to-end deep learning can accurately model many transient protein complexes, and highlights areas of improvement for future developments to reliably model any protein–protein interaction of interest.
dc.description.urihttps://doi.org/10.1002/pro.4379
dc.identifierhttps://doi.org/10.13016/dspace/xbwe-1rix
dc.identifier.citationYin, R, Feng, BY, Varshney, A, Pierce, BG. Benchmarking AlphaFold for protein complex modeling reveals accuracy determinants. Protein Science. 2022; 31(8):e4379.
dc.identifier.urihttp://hdl.handle.net/1903/30597
dc.language.isoen_US
dc.publisherWiley
dc.relation.isAvailableAtCell Biology & Molecular Geneticsen_us
dc.relation.isAvailableAtDigital Repository at the University of Marylanden_us
dc.relation.isAvailableAtCollege of Computer, Mathematical & Natural Sciencesen_us
dc.relation.isAvailableAtUniversity of Maryland (College Park, MD)en_us
dc.titleBenchmarking AlphaFold for protein complex modeling reveals accuracy determinants
dc.typeArticle
local.equitableAccessSubmissionNo

Files

Original bundle
Now showing 1 - 1 of 1
Loading...
Thumbnail Image
Name:
Protein Science - 2022 - Yin.pdf
Size:
5.06 MB
Format:
Adobe Portable Document Format
License bundle
Now showing 1 - 1 of 1
No Thumbnail Available
Name:
license.txt
Size:
1.55 KB
Format:
Item-specific license agreed upon to submission
Description: