Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Am Chem Soc ; 142(22): 9896-9901, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32412752

ABSTRACT

Polyketide synthase (PKS) engineering is an attractive method to generate new molecules such as commodity, fine and specialty chemicals. A significant challenge is re-engineering a partially reductive PKS module to produce a saturated ß-carbon through a reductive loop (RL) exchange. In this work, we sought to establish that chemoinformatics, a field traditionally used in drug discovery, offers a viable strategy for RL exchanges. We first introduced a set of donor RLs of diverse genetic origin and chemical substrates  into the first extension module of the lipomycin PKS (LipPKS1). Product titers of these engineered unimodular PKSs correlated with chemical structure similarity between the substrate of the donor RLs and recipient LipPKS1, reaching a titer of 165 mg/L of short-chain fatty acids produced by the host Streptomyces albus J1074. Expanding this method to larger intermediates that require bimodular communication, we introduced RLs of divergent chemosimilarity into LipPKS2 and determined triketide lactone production. Collectively, we observed a statistically significant correlation between atom pair chemosimilarity and production, establishing a new chemoinformatic method that may aid in the engineering of PKSs to produce desired, unnatural products.


Subject(s)
Computational Biology , Polyketide Synthases/chemistry , Protein Engineering , Molecular Structure , Polyketide Synthases/metabolism
2.
PLoS One ; 12(2): e0171413, 2017.
Article in English | MEDLINE | ID: mdl-28178331

ABSTRACT

This study presents an analysis of the small molecule bioactivity profiles across large quantities of diverse protein families represented in PubChem BioAssay. We compared the bioactivity profiles of FDA approved drugs to non-FDA approved compounds, and report several distinct patterns characteristic of the approved drugs. We found that a large fraction of the previously reported higher target promiscuity among FDA approved compounds, compared to non-FDA approved bioactives, was frequently due to cross-reactivity within rather than across protein families. We identified 804 potentially novel protein target candidates for FDA approved drugs, as well as 901 potentially novel target candidates with active non-FDA approved compounds, but no FDA approved drugs with activity against these targets. We also identified 486348 potentially novel compounds active against the same targets as FDA approved drugs, as well as 153402 potentially novel compounds active against targets without active FDA approved drugs. By quantifying the agreement among replicated screens, we estimated that more than half of these novel outcomes are reproducible. Using biclustering, we identified many dense clusters of FDA approved drugs with enriched activity against a common set of protein targets. We also report the distribution of compound promiscuity using a Bayesian statistical model, and report the sensitivity and specificity of two common methods for identifying promiscuous compounds. Aggregator assays exhibited greater accuracy in identifying highly promiscuous compounds, while PAINS substructures were able to identify a much larger set of "middle range" promiscuous compounds. Additionally, we report a large number of promiscuous compounds not identified as aggregators or PAINS. In summary, the results of this study represent a rich reference for selecting novel drug and target protein candidates, as well as for eliminating candidate compounds with unselective activities.


Subject(s)
Drug Discovery , Proteome , Proteomics , Small Molecule Libraries , Cluster Analysis , Computational Biology/methods , Data Mining , Drug Discovery/methods , High-Throughput Screening Assays , Models, Statistical , Protein Binding , Proteomics/methods , Reproducibility of Results
3.
J Chem Inf Model ; 56(7): 1237-42, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27367556

ABSTRACT

Despite a large and rapidly growing body of small molecule bioactivity screens available in the public domain, systematic leverage of the data to assess target druggability and compound selectivity has been confounded by a lack of suitable cross-target analysis software. We have developed bioassayR, a computational tool that enables simultaneous analysis of thousands of bioassay experiments performed over a diverse set of compounds and biological targets. Unique features include support for large-scale cross-target analyses of both public and custom bioassays, generation of high throughput screening fingerprints (HTSFPs), and an optional preloaded database that provides access to a substantial portion of publicly available bioactivity data. bioassayR is implemented as an open-source R/Bioconductor package available from https://bioconductor.org/packages/bioassayR/ .


Subject(s)
Biological Assay , Computational Biology/methods , Small Molecule Libraries/pharmacology , Databases, Pharmaceutical , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...