SIMMER employs similarity algorithms to accurately identify human gut microbiome species and enzymes capable of known chemical transformations

  1. Annamarie E Bustion
  2. Renuka R Nayak
  3. Ayushi Agrawal
  4. Peter J Turnbaugh
  5. Katie S Pollard  Is a corresponding author
  1. University of California, San Francisco, United States
  2. Gladstone Institutes, United States

Abstract

Bacteria within the gut microbiota possess the ability to metabolize a wide array of human drugs, foods, and toxins, but the responsible enzymes for these chemical events remain largely uncharacterized due to the time-consuming nature of current experimental approaches. Attempts have been made in the past to computationally predict which bacterial species and enzymes are responsible for chemical transformations in the gut environment, but with low accuracy due to minimal chemical representation and sequence similarity search schemes. Here, we present an in silico approach that employs chemical and protein Similarity algorithms that Identify MicrobioMe Enzymatic Reactions (SIMMER). We show that SIMMER accurately predicts the responsible species and enzymes for a queried reaction, unlike previous methods. We demonstrate SIMMER use cases in the context of drug metabolism by predicting previously uncharacterized enzymes for 88 drug transformations known to occur in the human gut. We validate these predictions on external datasets and provide an in vitro validation of SIMMER's predictions for metabolism of methotrexate, an anti-arthritic drug. After demonstrating its utility and accuracy, we made SIMMER available as both a command-line and web tool, with flexible input and output options for determining chemical transformations within the human gut. We present SIMMER as a computational addition to the microbiome researcher's toolbox, enabling them to make informed hypotheses before embarking on the lengthy laboratory experiments required to characterize novel bacterial enzymes that can alter human ingested compounds.

Data availability

Data generated and analyzed during this study are provided in Figures 2-10 source data files, Table 1 source data file, supplemental files, and at https://github.com/aebustion/SIMMER. Accession numbers of previously published datasets are provided in the Materials and Methods section. SIMMER code can either be run at the SIMMER website (https://simmer.pollard.gladstone.org/) or downloaded directly from the above-linked GitHub.

The following previously published data sets were used

Article and author information

Author details

  1. Annamarie E Bustion

    Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-7380-3619
  2. Renuka R Nayak

    Department of Medicine, University of California, San Francisco, San Francisco, United States
    Competing interests
    No competing interests declared.
  3. Ayushi Agrawal

    Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-2940-8926
  4. Peter J Turnbaugh

    Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
    Competing interests
    Peter J Turnbaugh, Reviewing editor, eLife.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-0888-2875
  5. Katie S Pollard

    Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, United States
    For correspondence
    kpollard@gladstone.ucsf.edu
    Competing interests
    Katie S Pollard, is a consultant for Phylagen Inc..
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-9870-6196

Funding

PhRMA Foundation (Predoctoral Fellowship)

  • Annamarie E Bustion

ARCS Foundation (Graduate Student Scholarship)

  • Annamarie E Bustion

UCSF Benioff Center for Microbiome Medicine (Trainee Pilot Award)

  • Annamarie E Bustion

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Reviewing Editor

  1. Matthew Redinbo, The University of North Carolina - Chapel Hill, United States

Version history

  1. Received: August 3, 2022
  2. Preprint posted: August 4, 2022 (view preprint)
  3. Accepted: June 11, 2023
  4. Accepted Manuscript published: June 12, 2023 (version 1)
  5. Accepted Manuscript updated: June 14, 2023 (version 2)
  6. Version of Record published: June 23, 2023 (version 3)

Copyright

© 2023, Bustion et al.

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 1,260
    views
  • 123
    downloads
  • 3
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Annamarie E Bustion
  2. Renuka R Nayak
  3. Ayushi Agrawal
  4. Peter J Turnbaugh
  5. Katie S Pollard
(2023)
SIMMER employs similarity algorithms to accurately identify human gut microbiome species and enzymes capable of known chemical transformations
eLife 12:e82401.
https://doi.org/10.7554/eLife.82401

Share this article

https://doi.org/10.7554/eLife.82401

Further reading

    1. Biochemistry and Chemical Biology
    2. Microbiology and Infectious Disease
    Natalia E Ketaren, Fred D Mast ... John D Aitchison
    Research Advance

    To date, all major modes of monoclonal antibody therapy targeting SARS-CoV-2 have lost significant efficacy against the latest circulating variants. As SARS-CoV-2 omicron sublineages account for over 90% of COVID-19 infections, evasion of immune responses generated by vaccination or exposure to previous variants poses a significant challenge. A compelling new therapeutic strategy against SARS-CoV-2 is that of single-domain antibodies, termed nanobodies, which address certain limitations of monoclonal antibodies. Here, we demonstrate that our high-affinity nanobody repertoire, generated against wild-type SARS-CoV-2 spike protein (Mast et al., 2021), remains effective against variants of concern, including omicron BA.4/BA.5; a subset is predicted to counter resistance in emerging XBB and BQ.1.1 sublineages. Furthermore, we reveal the synergistic potential of nanobody cocktails in neutralizing emerging variants. Our study highlights the power of nanobody technology as a versatile therapeutic and diagnostic tool to combat rapidly evolving infectious diseases such as SARS-CoV-2.

    1. Biochemistry and Chemical Biology
    Benjamin R Duewell, Naomi E Wilson ... Scott D Hansen
    Research Article

    Phosphoinositide 3-kinase (PI3K) beta (PI3Kβ) is functionally unique in the ability to integrate signals derived from receptor tyrosine kinases (RTKs), G-protein coupled receptors, and Rho-family GTPases. The mechanism by which PI3Kβ prioritizes interactions with various membrane-tethered signaling inputs, however, remains unclear. Previous experiments did not determine whether interactions with membrane-tethered proteins primarily control PI3Kβ localization versus directly modulate lipid kinase activity. To address this gap in our knowledge, we established an assay to directly visualize how three distinct protein interactions regulate PI3Kβ when presented to the kinase in a biologically relevant configuration on supported lipid bilayers. Using single molecule Total Internal Reflection Fluorescence (TIRF) Microscopy, we determined the mechanism controlling PI3Kβ membrane localization, prioritization of signaling inputs, and lipid kinase activation. We find that auto-inhibited PI3Kβ prioritizes interactions with RTK-derived tyrosine phosphorylated (pY) peptides before engaging either GβGγ or Rac1(GTP). Although pY peptides strongly localize PI3Kβ to membranes, stimulation of lipid kinase activity is modest. In the presence of either pY/GβGγ or pY/Rac1(GTP), PI3Kβ activity is dramatically enhanced beyond what can be explained by simply increasing membrane localization. Instead, PI3Kβ is synergistically activated by pY/GβGγ and pY/Rac1 (GTP) through a mechanism consistent with allosteric regulation.