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

  • 1,306
    views
  • 127
    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
    Zheng Ruan, Junuk Lee ... Wei Lü
    Research Article

    Protein phosphorylation is one of the major molecular mechanisms regulating protein activity and function throughout the cell. Pannexin 1 (PANX1) is a large-pore channel permeable to ATP and other cellular metabolites. Its tyrosine phosphorylation and subsequent activation have been found to play critical roles in diverse cellular conditions, including neuronal cell death, acute inflammation, and smooth muscle contraction. Specifically, the non-receptor kinase Src has been reported to phosphorylate Tyr198 and Tyr308 of mouse PANX1 (equivalent to Tyr199 and Tyr309 of human PANX1), resulting in channel opening and ATP release. Although the Src-dependent PANX1 activation mechanism has been widely discussed in the literature, independent validation of the tyrosine phosphorylation of PANX1 has been lacking. Here, we show that commercially available antibodies against the two phosphorylation sites mentioned above—which were used to identify endogenous PANX1 phosphorylation at these two sites—are nonspecific and should not be used to interpret results related to PANX1 phosphorylation. We further provide evidence that neither tyrosine residue is a major phosphorylation site for Src kinase in heterologous expression systems. We call on the field to re-examine the existing paradigm of tyrosine phosphorylation-dependent activation of the PANX1 channel.

    1. Biochemistry and Chemical Biology
    2. Cell Biology
    Christopher TA Lewis, Elise G Melhedegaard ... Julien Ochala
    Research Article

    Hibernation is a period of metabolic suppression utilized by many small and large mammal species to survive during winter periods. As the underlying cellular and molecular mechanisms remain incompletely understood, our study aimed to determine whether skeletal muscle myosin and its metabolic efficiency undergo alterations during hibernation to optimize energy utilization. We isolated muscle fibers from small hibernators, Ictidomys tridecemlineatus and Eliomys quercinus and larger hibernators, Ursus arctos and Ursus americanus. We then conducted loaded Mant-ATP chase experiments alongside X-ray diffraction to measure resting myosin dynamics and its ATP demand. In parallel, we performed multiple proteomics analyses. Our results showed a preservation of myosin structure in U. arctos and U. americanus during hibernation, whilst in I. tridecemlineatus and E. quercinus, changes in myosin metabolic states during torpor unexpectedly led to higher levels in energy expenditure of type II, fast-twitch muscle fibers at ambient lab temperatures (20 °C). Upon repeating loaded Mant-ATP chase experiments at 8 °C (near the body temperature of torpid animals), we found that myosin ATP consumption in type II muscle fibers was reduced by 77–107% during torpor compared to active periods. Additionally, we observed Myh2 hyper-phosphorylation during torpor in I. tridecemilineatus, which was predicted to stabilize the myosin molecule. This may act as a potential molecular mechanism mitigating myosin-associated increases in skeletal muscle energy expenditure during periods of torpor in response to cold exposure. Altogether, we demonstrate that resting myosin is altered in hibernating mammals, contributing to significant changes to the ATP consumption of skeletal muscle. Additionally, we observe that it is further altered in response to cold exposure and highlight myosin as a potentially contributor to skeletal muscle non-shivering thermogenesis.