10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      MICOM: Metagenome-Scale Modeling To Infer Metabolic Interactions in the Gut Microbiota

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The bacterial communities that live within the human gut have been linked to health and disease. However, we are still just beginning to understand how those bacteria interact and what potential interventions to our gut microbiome can make us healthier. Here, we present a mathematical modeling framework (named MICOM) that can recapitulate the growth rates of diverse bacterial species in the gut and can simulate metabolic interactions within microbial communities. We show that MICOM can unravel the ecological rules that shape the microbial landscape in our gut and that a given dietary or probiotic intervention can have widely different effects in different people.

          ABSTRACT

          Compositional changes in the gut microbiota have been associated with a variety of medical conditions such as obesity, Crohn’s disease, and diabetes. However, connecting microbial community composition to ecosystem function remains a challenge. Here, we introduce MICOM, a customizable metabolic model of the human gut microbiome. By using a heuristic optimization approach based on L2 regularization, we were able to obtain a unique set of realistic growth rates that corresponded well with observed replication rates. We integrated adjustable dietary and taxon abundance constraints to generate personalized metabolic models for individual metagenomic samples. We applied MICOM to a balanced cohort of metagenomes from 186 people, including a metabolically healthy population and individuals with type 1 and type 2 diabetes. Model results showed that individual bacterial genera maintained conserved niche structures across humans, while the community-level production of short-chain fatty acids (SCFAs) was heterogeneous and highly individual specific. Model output revealed complex cross-feeding interactions that would be difficult to measure in vivo. Metabolic interaction networks differed somewhat consistently between healthy and diabetic subjects. In particular, MICOM predicted reduced butyrate and propionate production in a diabetic cohort, with restoration of SCFA production profiles found in healthy subjects following metformin treatment. Overall, we found that changes in diet or taxon abundances have highly personalized effects. We believe MICOM can serve as a useful tool for generating mechanistic hypotheses for how diet and microbiome composition influence community function. All methods are implemented in an open-source Python package, which is available at https://github.com/micom-dev/micom.

          IMPORTANCE The bacterial communities that live within the human gut have been linked to health and disease. However, we are still just beginning to understand how those bacteria interact and what potential interventions to our gut microbiome can make us healthier. Here, we present a mathematical modeling framework (named MICOM) that can recapitulate the growth rates of diverse bacterial species in the gut and can simulate metabolic interactions within microbial communities. We show that MICOM can unravel the ecological rules that shape the microbial landscape in our gut and that a given dietary or probiotic intervention can have widely different effects in different people.

          Related collections

          Most cited references26

          • Record: found
          • Abstract: not found
          • Article: not found

          Ridge Regression: Biased Estimation for Nonorthogonal Problems

            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            COBRApy: COnstraints-Based Reconstruction and Analysis for Python

            Background COnstraint-Based Reconstruction and Analysis (COBRA) methods are widely used for genome-scale modeling of metabolic networks in both prokaryotes and eukaryotes. Due to the successes with metabolism, there is an increasing effort to apply COBRA methods to reconstruct and analyze integrated models of cellular processes. The COBRA Toolbox for MATLAB is a leading software package for genome-scale analysis of metabolism; however, it was not designed to elegantly capture the complexity inherent in integrated biological networks and lacks an integration framework for the multiomics data used in systems biology. The openCOBRA Project is a community effort to promote constraints-based research through the distribution of freely available software. Results Here, we describe COBRA for Python (COBRApy), a Python package that provides support for basic COBRA methods. COBRApy is designed in an object-oriented fashion that facilitates the representation of the complex biological processes of metabolism and gene expression. COBRApy does not require MATLAB to function; however, it includes an interface to the COBRA Toolbox for MATLAB to facilitate use of legacy codes. For improved performance, COBRApy includes parallel processing support for computationally intensive processes. Conclusion COBRApy is an object-oriented framework designed to meet the computational challenges associated with the next generation of stoichiometric constraint-based models and high-density omics data sets. Availability http://opencobra.sourceforge.net/
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Generation of genome-scale metabolic reconstructions for 773 members of the human gut microbiota

              A large set of microbial metabolic models (AGORA) could be applied to better understand the functions of the human gut microbiome.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                mSystems
                mSystems
                msys
                msys
                mSystems
                mSystems
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                2379-5077
                Jan-Feb 2020
                21 January 2020
                : 5
                : 1
                : e00606-19
                Affiliations
                [a ]Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, México
                [b ]Institute for Systems Biology, Seattle, Washington, USA
                [c ]eScience Institute, University of Washington, Seattle, Washington, USA
                [d ]Human Systems Biology Laboratory, Coordinación de la Investigación Científica - Red de Apoyo a la Investigación, Universidad Nacional Autonóma de México (UNAM), Mexico City, México
                Mayo Clinic
                Author notes
                Address correspondence to Sean M. Gibbons, sgibbons@ 123456isbscience.org , or Osbaldo Resendis-Antonio, oresendis@ 123456inmegen.gob.mx .

                Citation Diener C, Gibbons SM, Resendis-Antonio O. 2020. MICOM: metagenome-scale modeling to infer metabolic interactions in the gut microbiota. mSystems 5:e00606-19. https://doi.org/10.1128/mSystems.00606-19.

                Author information
                https://orcid.org/0000-0002-7476-0868
                https://orcid.org/0000-0002-8724-7916
                https://orcid.org/0000-0001-5220-541X
                Article
                mSystems00606-19
                10.1128/mSystems.00606-19
                6977071
                31964767
                f126620b-bfcc-4fd8-af5e-a5f4724cda59
                Copyright © 2020 Diener et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 20 September 2019
                : 19 December 2019
                Page count
                supplementary-material: 6, Figures: 6, Tables: 1, Equations: 23, References: 60, Pages: 17, Words: 12241
                Categories
                Research Article
                Novel Systems Biology Techniques
                Custom metadata
                January/February 2020

                flux balance analysis,gut microbiome,metagenome,systems biology

                Comments

                Comment on this article