Abstract

BackgroundDiverse microbiome communities drive biogeochemical processes and evolution of animals in their ecosystems. Many microbiome projects have demonstrated the power of using metagenomics to understand the structures and factors influencing the function of the microbiomes in their environments. In order to characterize the effects from microbiome composition for human health, diseases, and even ecosystems, one must first understand the relationship of microbes and their environment in different samples. Running machine learning model with metagenomic sequencing data is encouraged for this purpose, but it is not an easy task to make an appropriate machine learning model for all diverse metagenomic datasets.ResultsWe introduce MegaR, an R Shiny package and web application, to build an unbiased machine learning model effortlessly with interactive visual analysis. The MegaR employs taxonomic profiles from either whole metagenome sequencing or 16S rRNA sequencing data to develop machine learning models and classify the samples into two or more categories. It provides various options for model fine tuning throughout the analysis pipeline such as data processing, multiple machine learning techniques, model validation, and unknown sample prediction that can be used to achieve the highest prediction accuracy possible for any given dataset while still maintaining a user-friendly experience.ConclusionsMetagenomic sample classification and phenotype prediction is important particularly when it applies to a diagnostic method for identifying and predicting microbe-related human diseases. MegaR provides various interactive visualizations for user to build an accurate machine-learning model without difficulty. Unknown sample prediction with a properly trained model using MegaR will enhance researchers to identify the sample property in a fast turnaround time.

Highlights

  • Diverse microbiome communities drive biogeochemical processes and evolution of animals in their ecosystems

  • In the Type I diabetes (T1D) cohort there were 777 16S rRNA sequencing (16S) rRNA samples, out of which 175 had T1D, and 85 samples did not have T1D. 314 samples were from children from birth to one age, 297 samples were from children from one to two, and 166 children were from two to three in terms of the age group

  • In the 16S rRNA data set, there are 528 samples from children who were not treated with antibiotics while 520 samples were from children who were treated with antibiotics

Read more

Summary

Results

Dataset In order to demonstrate the efficacy of MegaR as a disease sample prediction tool, DIABIMMUNE (https://pubs.broadinstitute.org/diabimmune) microbiome project data sets were used to perform a sample pipeline execution in MegaR. Model and prediction accuracy We used MegaR to analyze different datasets from the DIABIMMUNE research group. We used MegaR to check if optimizing the threshold and percentage of sample with threshold as well as data split for training and testing improves the model (Fig. 3, Table 2). Using MegaR, we were able to obtain a slightly higher prediction accuracy for both datasets compared to the results reported by the MetAML project (Table 3). Optimal model parameters are the values used to obtain the highest accuracy for the data set. Bold values represent the highest obtained accuracy for each dataset believe that this slight increase is due to the ability of the MegaR package to fine tune the model parameters to optimize the model for each data set

Conclusions
Background
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call