1
|
Lutz KC, Neugent ML, Bedi T, De Nisco NJ, Li Q. A Generalized Bayesian Stochastic Block Model for Microbiome Community Detection. Stat Med 2025; 44:e10291. [PMID: 39853798 PMCID: PMC11760646 DOI: 10.1002/sim.10291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/02/2024] [Accepted: 11/11/2024] [Indexed: 01/26/2025]
Abstract
Advances in next-generation sequencing technology have enabled the high-throughput profiling of metagenomes and accelerated microbiome studies. Recently, there has been a rise in quantitative studies that aim to decipher the microbiome co-occurrence network and its underlying community structure based on metagenomic sequence data. Uncovering the complex microbiome community structure is essential to understanding the role of the microbiome in disease progression and susceptibility. Taxonomic abundance data generated from metagenomic sequencing technologies are high-dimensional and compositional, suffering from uneven sampling depth, over-dispersion, and zero-inflation. These characteristics often challenge the reliability of the current methods for microbiome community detection. To study the microbiome co-occurrence network and perform community detection, we propose a generalized Bayesian stochastic block model that is tailored for microbiome data analysis where the data are transformed using the recently developed modified centered-log ratio transformation. Our model also allows us to leverage taxonomic tree information using a Markov random field prior. The model parameters are jointly inferred by using Markov chain Monte Carlo sampling techniques. Our simulation study showed that the proposed approach performs better than competing methods even when taxonomic tree information is non-informative. We applied our approach to a real urinary microbiome dataset from postmenopausal women. To the best of our knowledge, this is the first time the urinary microbiome co-occurrence network structure in postmenopausal women has been studied. In summary, this statistical methodology provides a new tool for facilitating advanced microbiome studies.
Collapse
Affiliation(s)
- Kevin C. Lutz
- Peter O'Donnell Jr. School of Public HealthThe University of Texas Southwestern Medical CenterDallasTexas
| | - Michael L. Neugent
- Department of Biological SciencesThe University of Texas at DallasRichardsonTexas
| | - Tejasv Bedi
- Department of Mathematical SciencesThe University of Texas at DallasRichardsonTexas
| | - Nicole J. De Nisco
- Department of Biological SciencesThe University of Texas at DallasRichardsonTexas
- Department of UrologyThe University of Texas Southwestern Medical CenterDallasTexas
| | - Qiwei Li
- Department of Mathematical SciencesThe University of Texas at DallasRichardsonTexas
| |
Collapse
|
2
|
Liu Z, Sun Y, Li Y, Ma A, Willaims NF, Jahanbahkshi S, Hoyd R, Wang X, Zhang S, Zhu J, Xu D, Spakowicz D, Ma Q, Liu B. An Explainable Graph Neural Framework to Identify Cancer-Associated Intratumoral Microbial Communities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403393. [PMID: 39225619 PMCID: PMC11538693 DOI: 10.1002/advs.202403393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/26/2024] [Indexed: 09/04/2024]
Abstract
Microbes are extensively present among various cancer tissues and play critical roles in carcinogenesis and treatment responses. However, the underlying relationships between intratumoral microbes and tumors remain poorly understood. Here, a MIcrobial Cancer-association Analysis using a Heterogeneous graph transformer (MICAH) to identify intratumoral cancer-associated microbial communities is presented. MICAH integrates metabolic and phylogenetic relationships among microbes into a heterogeneous graph representation. It uses a graph transformer to holistically capture relationships between intratumoral microbes and cancer tissues, which improves the explainability of the associations between identified microbial communities and cancers. MICAH is applied to intratumoral bacterial data across 5 cancer types and 5 fungi datasets, and its generalizability and reproducibility are demonstrated. After experimentally testing a representative observation using a mouse model of tumor-microbe-immune interactions, a result consistent with MICAH's identified relationship is observed. Source tracking analysis reveals that the primary known contributor to a cancer-associated microbial community is the organs affected by the type of cancer. Overall, this graph neural network framework refines the number of microbes that can be used for follow-up experimental validation from thousands to tens, thereby helping to accelerate the understanding of the relationship between tumors and intratumoral microbiomes.
Collapse
Affiliation(s)
- Zhaoqian Liu
- School of MathematicsShandong UniversityJinanShandong250100China
- College of SciencesXi'an University of Science and TechnologyXi'anShanxi710054China
| | - Yuhan Sun
- School of MathematicsShandong UniversityJinanShandong250100China
| | - Yingjie Li
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
| | - Anjun Ma
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Pelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOH43210USA
| | - Nyelia F. Willaims
- Department of Internal MedicineCollege of MedicineThe Ohio State UniversityColumbusOH43210USA
| | - Shiva Jahanbahkshi
- Department of Food Science and TechnologyCollege of FoodAgricultural, and Environmental SciencesThe Ohio State UniversityColumbusOH43210USA
| | - Rebecca Hoyd
- Department of Internal MedicineCollege of MedicineThe Ohio State UniversityColumbusOH43210USA
| | - Xiaoying Wang
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Pelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOH43210USA
| | - Shiqi Zhang
- Department of Human SciencesCollege of Education and Human EcologyThe Ohio State UniversityColumbusOH43210USA
| | - Jiangjiang Zhu
- Department of Human SciencesCollege of Education and Human EcologyThe Ohio State UniversityColumbusOH43210USA
| | - Dong Xu
- Department of Electrical Engineering and Computer ScienceUniversity of MissouriColumbiaMO65201USA
- Christopher S. Bond Life Sciences CenterUniversity of MissouriColumbiaMO65201USA
| | - Daniel Spakowicz
- Pelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOH43210USA
- Department of Internal MedicineCollege of MedicineThe Ohio State UniversityColumbusOH43210USA
| | - Qin Ma
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Pelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOH43210USA
| | - Bingqiang Liu
- School of MathematicsShandong UniversityJinanShandong250100China
- Shandong National Center for Applied MathematicsJinanShandong250199China
| |
Collapse
|
3
|
Abstract
The microbiome represents a hidden world of tiny organisms populating not only our surroundings but also our own bodies. By enabling comprehensive profiling of these invisible creatures, modern genomic sequencing tools have given us an unprecedented ability to characterize these populations and uncover their outsize impact on our environment and health. Statistical analysis of microbiome data is critical to infer patterns from the observed abundances. The application and development of analytical methods in this area require careful consideration of the unique aspects of microbiome profiles. We begin this review with a brief overview of microbiome data collection and processing and describe the resulting data structure. We then provide an overview of statistical methods for key tasks in microbiome data analysis, including data visualization, comparison of microbial abundance across groups, regression modeling, and network inference. We conclude with a discussion and highlight interesting future directions.
Collapse
Affiliation(s)
- Christine B Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Satabdi Saha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Zhou F, He K, Cai JJ, Davidson LA, Chapkin RS, Ni Y. A Unified Bayesian Framework for Bi-overlapping-Clustering Multi-omics Data via Sparse Matrix Factorization. STATISTICS IN BIOSCIENCES 2023; 15:669-691. [PMID: 38179127 PMCID: PMC10766378 DOI: 10.1007/s12561-022-09350-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 04/10/2021] [Accepted: 06/06/2022] [Indexed: 11/27/2022]
Abstract
The advances of modern sequencing techniques have generated an unprecedented amount of multi-omics data which provide great opportunities to quantitatively explore functional genomes from different but complementary perspectives. However, distinct modalities/sequencing technologies generate diverse types of data which greatly complicate statistical modeling because uniquely optimized methods are required for handling each type of data. In this paper, we propose a unified framework for Bayesian nonparametric matrix factorization that infers overlapping bi-clusters for multi-omics data. The proposed method adaptively discretizes different types of observations into common latent states on which cluster structures are built hierarchically. The proposed Bayesian nonparametric method is able to automatically determine the number of clusters. We demonstrate the utility of the proposed method using simulation studies and applications to a single-cell RNA-sequencing dataset, a combination of single-cell RNA-sequencing and single-cell ATAC-sequencing dataset, a bulk RNA-sequencing dataset, and a DNA methylation dataset which reveal several interesting findings that are consistent with biological literature.
Collapse
Affiliation(s)
- Fangting Zhou
- Institute of Statistics and Big Data, Renmin University of China, Beijing, China
- Department of Statistics, Texas A&M University, College Station, USA
| | - Kejun He
- Institute of Statistics and Big Data, Renmin University of China, Beijing, China
| | - James J. Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, USA
| | - Laurie A. Davidson
- Department of Nutrition and Food Science, Texas A&M University, College Station, USA
- Program in Integrative Nutrition and Complex Diseases, Texas A &M University, College Station, USA
| | - Robert S. Chapkin
- Department of Nutrition and Food Science, Texas A&M University, College Station, USA
- Program in Integrative Nutrition and Complex Diseases, Texas A &M University, College Station, USA
| | - Yang Ni
- Department of Statistics, Texas A&M University, College Station, USA
| |
Collapse
|
5
|
Wang J, Liu A, Li A, Song H, Luo P, Zhan M, Zhou X, Chen L, Zhang J, Wang R. Lactobacillus fermentum CKCC1858 alleviates hyperlipidemia in golden hamsters on a high-fat diet via modulating gut microbiota. Food Funct 2023; 14:9580-9590. [PMID: 37823897 DOI: 10.1039/d3fo02618c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
To investigate the effect of probiotic Lactobacillus fermentum CKCC1858, LF on the prevention of hyperlipidemia and its correlation with gut microbiota, golden hamsters were fed a high-fat diet alone or in combination with the probiotic for 6 weeks. The results showed that the LF intervention alleviated HFD-induced hyperlipidemia and liver damage, as evidenced by the reduced serum lipid profile levels and liver function markers. More importantly, the LF intervention attenuated HFD-induced microbiota dysbiosis by enhancing the abundance of SCFA-producing bacteria and reshaping the metabolic functions of the gut microbiota, likely contributing to its pronounced preventive effects on hyperlipidemia. This study elucidated the mechanism of the preventive effect of probiotics on hyperlipidemia in terms of regulating gut microbiota, and provided suggestions for regulating gut microbiota through probiotic interventions to improve lipid metabolism.
Collapse
Affiliation(s)
- Jun Wang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| | - Aijie Liu
- ClassyKiss Dairy (Shenzhen) Co., Ltd, China
| | - Ao Li
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| | - Hainan Song
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| | | | - Meng Zhan
- ClassyKiss Dairy (Shenzhen) Co., Ltd, China
| | | | - Lihao Chen
- ClassyKiss Dairy (Shenzhen) Co., Ltd, China
| | - Jiachao Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| | - Ruimin Wang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| |
Collapse
|
6
|
Liu Z, Wang Q, Ma A, Feng S, Chung D, Zhao J, Ma Q, Liu B. Inference of disease-associated microbial gene modules based on metagenomic and metatranscriptomic data. Comput Biol Med 2023; 165:107458. [PMID: 37703713 DOI: 10.1016/j.compbiomed.2023.107458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/22/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023]
Abstract
The identification of microbial characteristics associated with diseases is crucial for disease diagnosis and therapy. However, the presence of heterogeneity, high dimensionality, and large amounts of microbial data presents tremendous challenges in discovering key microbial features. In this paper, we present IDAM, a novel computational method for inferring disease-associated gene modules from metagenomic and metatranscriptomic data. This method integrates gene context conservation (uber-operons) and regulatory mechanisms (gene co-expression patterns) within a mathematical graph model to explore gene modules associated with specific diseases. It alleviates reliance on prior meta-data. We applied IDAM to publicly available datasets from inflammatory bowel disease, melanoma, type 1 diabetes mellitus, and irritable bowel syndrome. The results demonstrated the superior performance of IDAM in inferring disease-associated characteristics compared to existing popular tools. Furthermore, we showcased the high reproducibility of the gene modules inferred by IDAM using independent cohorts with inflammatory bowel disease. We believe that IDAM can be a highly advantageous method for exploring disease-associated microbial characteristics. The source code of IDAM is freely available at https://github.com/OSU-BMBL/IDAM, and the web server can be accessed at https://bmblx.bmi.osumc.edu/idam/.
Collapse
Affiliation(s)
- Zhaoqian Liu
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China
| | - Qi Wang
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China
| | - Anjun Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Shaohong Feng
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, 43210, USA
| | - Jing Zhao
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, 43210, USA.
| | - Bingqiang Liu
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China; Shandong National Center for Applied Mathematics, Jinan, Shandong, 250100, China.
| |
Collapse
|
7
|
Wu Y, Jha R, Li A, Liu H, Zhang Z, Zhang C, Zhai Q, Zhang J. Probiotics (Lactobacillus plantarum HNU082) Supplementation Relieves Ulcerative Colitis by Affecting Intestinal Barrier Functions, Immunity-Related Gene Expression, Gut Microbiota, and Metabolic Pathways in Mice. Microbiol Spectr 2022; 10:e0165122. [PMID: 36321893 PMCID: PMC9769980 DOI: 10.1128/spectrum.01651-22] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022] Open
Abstract
Probiotics can effectively improve ulcerative colitis (UC), but the mechanism is still unclear. Here, shotgun metagenome and transcriptome analyses were performed to explore the therapeutic effect and the mechanism of the probiotic Lactobacillus plantarum HNU082 (Lp082) on UC. The results showed that Lp082 treatment significantly ameliorated dextran sulfate sodium (DSS)-induced UC in mice, which was manifested as increases in body weight, water intake, food intake, and colon length and decreases in disease activity index (DAI), immune organ index, inflammatory factors, and histopathological scores after Lp082 intake. An in-depth study discovered that Lp082 could improve the intestinal mucosal barrier and relieve inflammation by cooptimizing the biological barrier, chemical barrier, mechanical barrier, and immune barrier. Specifically, Lp082 rebuilt the biological barrier by regulating the intestinal microbiome and increasing the production of short-chain fatty acids (SCFAs). Lp082 improved the chemical barrier by reducing intercellular cell adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule (VCAM) and increasing goblet cells and mucin2. Lp082 ameliorated the mechanical barrier by increasing zonula occludens-1 (ZO-1), zonula occludens-2 (ZO-2), and occludin while decreasing claudin-1 and claudin-2. Lp082 optimized the immune barrier by reducing the content of interleukin-1β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α), myeloperoxidase (MPO), and interferon-γ (IFN-γ) and increasing IL-10, transforming growth factor-β1 (TGF-β1), and TGF-β2, inhibiting the NF-κB signaling pathway. Taken together, probiotic Lp082 can play a protective role in a DSS-induced colitis mouse model by protecting the intestinal mucosal barrier, attenuating the inflammatory response, and regulating microbial imbalance. This study provides support for the development of probiotic-based microbial products as an alternative treatment strategy for UC. IMPORTANCE Many studies have focused on the therapeutic effect of probiotics on ulcerative colitis (UC), but few studies have paid attention to the mechanism of probiotics, especially the therapeutic effect. This study suggests that Lp082 has a therapeutic effect on colitis in mice. Its mechanisms of action include protecting the mucosal barrier and actively modulating the gut microbiome, modulating inflammatory pathways, and reducing neutrophil infiltration. Our study enriches the mechanism and provides a new prospect for probiotics in the treatment of colitis, helps to deepen the understanding of the intestinal mucosal barrier, and provides guidance for the future probiotic treatment of human colitis.
Collapse
Affiliation(s)
- Yuqing Wu
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
| | - Rajesh Jha
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Ao Li
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
| | - Huanwei Liu
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
| | - Zeng Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
| | - Chengcheng Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qixiao Zhai
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jiachao Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
- One Health Institute, Hainan University, Haikou, China
| |
Collapse
|
8
|
Chung HC, Gaynanova I, Ni Y. Phylogenetically informed Bayesian truncated copula graphical models for microbial association networks. Ann Appl Stat 2022. [DOI: 10.1214/21-aoas1598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
| | | | - Yang Ni
- Department of Statistics, Texas A&M University
| |
Collapse
|
9
|
Jin Z, Kang J, Yu T. Feature selection and classification over the network with missing node observations. Stat Med 2022; 41:1242-1262. [PMID: 34816464 PMCID: PMC9773124 DOI: 10.1002/sim.9267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/14/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022]
Abstract
Jointly analyzing transcriptomic data and the existing biological networks can yield more robust and informative feature selection results, as well as better understanding of the biological mechanisms. Selecting and classifying node features over genome-scale networks has become increasingly important in genomic biology and genomic medicine. Existing methods have some critical drawbacks. The first is they do not allow flexible modeling of different subtypes of selected nodes. The second is they ignore nodes with missing values, very likely to increase bias in estimation. To address these limitations, we propose a general modeling framework for Bayesian node classification (BNC) with missing values. A new prior model is developed for the class indicators incorporating the network structure. For posterior computation, we resort to the Swendsen-Wang algorithm for efficiently updating class indicators. BNC can naturally handle missing values in the Bayesian modeling framework, which improves the node classification accuracy and reduces the bias in estimating gene effects. We demonstrate the advantages of our methods via extensive simulation studies and the analysis of the cutaneous melanoma dataset from The Cancer Genome Atlas.
Collapse
Affiliation(s)
| | - Jian Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Tianwei Yu
- School of Data Science and Warshel Institute, The Chinese University of Hong Kong - Shenzhen, and Shenzhen Research Institute of Big Data, Shenzhen, China
| |
Collapse
|
10
|
Cardinale S, Kadarmideen HN. Host Genome-Metagenome Analyses Using Combinatorial Network Methods Reveal Key Metagenomic and Host Genetic Features for Methane Emission and Feed Efficiency in Cattle. Front Genet 2022; 13:795717. [PMID: 35281842 PMCID: PMC8905538 DOI: 10.3389/fgene.2022.795717] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/10/2022] [Indexed: 12/22/2022] Open
Abstract
Cattle production is one of the key contributors to global warming due to methane emission, which is a by-product of converting feed stuff into milk and meat for human consumption. Rumen hosts numerous microbial communities that are involved in the digestive process, leading to notable amounts of methane emission. The key factors underlying differences in methane emission between individual animals are due to, among other factors, both specific enrichments of certain microbial communities and host genetic factors that influence the microbial abundances. The detection of such factors involves various biostatistical and bioinformatics methods. In this study, our main objective was to reanalyze a publicly available data set using our proprietary Synomics Insights platform that is based on novel combinatorial network and machine learning methods to detect key metagenomic and host genetic features for methane emission and residual feed intake (RFI) in dairy cattle. The other objective was to compare the results with publicly available standard tools, such as those found in the microbiome bioinformatics platform QIIME2 and classic GWAS analysis. The data set used was publicly available and comprised 1,016 dairy cows with 16S short read sequencing data from two dairy cow breeds: Holstein and Nordic Reds. Host genomic data consisted of both 50 k and 150 k SNP arrays. Although several traits were analyzed by the original authors, here, we considered only methane emission as key phenotype for associating microbial communities and host genetic factors. The Synomics Insights platform is based on combinatorial methods that can identify taxa that are differentially abundant between animals showing high or low methane emission or RFI. Focusing exclusively on enriched taxa, for methane emission, the study identified 26 order-level taxa that combinatorial networks reported as significantly enriched either in high or low emitters. Additionally, a Z-test on proportions found 21/26 (81%) of these taxa were differentially enriched between high and low emitters (p value <.05). In particular, the phylum of Proteobacteria and the order Desulfovibrionales were found enriched in high emitters while the order Veillonellales was found to be more abundant in low emitters as previously reported for cattle (Wallace et al., 2015). In comparison, using the publicly available tool ANCOM only the order Methanosarcinales could be identified as differentially abundant between the two groups. We also investigated a link between host genome and rumen microbiome by applying our Synomics Insights platform and comparing it with an industry standard GWAS method. This resulted in the identification of genetic determinants in cows that are associated with changes in heritable components of the rumen microbiome. Only four key SNPs were found by both our platform and GWAS, whereas the Synomics Insights platform identified 1,290 significant SNPs that were not found by GWAS. Gene Ontology (GO) analysis found transcription factor as the dominant biological function. We estimated heritability of a core 73 taxa from the original set of 150 core order-level taxonomies and showed that some species are medium to highly heritable (0.25–0.62), paving the way for selective breeding of animals with desirable core microbiome characteristics. We identified a set of 113 key SNPs associated with >90% of these core heritable taxonomies. Finally, we have characterized a small set (<10) of SNPs strongly associated with key heritable bacterial orders with known role in methanogenesis, such as Desulfobacterales and Methanobacteriales.
Collapse
Affiliation(s)
- Stefano Cardinale
- Synomics Ltd, Hanborough Business Park, Long Hanborough, United Kingdom
| | | |
Collapse
|
11
|
Ramkumar M, Basker N, Pradeep D, Prajapati R, Yuvaraj N, Arshath Raja R, Suresh C, Vignesh R, Barakkath Nisha U, Srihari K, Alene A. Healthcare Biclustering-Based Prediction on Gene Expression Dataset. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2263194. [PMID: 35265709 PMCID: PMC8901349 DOI: 10.1155/2022/2263194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/02/2022] [Accepted: 02/10/2022] [Indexed: 12/20/2022]
Abstract
In this paper, we develop a healthcare biclustering model in the field of healthcare to reduce the inconveniences linked to the data clustering on gene expression. The present study uses two separate healthcare biclustering approaches to identify specific gene activity in certain environments and remove the duplication of broad gene information components. Moreover, because of its adequacy in the problem where populations of potential solutions allow exploration of a greater portion of the research area, machine learning or heuristic algorithm has become extensively used for healthcare biclustering in the field of healthcare. The study is evaluated in terms of average match score for nonoverlapping modules, overlapping modules through the influence of noise for constant bicluster and additive bicluster, and the run time. The results show that proposed FCM blustering method has higher average match score, and reduced run time proposed FCM than the existing PSO-SA and fuzzy logic healthcare biclustering methods.
Collapse
Affiliation(s)
- M. Ramkumar
- Department of Computer Science and Engineering, HKBK College of Engineering, India
| | - N. Basker
- Department of Computer Science and Engineering, Sona College of Technology, India
| | - D. Pradeep
- Department of Computer Science and Engineering, M.Kumarasamy College of Engineering, Karur, India
| | - Ramesh Prajapati
- Department of Computer Engineering, Shree Swaminarayan Institute of Technology (SSIT), India
| | - N. Yuvaraj
- Research and Publications, ICT Academy, IIT Madras Research Park, India
| | - R. Arshath Raja
- Research and Publications, ICT Academy, IIT Madras Research Park, India
| | - C. Suresh
- CSE, Sri Ranganathar Institute of Engineering and Technology, Coimbatore, India
| | - Rahul Vignesh
- CSE, Dhanalakshmi Srinivasan College of Engineering, Coimbatore, India
| | - U. Barakkath Nisha
- IT Department, Sri Krishna College of Engineering and Technology, Coimbatore, India
| | - K. Srihari
- Department of Computer Science and Engineering, SNS College of Technology, India
| | - Assefa Alene
- Department of Chemical Engineering, College of Biological and Chemical Engineering, Addis Ababa Science and Technology University, Ethiopia
| |
Collapse
|
12
|
Hintikka J, Lensu S, Mäkinen E, Karvinen S, Honkanen M, Lindén J, Garrels T, Pekkala S, Lahti L. Xylo-Oligosaccharides in Prevention of Hepatic Steatosis and Adipose Tissue Inflammation: Associating Taxonomic and Metabolomic Patterns in Fecal Microbiomes with Biclustering. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:4049. [PMID: 33921370 PMCID: PMC8068902 DOI: 10.3390/ijerph18084049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/26/2021] [Accepted: 04/08/2021] [Indexed: 12/15/2022]
Abstract
We have shown that prebiotic xylo-oligosaccharides (XOS) increased beneficial gut microbiota (GM) and prevented high fat diet-induced hepatic steatosis, but the mechanisms associated with these effects are not clear. We studied whether XOS affects adipose tissue inflammation and insulin signaling, and whether the GM and fecal metabolome explain associated patterns. XOS was supplemented or not with high (HFD) or low (LFD) fat diet for 12 weeks in male Wistar rats (n = 10/group). Previously analyzed GM and fecal metabolites were biclustered to reduce data dimensionality and identify interpretable groups of co-occurring genera and metabolites. Based on our findings, biclustering provides a useful algorithmic method for capturing such joint signatures. On the HFD, XOS-supplemented rats showed lower number of adipose tissue crown-like structures, increased phosphorylation of AKT in liver and adipose tissue as well as lower expression of hepatic miRNAs. XOS-supplemented rats had more fecal glycine and less hypoxanthine, isovalerate, branched chain amino acids and aromatic amino acids. Several bacterial genera were associated with the metabolic signatures. In conclusion, the beneficial effects of XOS on hepatic steatosis involved decreased adipose tissue inflammation and likely improved insulin signaling, which were further associated with fecal metabolites and GM.
Collapse
Affiliation(s)
- Jukka Hintikka
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
| | - Sanna Lensu
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
- Department of Psychology, University of Jyväskylä, FI-40014 Jyväskylä, Finland
| | - Elina Mäkinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
| | - Sira Karvinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
| | - Marjaana Honkanen
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
| | - Jere Lindén
- Veterinary Pathology and Parasitology and Finnish Centre for Laboratory Animal Pathology/HiLIFE, University of Helsinki, FIN-00014 Helsinki, Finland;
| | - Tim Garrels
- Department of Computing, University of Turku, FI-20014 Turku, Finland; (T.G.); (L.L.)
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
- Department of Clinical Microbiology, Turku University Hospital, FI-20521 Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, FI-20014 Turku, Finland; (T.G.); (L.L.)
| |
Collapse
|
13
|
Marcos-Zambrano LJ, Karaduzovic-Hadziabdic K, Loncar Turukalo T, Przymus P, Trajkovik V, Aasmets O, Berland M, Gruca A, Hasic J, Hron K, Klammsteiner T, Kolev M, Lahti L, Lopes MB, Moreno V, Naskinova I, Org E, Paciência I, Papoutsoglou G, Shigdel R, Stres B, Vilne B, Yousef M, Zdravevski E, Tsamardinos I, Carrillo de Santa Pau E, Claesson MJ, Moreno-Indias I, Truu J. Applications of Machine Learning in Human Microbiome Studies: A Review on Feature Selection, Biomarker Identification, Disease Prediction and Treatment. Front Microbiol 2021; 12:634511. [PMID: 33737920 PMCID: PMC7962872 DOI: 10.3389/fmicb.2021.634511] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
The number of microbiome-related studies has notably increased the availability of data on human microbiome composition and function. These studies provide the essential material to deeply explore host-microbiome associations and their relation to the development and progression of various complex diseases. Improved data-analytical tools are needed to exploit all information from these biological datasets, taking into account the peculiarities of microbiome data, i.e., compositional, heterogeneous and sparse nature of these datasets. The possibility of predicting host-phenotypes based on taxonomy-informed feature selection to establish an association between microbiome and predict disease states is beneficial for personalized medicine. In this regard, machine learning (ML) provides new insights into the development of models that can be used to predict outputs, such as classification and prediction in microbiology, infer host phenotypes to predict diseases and use microbial communities to stratify patients by their characterization of state-specific microbial signatures. Here we review the state-of-the-art ML methods and respective software applied in human microbiome studies, performed as part of the COST Action ML4Microbiome activities. This scoping review focuses on the application of ML in microbiome studies related to association and clinical use for diagnostics, prognostics, and therapeutics. Although the data presented here is more related to the bacterial community, many algorithms could be applied in general, regardless of the feature type. This literature and software review covering this broad topic is aligned with the scoping review methodology. The manual identification of data sources has been complemented with: (1) automated publication search through digital libraries of the three major publishers using natural language processing (NLP) Toolkit, and (2) an automated identification of relevant software repositories on GitHub and ranking of the related research papers relying on learning to rank approach.
Collapse
Affiliation(s)
- Laura Judith Marcos-Zambrano
- Computational Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute, Madrid, Spain
| | | | | | - Piotr Przymus
- Faculty of Mathematics and Computer Science, Nicolaus Copernicus University, Toruń, Poland
| | - Vladimir Trajkovik
- Faculty of Computer Science and Engineering, Ss. Cyril and Methodius University, Skopje, North Macedonia
| | - Oliver Aasmets
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Magali Berland
- Université Paris-Saclay, INRAE, MGP, Jouy-en-Josas, France
| | - Aleksandra Gruca
- Department of Computer Networks and Systems, Silesian University of Technology, Gliwice, Poland
| | - Jasminka Hasic
- University Sarajevo School of Science and Technology, Sarajevo, Bosnia and Herzegovina
| | - Karel Hron
- Department of Mathematical Analysis and Applications of Mathematics, Palacký University, Olomouc, Czechia
| | | | - Mikhail Kolev
- South West University “Neofit Rilski”, Blagoevgrad, Bulgaria
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Marta B. Lopes
- NOVA Laboratory for Computer Science and Informatics (NOVA LINCS), FCT, UNL, Caparica, Portugal
- Centro de Matemática e Aplicações (CMA), FCT, UNL, Caparica, Portugal
| | - Victor Moreno
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO)Barcelona, Spain
- Colorectal Cancer Group, Institut de Recerca Biomedica de Bellvitge (IDIBELL), Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Irina Naskinova
- South West University “Neofit Rilski”, Blagoevgrad, Bulgaria
| | - Elin Org
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| | - Inês Paciência
- EPIUnit – Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
| | | | - Rajesh Shigdel
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Blaz Stres
- Group for Microbiology and Microbial Biotechnology, Department of Animal Science, University of Ljubljana, Ljubljana, Slovenia
| | - Baiba Vilne
- Bioinformatics Research Unit, Riga Stradins University, Riga, Latvia
| | - Malik Yousef
- Department of Information Systems, Zefat Academic College, Zefat, Israel
- Galilee Digital Health Research Center (GDH), Zefat Academic College, Zefat, Israel
| | - Eftim Zdravevski
- Faculty of Computer Science and Engineering, Ss. Cyril and Methodius University, Skopje, North Macedonia
| | | | | | - Marcus J. Claesson
- School of Microbiology & APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Isabel Moreno-Indias
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jaak Truu
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| |
Collapse
|