Inquiry
Microbiome Network Analysis: Unveiling Interactions and Functional Dynamics in Microbial Communities

Microbiome Network Analysis: Unveiling Interactions and Functional Dynamics in Microbial Communities

Online Inquiry

What is Microbiome Network Analysis?

A microbiome encompasses the comprehensive assemblage of microorganisms, encompassing bacteria, fungi, viruses, and various microbial entities, that populate a designated ecological milieu. Within this intricate network, these microorganisms engage in intricate interplay, both amongst themselves and with the host, exerting profound influences on a myriad of biological processes and profoundly contributing to the overall stability of the ecosystem in question. Notably, the human microbiome, an intricately structured consortium of trillions of microorganisms, occupies a central role in modulating human health and disease outcomes. The advent of cutting-edge high-throughput sequencing technologies has ushered in a paradigm shift in microbiome research, enabling in-depth exploration of microbial communities and elucidation of their functional dynamics.

Microbiome network analysis plays a pivotal role in deciphering the intricate web of interactions between microorganisms by constructing and analyzing complex networks. In a microbiome network, nodes represent individual microbial taxa or species, and edges represent interactions between them. These interactions can be positive (e.g., cooperation, mutualism) or negative (e.g., competition, predation), and they play a vital role in shaping the structure and dynamics of the microbial community.

Taxonomic profiling of bacteria, fungi and the virome.Taxonomic profiling of bacteria, fungi and the virome. (Matchado et al., 2021)

Once the microbiome network is constructed, various network analysis techniques can be applied to gain insights into its structure and properties. You can read our article Network Analysis in Biology for basic knowledge of biological network analysis.

What can microbiome network analysis do?

Microbiome network analysis allows for the integration of additional data types, such as metagenomics or metatranscriptomics, to infer functional relationships between microorganisms and explore their metabolic potential. By combining information on microbial interactions, community structure, and functional profiles, researchers can gain a deeper understanding of the roles and contributions of specific microorganisms in the microbiome ecosystem.

Methods and Tools for Microbiome Network Analysis

Data Preprocessing

Microbiome network analysis begins with the acquisition of high-quality sequencing data, obtained through state-of-the-art technologies such as 16S rRNA gene sequencing or shotgun metagenomics. These data undergo rigorous preprocessing steps, including quality control, adapter trimming, error correction, and read filtering. Advanced tools and algorithms like FastQC, Trimmomatic, DADA2, or KneadData are employed for efficient data preprocessing, ensuring the reliability and accuracy of subsequent analyses.

For more information about How to build and analyze biological networks?

Taxonomic Profiling and Abundance Quantification

Taxonomic profiling assigns taxonomic labels to sequencing reads, enabling identification and characterization of microbial taxa present in the community. Advanced algorithms and databases such as SILVA, Greengenes, or NCBI's RefSeq facilitate accurate taxonomic classification. Subsequently, relative abundance estimation is performed to quantify the presence and abundance of microbial taxa. Tools like QIIME, mothur, or MetaPhlAn leverage statistical models and reference databases for robust taxonomic profiling and abundance quantification.

Microbiome Network Construction

Once the preprocessed data is available, the construction of microbiome networks involves quantifying the relationships between microbial taxa. Two common approaches are co-occurrence networks and co-abundance networks.

a. Co-occurrence Networks

Co-occurrence networks focus on identifying taxa that tend to co-occur together across samples, indicating potential ecological interactions. Statistical measures such as correlation coefficients (e.g., Pearson's, Spearman's) or mutual information are utilized to quantify the strength of associations. Advanced algorithms including SPIEC-EASI, CoNet, or WGCNA enable the construction of robust co-occurrence networks, providing insights into microbial community structure and dynamics.

b. Co-abundance Networks

Co-abundance networks highlight microbial taxa that exhibit similar abundance patterns across samples, implying potential functional relationships. These networks shed light on cooperative or competitive interactions among taxa. Cutting-edge tools like SPIEC-EASI, CoNet, or MINT facilitate the construction of co-abundance networks, facilitating deeper understanding of functional dynamics within microbial communities.

Overview of network approaches for microbial intra- and inter-kingdom interactions.Overview of network approaches for microbial intra- and inter-kingdom interactions. (Matchado et al., 2021)

Network Topology Analysis

Once the networks are constructed, various network topology measures are employed to characterize the structure and organization of the microbiome networks. These measures provide insights into the connectivity, centrality, and modularity of the network.

Network topology analysis of predicted gene network.Network topology analysis of predicted gene network. (Shamloo-Dashtpagerdi et al., 2019)

a. Degree Centrality

Degree centrality quantifies the number of connections (edges) associated with a particular node (microbial taxon). High-degree nodes (hubs) are considered crucial for maintaining network stability and are often associated with keystone species. Tools like NetworkX and igraph provide functions to calculate degree centrality.

b. Clustering Coefficient

The clustering coefficient quantifies the extent to which nodes tend to form clusters or communities within the network. It measures the density of connections among a node's neighbors, elucidating the presence of tightly connected groups of taxa. Employing powerful algorithms within NetworkX, igraph, or Cytoscape, the clustering coefficient can be accurately calculated, providing insights into the modular organization of microbial communities.

c. Betweenness Centrality

Betweenness centrality identifies nodes that act as bridges or mediators within the network. It quantifies the extent to which a node lies on the shortest paths between other pairs of nodes. High betweenness centrality nodes play a critical role in maintaining network connectivity. NetworkX and igraph provide functions to calculate betweenness centrality.

d. Modularity

Modularity measures the presence of densely connected modules or communities within the network. It quantifies the extent to which the network can be partitioned into distinct groups of taxa. Higher modularity values indicate stronger within-module connections and weaker between-module connections. Cutting-edge community detection algorithms such as Louvain, Infomap, or Walktrap, implemented in NetworkX, igraph, or Cytoscape, facilitate the identification of modules within microbiome networks, unraveling functional substructures and potential ecological niches.

Biological Interpretation

Microbiome network analysis is not solely limited to the construction and characterization of networks. Integration of additional metadata, such as clinical outcomes, dietary information, or functional annotations, can provide a deeper understanding of the biological significance of the network.

a. Module Identification and Functional Annotation

Modules identified within the network represent groups of taxa that potentially share similar functions or ecological niches. Functional annotations can be assigned to these modules by integrating information from databases such as KEGG (Kyoto Encyclopedia of Genes and Genomes) or MetaCyc. Functional enrichment analysis tools, such as LEfSe (Linear discriminant analysis Effect Size) or PICRUSt (Phylogenetic Investigation of Communities by Reconstruction of Unobserved States), can also be utilized to identify significantly enriched functional pathways within the modules.

b. Integration with Host Data

Integrating microbiome network analysis with host data, such as host gene expression or clinical metadata, allows for a comprehensive understanding of the host-microbiome interactions. Tools like MaAsLin2 (Multivariate Association with Linear Models) and MixMC (Mixtures of Multiple Correspondence Analysis) can be employed for the integration of multi-omics data. Visualizing microbiome networks is crucial for comprehending complex interactions and facilitating biological interpretations. Tools such as Cytoscape, Gephi, or iGraph provide powerful visualization capabilities to explore network structures, node attributes, and edge properties.

Dynamic Network Analysis

Microbiome networks are dynamic and can exhibit temporal variations influenced by factors like diet, host genetics, and environmental changes. Analyzing microbiome dynamics provides insights into community stability, resilience, and response to perturbations.

General steps for the execution of dynamic network inference.General steps for the execution of dynamic network inference. (Garcia J et al., 2020)

a. Time-Series Analysis

Time-series data, collected at multiple time points, enable the study of microbial community dynamics over time. Sophisticated statistical methods like dynamic Bayesian networks, state-space models, or ARIMA models capture the temporal relationships and fluctuations within microbiome networks. Tools such as WGCNA, iTAS, or GeneNet provide advanced functionalities for analyzing time-series data, identifying temporal patterns, and inferring dynamic microbial interactions.

b. Differential Network Analysis

Differential network analysis compares network structures between different conditions or time points, revealing variations in microbial interactions. Innovative tools such as ALDEx2, DiffCoEx, or CCREPE leverage statistical modeling and differential abundance analysis to identify condition-specific or temporally regulated microbial interactions. These approaches contribute to a deeper understanding of the functional changes and adaptation mechanisms within microbial communities.

Challenges and Future Perspectives in Microbiome Network Analysis

Microbiome network analysis poses several challenges and offers exciting avenues for future research.

Statistical Inference in Microbiome Networks

Microbiome network analysis involves addressing statistical challenges inherent in high-dimensional, compositional, and sparse microbiome data. Developing robust statistical models and inference methods that consider these unique characteristics is crucial for accurate network construction and interpretation.

Multi-Omics Integration

Integrating microbiome data with other omics data, such as metabolomics or transcriptomics, provides a comprehensive understanding of host-microbiome interactions. Advancements in multi-omics integration methods and tools will facilitate a holistic view of the complex interplay between different biological layers.

Causal Inference

Unraveling causal relationships within microbiome networks is a challenging yet essential task. Advancements in causal inference methodologies, such as causal graphical models or causal discovery algorithms, will enable the identification of key drivers and mechanisms underlying microbial interactions.

Translational Applications

Translating microbiome network analysis into clinical practice requires the development of robust biomarkers, predictive models, and therapeutic interventions. Future research should focus on identifying network-based signatures associated with specific diseases or treatment responses, facilitating personalized medicine approaches.

References

  1. Matchado, Monica Steffi, et al. "Network analysis methods for studying microbial communities: A mini review." Computational and structural biotechnology journal 19 (2021): 2687-2698.
  2. Shamloo-Dashtpagerdi, Roohollah, et al. "LOS2 gene plays a potential role in barley (Hordeum vulgare L.) salinity tolerance as a hub gene." Molecular breeding 39.8 (2019): 119.
  3. Garcia J, Kao‐Kniffin J. Can dynamic network modelling be used to identify adaptive microbiomes?[J]. Functional Ecology, 2020, 34(10): 2065-2074.
* For Research Use Only. Not for use in diagnostic procedures.
Online Inquiry