Open access
Host-Microbial Interactions
Research Article
1 April 2024

Decoding host-microbiome interactions through co-expression network analysis within the non-human primate intestine

ABSTRACT

The gut microbiome affects the health status of the host through complex interactions with the host’s intestinal wall. These host-microbiome interactions may spatially vary along the physical and chemical environment of the intestine, but these changes remain unknown. This study investigated these intricate relationships through a gene co-expression network analysis based on dual transcriptome profiling of different intestinal sites—cecum, transverse colon, and rectum—of the primate common marmoset. We proposed a gene module extraction algorithm based on the graph theory to find tightly interacting gene modules of the host and the microbiome from a vast co-expression network. The 27 gene modules identified by this method, which include both host and microbiome genes, not only produced results consistent with previous studies regarding the host-microbiome relationships, but also provided new insights into microbiome genes acting as potential mediators in host-microbiome interplays. Specifically, we discovered associations between the host gene FBP1, a cancer marker, and polysaccharide degradation-related genes (pfkA and fucI) coded by Bacteroides vulgatus, as well as relationships between host B cell-specific genes (CD19, CD22, CD79B, and PTPN6) and a tryptophan synthesis gene (trpB) coded by Parabacteroides distasonis. Furthermore, our proposed module extraction algorithm surpassed existing approaches by successfully defining more functionally related gene modules, providing insights for understanding the complex relationship between the host and the microbiome.

IMPORTANCE

We unveiled the intricate dynamics of the host-microbiome interactions along the colon by identifying closely interacting gene modules from a vast gene co-expression network, constructed based on simultaneous profiling of both host and microbiome transcriptomes. Our proposed gene module extraction algorithm, designed to interpret inter-species interactions, enabled the identification of functionally related gene modules encompassing both host and microbiome genes, which was challenging with conventional modularity maximization algorithms. Through these identified gene modules, we discerned previously unrecognized bacterial genes that potentially mediate in known relationships between host genes and specific bacterial species. Our findings underscore the spatial variations in host-microbiome interactions along the colon, rather than displaying a uniform pattern throughout the colon.

INTRODUCTION

The mutual interaction between the intestinal microbiome and the host constitutes a complex biological process, with intertwined dependencies that are essential for the survival and health of both. The interactions between the intestinal microbiome and the host are formed by various factors such as the effects of metabolites produced by bacteria on the host, the binding of bacterial-derived proteins and host receptors, and the binding of bacterial cell surface structures and host receptors. Studies on host-microbiome interaction through bacterial-derived proteins have discovered that a novel protein P9 secreted by Akkermansia muciniphila binds to host ICAM-2, thereby increasing the secretion of glucagon-like peptide 1 (GLP-1), suggesting that the interaction between P9 and ICAM-2 could be a potential therapeutic target in type 2 diabetes patients (1). Excluding certain Gram-negative bacteria, it has also been reported that bacterial lipopolysaccharides, which are components of the Gram-negative bacterial cell wall, bind to the Toll-like receptor 4 complex and can induce innate immune responses (2). Among these interactions, the host-microbiome interaction mediated by metabolites is particularly well studied. For instance, primary bile acids, produced in the host’s liver cells, are converted into secondary bile acids by the microbiome in the colon. This microbiome-derived secondary bile acid is known to regulate the homeostasis of regulatory T cells, which are immune-suppressive cells in the host (3). Through these processes, the intestinal microbiome and host mutually interact and establish a symbiotic relationship while forming a metabolic pathway or physiological response.
To understand such complex interactions between the host and the microbiome, it is beneficial to analyze combined profiles of both the microbiome and the host. Recent studies have reported on dual profiling of host and microbiome gene expression levels to understand the interactions between the host and microbiome (46). Previously, the focus was primarily on identifying bacterial species related to host phenotypes (such as diseases) based on the relative abundance of microbiome (7). However, profiling both host and microbiome gene expression levels has provided further insights into which genes of which bacterial species are related to host physiological functions and phenotypes. Gene co-expression network analysis is one of the useful approaches for comprehensively deciphering complex interactions from the vast number of gene expression profiles of the host and microbiome. A gene co-expression network is constructed by representing genes as nodes and connecting genes with correlated expression levels via edges. Representing gene relationships as a network enables the extraction of closely interconnected gene modules (subgraphs) and the identification of hub genes connected to multiple genes, thereby enhancing interpretability. For instance, a study (5) characterizing the host-microbiome interaction during influenza virus infection identified modules from the host’s gene co-expression network. It discovered correlations between host gene modules involved in interferon signal transduction and the sulfonamide antibiotic resistance genes of the microbiome. In another study (6), network analysis was applied to the gene expression profiles of the host and microbiome in the nasopharyngeal airway during bronchiolitis. Networks were constructed for both the host and microbiome and modules were identified from these networks. The study revealed that host modules were involved in regulating host-T cell activity. Additionally, microbiome modules involved in microbial-branched-chain amino acid metabolism and oxidative stress response were associated with disease severity.
Despite such progress, several limitations remain in understanding the interactions between the host and the intestinal microbiome. First, there’s a lack of understanding of the spatial variation in host-microbiome interactions. The colon, rich in microbiota, can be subdivided into the cecum, ascending colon, transverse colon, descending colon, sigmoid colon, and rectum. Each section possesses distinct functions and differs in its physicochemical environment, including variations in nutrient content, pH, and luminal flow (810), and the microbiome could vary as well. Indeed, studies using animal models have reported that the composition and functional fluctuation of the microbiome vary due to environmental changes depending on the intestinal location (biogeographical location) (1113). However, these studies focused solely on the microbiome, and the coordinated spatial variation of both the microbiome and host gut cells along the intestinal tract has yet to be clarified.
Second, there is a lack of computational methods for extracting significant modules (subgraphs) from the host-microbiome gene co-expression network. Analyzing a massive gene co-expression network integrating the gene expression profiles of both the host and microbiome helps interpret the interactions between the genes of the host and the microbiome. Especially in a huge gene co-expression network, it is crucial to extract densely connected subgraphs (modules) to divide the network into understandable units; however, existing algorithms for extracting modules from the network are insufficient for accurately capturing the interaction between the host and microbiome because they were not designed with the purpose of understanding interactions between organisms (1417).
Lastly, there are concerns regarding the applicability of the results of studies using animal models to humans. While mouse models have been the basis of biological research for a long time due to ethical constraints, experiments in controlled environments, and considerations of reproducibility, there are several issues and limitations to their use. The physiology, anatomy, and pharmacology of mice differ from those of humans, and there are differences in the composition and function of the intestinal microbiome between mice and humans (13, 18). Particularly considering the complexity of host-microbiome interactions, it is challenging to precisely replicate the relationship between humans and their intestinal microbiome. In contrast, the common marmoset, a small New World primate, is considered a useful model in preclinical trials because it is genetically close to humans and has common physiological and anatomical characteristics (19). Moreover, the common marmoset is the only non-human primate in which a germ-free state has been successfully achieved (20), and it has a fecal microbiota similar to that of humans compared with other animal models, including mice (13), potentially broadening the scope of intestinal microbiome research.
To address these challenges, this study aimed to reveal the interactions between the host and the microbiome along multiple sites of the common marmoset colon—cecum, transverse colon, and rectum—by developing a module extraction method applicable to the gene co-expression network that represents the complex interactions between the host and the microbiome. Through simultaneous profiling of host and microbiome transcripts in the intestinal sites of the common marmoset, we constructed an integrated gene co-expression network, incorporating both host and microbiome genes. From this network, we further identified densely covariant gene modules (subgraphs) to interpret the interactions between genes. As conventional methods for extracting gene modules from the network were insufficient for interpreting the interactions between organisms examined in this study, we proposed a module extraction algorithm based on the graph theory. Our proposed method successfully extracted more modules containing functionally related genes of both the host and microbiome compared with existing algorithms, underlining its suitability for predicting host-microbiome interactions. This method extracted 27 gene modules containing both the host and microbiome, indicating that the gut microbiome is functionally closely involved in the variation of host gene expression along the colon.

RESULTS

Overview of the analysis pipeline integrating host and microbiome transcriptomes: identification of closely interacting gene modules from gene co-expression networks

To investigate the host and microbiome interactions along the colon, we proposed an analysis method integrating the host transcriptome and the microbiome metatranscriptome (Fig. 1). First, we computed gene expression profiles from the host transcriptome and microbiome metatranscriptome obtained via dual RNA sequencing (RNA-seq) and created three types of gene co-expression networks: host-host, host-microbiome, and microbiome-microbiome. By unifying these three networks, we constructed a host and microbiome gene co-expression network, with the genes of the host and microbiome serving as nodes and covariant genes interconnected. Second, from the gene co-expression network, we identified gene modules containing closely interacting genes from both the host and microbiome. We aimed to interpret the interactions between host genes and microbiome genes within modules extracted from a gene co-expression network, based on the assumption that functionally similar genes are co-expressed (21). Conventional module extraction methods are not designed to understand interspecies interactions (1417) and do not distinguish between host gene and microbiome gene nodes in the network. Consequently, modules composed solely of host genes or exclusively of microbiome genes can be extracted, making it challenging to understand the interactions between the host and the microbiome when such single-species modules are present. Therefore, we proposed a clique finding-based module extraction method that takes into account the structure of three networks: host-host, host-microbiome, and microbiome-microbiome. This method begins by exploring cliques from the gene co-expression network. In graph theory, a “clique” refers to a subgraph within the network where all nodes are fully connected. For overlapping cliques, the method calculates the cliqueness in each of the original three networks and merges them if the cliqueness of all networks exceeds a certain threshold, thereby obtaining the final gene modules (refer to Materials and Methods for details).
Fig 1
Fig 1 Overview of the main analytical approach used in this study. This method involves the simultaneous acquisition of the host and microbiome transcriptome (metatranscriptome) from intestinal sites and conducts a gene co-expression network analysis of the host and microbiome community. The procedure is outlined from steps A to E. (A) Perform dual RNA-seq of host and microbiome transcriptomes in internal site samples. (B) Separate host and microbiome RNA reads in silico using the host reference genome. (C) Compute the gene expression profiles of the host and microbiome, respectively. (D) Unify gene co-expression networks between host-host, host-microbiome, and microbiome-microbiome. The host-host and microbiome-microbiome gene co-expression networks are connected when there is a positive correlation between genes, while the host-microbiome gene co-expression network is connected when there is either a positive or negative correlation between genes. (E) Identify closely interacting gene modules from the gene co-expression network using a proposed clique-based algorithm. (E step 1) This process begins with searching for cliques. (E step 2) If overlaps exist between cliques, the cliqueness score is computed in three gene co-expression networks (host-host, host-microbiome, and microbiome-microbiome) when overlapping cliques are merged. (E step 3) If the cliqueness score exceeds the merging threshold τ in all three networks, the cliques are merged into a single module. If the score does not exceed the merging threshold τ in at least one network, the original cliques are retained without merging. This process is repeated until no further modules can be merged.

Computing gene expression profiles of the host and the microbiome from dual RNA-seq data

First, we extracted high-quality RNA (RNA integrity number [RIN] scores ≥ 8.6; Table S1) from the intestinal sites (cecum, transverse colon, and rectum) of five common marmosets (Supplemental Note Section 1) and carried out dual RNA-seq with NovaSeq 6000 v1.5 (100 bp paired-end). The obtained RNA read data amounted to a total of 4,724 M reads (mean 157,450,033 paired-end reads/sample; Table S2; Supplemental Note Section 2).
Next, as the RNA read data obtained by dual RNA-seq contains a mixture of reads derived from both the host and the microbiome, it is necessary to separate these read data in silico (Table S3; Supplemental Note Section 3). The pipeline established in this study separates the host-derived read data and the microbiome-derived read data by aligning the RNA reads to the host reference genome (Fig. 1B). To avoid transcriptomic cross-contamination of read data between host and microbiome read data, we adjusted the pipeline parameters using simulated RNA read data from the host and the microbiome (Supplemental Note Section 4). As a result, we achieved a precision of 1.0 in separating the read data of the host and the microbiome, and our established pipeline completely separated the reads derived from the host and those from the microbiome (Table S9 posted at https://doi.org/10.5281/zenodo.10425960).
After the above preprocessing, we calculated the gene expression levels (transcripts per million [TPM]) for both the host and the microbiome. As a result, we detected 16,578 host-expressed genes and 8,533 microbiome-expressed genes in total (Supplemental Note Sections 5 and 6). To examine the expression patterns across different sites and across different individuals for both host genes and microbiome genes, we applied principal component analysis (PCA) to the gene expression profiles for visualization. There is a clear separation between the host gene expression profiles for each intestinal site, and the host samples were not grouped based on individuals (Fig. 2A and B). The microbiome profiles at the cecum and rectum, which are the beginning and end of the colon, respectively, were separated by PC1, with samples from the transverse colon, situated between the cecum and rectum, spanning across both sites (Fig. 2C). For the microbiome profiles as well as for the host ones, samples were not separated based on individuals (Fig. 2D). These results suggest that both host and microbiome gene expression profiles are more variable across different sites compared with those across individuals.
Fig 2
Fig 2 PCA analysis of gene expression data for host and microbiome. PCA was applied to the host gene expression profiles to visualize groupings according to site (A) and individual (B). PCA was similarly applied to the microbiome gene expression profiles to visualize groupings according to site (C) and individual (D).

Spatial variations in host gene expression among intestinal sites

Of the 16,578 host genes detected in three regions of the common marmoset’s intestinal tract—cecum, transverse colon, and rectum—a total of 2,615 genes showed significant expression variations among these sites (FDR < 0.01) (Table S4; Supplemental Note Section 5). Furthermore, upon conducting functional pathway analysis, 40 pathways were identified, all of which were significantly upregulated in the cecum compared with the transverse colon (normalized enrichment score > 1.5, FDR < 0.1; Table S10 posted at https://doi.org/10.5281/zenodo.10425960). In particular, pathways with high normalized enrichment scores, such as the Fc epsilon RI signaling pathway, T cell receptor signaling pathway, leukocyte transendothelial migration, Yersinia infection, and B cell receptor signaling pathway, were detected, indicating that pathways related to immune functions were upregulated in the cecum relative to the transverse colon (Fig. 3A). Additionally, a substantial number of genes involved in these functional pathways were found to be significantly upregulated in the cecum (FDR < 0.01). For example, among the genes specifically upregulated in the cecum, we found genes related to B cell receptor signaling. These include CD19, which acts as a coreceptor for the B cell antigen receptor complex and augments B cell activation (22); CD79A and CD79B, components of the B cell antigen receptor that generate signals upon antigen recognition (23); and CD22 and CD72, which inhibit the signal transduction of the B cell antigen receptor (24, 25). In addition, genes associated with T cell receptor signaling were also found as being highly expressed specifically in the cecum. These include CD28, a co-stimulatory receptor for T cell receptors that activates T cells (26); CD3D, a component of CD3 that plays a role in signal transduction during T cell activation (27); CD40LG, which is induced on the T cell surface following T cell receptor (TCR) activation (28); and CTLA4, which attenuates T cell activation and fine tunes immune responses (29) (Fig. 3B).
Fig 3
Fig 3 Variations in host gene expression and functional pathways among intestinal sites. (A) The top 20 functional pathways significantly upregulated in the cecum compared with the transverse colon (normalized enrichment score > 1.5, FDR < 0.1), as revealed by gene set enrichment analysis, are presented alongside their normalized enrichment score and FDR. (B) Heatmap of differentially expressed genes belonging to the top five enriched functional pathways. The heatmap on the left represents the z-scores of gene expression levels, with genes significantly upregulated in the cecum compared with the transverse colon and rectum (FDR < 0.01) colored in pink on the middle heatmap. The heatmap on the right indicates the functional pathways to which the genes belong, represented in green.

Interpreting host-microbiome interactions through spatial gene co-expression network analysis within the intestine

We constructed the unified gene co-expression network composed of 1,359 gene nodes, which embodies co-expression information on host-host, host-microbiome, and microbiome-microbiome interactions (Tables S11 to S13 posted at https://doi.org/10.5281/zenodo.10425960; Supplemental Note Section 7). Subsequently, to identify densely co-expressed gene sets within this network, we implemented the clique-based module extraction algorithm proposed in this study. As a result, we then identified 27 gene modules containing both host and microbiome genes (Tables S14 and S15 posted at https://doi.org/10.5281/zenodo.10425960). Furthermore, upon conducting a functional pathway enrichment analysis on these gene modules, we found that 70.4% (19 out of 27) of gene modules could be functionally characterized (Supplemental Note Section 8).
The modules we identified were characterized by functional pathways including the B cell receptor signaling pathway, fructose and mannose metabolism, oxidative phosphorylation, and biosynthesis of amino acids (Fig. 4). In addition, we provided visualizations of the bacterial species that contribute to microbiome gene expression within these identified modules, together with host and microbiome genes (Fig. 5 through 8).
Fig 4
Fig 4 Functional pathways characterized in modules extracted from the host-microbiome gene co-expression network. This figure displays the identified modules along with the results of their functional pathway enrichment analysis (Supplemental Note Section 8). To enhance clarity, the background of the bar graphs alternates between gray and white for each module. Modules were characterized using KEGG pathways, with those meeting an FDR < 0.05 as determined by Fisher’s exact test. The dashed line represents an FDR < 0.05 [−log10(FDR) > 1.30]. All pathways with an FDR < 0.1 are represented, and modules with no identified function are denoted as “None.”
Fig 5
Fig 5 Metabolic pathway-enriched Module_07. A module showing a positive correlation between the host gene FBP1, which is highly expressed in the transverse colon, and 40 microbiome genes, including those involved in fructose-mannose metabolism and oxidative phosphorylation, was extracted. These genes were visualized alongside bacterial species nodes that contribute to the expression of the microbiome genes. Note that the bacterial species nodes were added after the module detection analysis. Edges among genes indicate an absolute Spearman’s correlation coefficient greater than 0.8. Edges connecting microbiome genes to bacterial species are shown when the contribution rate of bacterial species to microbiome gene expression exceeds 0.2. The thickness of the edge increases with higher contribution rates.
Fig 6
Fig 6 B cell receptor signaling pathway-enriched Module_01. Host genes involved in the immune response of B cells, including CD19, CD22, CD79B, and PTPN6, which were highly expressed in the cecum, co-expressed with the K01696 (trpB). These genes were visualized alongside bacterial species nodes that contribute to the expression of the microbiome genes. K01696 (trpB) expression was contributed to by Parabacteroides distasonis. Edges among genes indicate an absolute Spearman’s correlation coefficient greater than 0.8. Edges connecting microbiome genes to bacterial species are shown when the contribution rate of bacterial species to microbiome gene expression exceeds 0.2. Note that the bacterial species nodes were added after the module detection analysis. The thickness of the edge increases with higher contribution rates.
Fig 7
Fig 7 Carbon metabolism pathway-enriched Module_21. Host genes, including MMP10, which is specifically highly expressed in the cecum, and 40 microbiome genes involved in carbon metabolism, were identified. These genes were visualized alongside bacterial species nodes that contribute to the expression of the microbiome genes. Note that the bacterial species nodes were added after the module detection analysis. Edges among genes indicate an absolute Spearman’s correlation coefficient greater than 0.8. Edges connecting microbiome genes to bacterial species are shown when the contribution rate of bacterial species to microbiome gene expression exceeds 0.2. The thickness of the edge increases with higher contribution rates.
Fig 8
Fig 8 Module_18 containing host genes and microbiome genes related to the production and breakdown of bile acids. These genes were visualized alongside bacterial species nodes that contribute to the expression of the microbiome genes. Note that the bacterial species nodes were added after the module detection analysis. Edges among genes indicate an absolute Spearman’s correlation coefficient greater than 0.8. Edges connecting microbiome genes to bacterial species are shown when the contribution rate of bacterial species to microbiome gene expression exceeds 0.2. The thickness of the edge increases with higher contribution rates.
Module_07, as shown in Fig. 5, was identified as a module with a positive correlation between the host gene fructose-1,6-bisphosphatase 1 (FBP1), which is highly expressed in the transverse colon, and 40 microbiome genes. These 40 gut microbiome genes included those associated with fructose-mannose metabolism, such as K01840 (manB), K00850 (pfkA), and K01818 (fucI), as well as those related to oxidative phosphorylation, such as K00241 (sdhC, frdC), K00426 (cydB), K02112 (atpD), and K02121 (ntpE, atpE). The most substantial contributor to the expression of these microbiome genes was Bacteroides vulgatus.
FBP1 is known to have reduced expression in various tumor tissues, including the esophagus (30), liver (31), lung (32), and ovarian (33) cancer. It has been confirmed that its expression increases with fructose treatment in intestinal organoids (34). Previous studies have identified FBP1 as a differentially expressed gene in the small intestine of germ-free mice, suggesting that the intestinal microbiome influences the expression of the host’s FBP1 gene (35), but the bacterial species and genes affecting the expression of the FBP1 gene have not been identified. Bacteroides vulgatus, which contributed to the expression of fructose-mannose metabolism-related microbiome genes observed in Module_07, has been reported to release fructose by decomposing polysaccharides (36, 37). In this module, the gene K00850 (pfkA) involved in the fructo-oligosaccharide degradation and the gene K01818 (fucI) involved in the 2′-fucosyllactose degradation were detected, revealing a novel association between polysaccharide degradation genes encoded by Bacteroides vulgatus and the host gene FBP1.
In Module_01, the B cell receptor signaling pathway was enriched (Fig. 6). This module featured covariation between host B cell-specific genes, including CD19, CD22, CD79B, and PTPN6, which were highly expressed in the cecum, and the tryptophan synthesis gene K01696 (trpB) from the microbiome. The upregulation of these host B cell-specific genes indicates an increase in B cells within the cecum. This tryptophan synthesis gene K01696 (trpB) expression was contributed to by Parabacteroides distasonis. Tryptophan is an essential amino acid that, upon uptake by intestinal cells and subsequent metabolism, can activate the Aryl hydrocarbon receptor and influence B cell immune responses (38). Tryptophan is also known to ameliorate autoimmune diseases, such as inflammatory bowel disease (39). Parabacteroides distasonis has been suggested to be involved in various autoimmune conditions in several studies (40, 41). For instance, Parabacteroides distasonis levels have been shown to be lower in multiple sclerosis (MS) patients compared with healthy controls, and transplanting the intestinal microbiota from MS patients to germ-free mice resulted in increased severity of autoimmune encephalomyelitis symptoms (40). While previous studies have reported associations between tryptophan and autoimmune diseases via B cell immune responses, as well as between autoimmune diseases and Parabacteroides distasonis, this study identifies a correlation between the tryptophan synthesis gene K01696 (trpB) from Parabacteroides distasonis and B cell-specific genes (CD19, CD22, CD79B, and PTPN6).
In Module_21, a group consisting of three host genes and 40 microbiome genes was identified. The three host genes included MMP10 and two uncharacterized genes LOC103795407 and CCDC162P, all of which showed high expression in the cecum (Fig. 7). The microbiome genes that showed a positive correlation with these genes included K01491 (folD), K00605 (gcvT), K01803 (tpiA), K00831 (serC), K15633 (gpmI), and K00024 (mdh), all of which are involved in carbon metabolism pathways. Bacteroides vulgatus was identified as the bacteria contributing to the expression of these microbiome genes. The MMP10 gene plays a crucial role in maintaining epithelial barrier function and is necessary for resolving colonic epithelial damage (42). A study examining the host transcriptome and microbiota composition of colorectal cancer tumors found high expression of MMP10 in tumor tissues and a significant increase in the genus Bacteroides, indicating a link between the host gene MMP10 and the genus Bacteroides (43). These findings are consistent with our observations. Furthermore, in the identified Module_21, not only the relationship between the host gene MMP10 and Bacteroides vulgatus but also a group of microbiome genes related to carbon metabolism pathways showing a positive correlation with the host gene MMP10 was identified. One hypothesis explaining the observed positive correlation between carbon metabolism-related genes encoded by Bacteroides vulgatus and the host gene MMP10 is that the microbiome may form colonies within the host’s mucosal layer. Mucin, the principal component of mucus secreted by the gastrointestinal mucosal epithelium, contains galactose and promotes bacterial settlement and adhesion (44), serving as a nutrient source for certain bacteria. Bacteroides vulgatus possesses mucosal adherence factors, allowing access to host-derived carbon sources (45). The upregulation of MMP10 in the cecum might represent a protective response against colonization by Bacteroides vulgatus. The detection of carbon metabolism-related genes of Bacteroides vulgatus in module_21 suggests they might be utilizing carbon sources derived from mucin or dietary glucose. Specifically, while Bacteroides vulgatus cannot degrade human mucin independently, it can acquire carbon sources from mucin in co-culture with Akkermansia muciniphila (46), which possesses genes detected in this module. When Akkermansia muciniphila converts galactose to glucose, Bacteroides vulgatus might then activate these carbon metabolism-related genes to utilize this glucose (Fig. S1). Our findings indicate a relationship between the carbon metabolism-related genes of Bacteroides vulgatus and the host gene MMP10; however, further experiments are required to ascertain whether the upregulation of MMP10 serves a protective role against colonization by Bacteroides vulgatus and Akkermansia muciniphila or if Bacteroides vulgatus utilizes mucin as a carbon source.
In Module_18, 3 host genes and 54 microbiome genes were identified, and the expression of these microbiome genes was further influenced by five bacterial species. The relationships between these host genes and microbiome genes, as well as between the host genes and bacterial species, were consistent with existing knowledge about bile acid synthesis and degradation (Fig. 8). This module included the host gene CYP27A1, which is involved in bile acid production (47), and NR1H3, a host gene that codes for a bile acid receptor (48). These genes were upregulated in the cecum and transverse colon compared with the rectum. The microbiome genes interacting with these host genes included K03664 (smpB), a gene essential for bacterial survival when upregulated in the presence of secondary bile acid deoxycholic acid, K02835 (prfA), which positively regulates the expression of a gene coding for bile acid hydrolase, and K03527 (ispH, lytB), a gene involved in bile acid sensitivity. Additionally, Bacteroides vulgatus contributed to the expression of all these microbiome genes, which aligns with previous studies showing an increase in Bacteroides spp. correlating with the expression of the host gene NR1H3 (49).

DISCUSSION

In this study, we extracted gene modules from a host and microbiome gene co-expression network based on dual transcriptome profiling. The module extraction method proposed in this study is based on clique finding and takes into account the structure of three types of networks: host-host, host-microbiome, and microbiome-microbiome. This approach enables the extraction of modules containing both host genes and microbiome genes. Conventional module extraction methods do not differentiate between host gene nodes and microbiome gene nodes within the network, potentially resulting in modules composed exclusively of either host genes or microbiome genes. Indeed, when conventional module extraction methods such as the Newman algorithm (15), the Louvain algorithm (16), the Leiden algorithm (17), and weighted correlation network analysis (WGCNA) (18) were applied to our data set (Supplemental Note Section 9), we observed that these modules included those composed solely of host genes or microbiome genes (Table S5). Notably, about half of the genes in WGCNA modules were found in modules comprising only one species. While the purpose of module extraction is to interpret the interactions among genes within a module, these single-species modules do not allow for an interpretation of the relationship between host genes and microbiome genes. Furthermore, to verify whether functionally similar genes were being extracted, we conducted enrichment analysis on the extracted modules (Supplemental Note Section 8). The results showed that our method had the highest proportion of functionally characterized modules (enriched modules) (Table S6).
The gene modules extracted, which encompassed both host and microbiome genes, frequently exhibited relationships where a small number of genes regulated a larger number of genes between the host and microbiome genes. These observations are thought to reflect the characteristics of the interactions between the host and the microbiome. Several studies have reported the impact of host gene mutations on the intestinal microbiome. For instance, it has been reported that the host gene VDR, which codes for the vitamin D receptor, influences the colonization of Parabacteroides (50) and that the host gene ABO is associated with Faecalicatena lactaris (51). In another study investigating the relationship between the intestinal microbiome and host gene loci through quantitative trait locus analysis, it was suggested that Akkermansia muciniphila regulates the expression of ATF3, which codes for a transcription factor that plays a crucial role in metabolic and immune regulation, and this bacterium is related to only four genes (52). These studies highlight the fact that a single or a small number of host genes are controlling or are controlled by the intestinal microbiome. Conversely, a small number of microbiome genes can affect the host’s metabolic (53), fitness (54), and longevity (55) phenotypes. For example, structural mutations in the gene related to butyrate transport in Anaerostipes hadrus are strongly associated with a reduction in the host’s metabolic risk (53), and the intestinal microbiome’s thiamine biosynthesis genes, transferred by horizontal transmission, influence the host’s fitness (54). Considering these previous studies and our results, it is suggested that the interaction between the host and the microbiome involves widespread influence of one’s genes on the activity of the other.
The common marmoset used in this study bears anatomical and pharmacological similarities to humans (19), and the fecal microbiome of the common marmoset is more similar to that of humans compared with other animal models (13). While the common marmoset’s cecum—one of the study’s targeted intestinal sites—does not possess an appendage as pronounced as the human appendix, it is known to share significant similarities with the human appendix in terms of microanatomical structure and immune reactivity (56, 57). The results of this study using common marmosets could therefore hold higher applicability to humans compared with other animal models.
In conclusion, we generated host transcriptome and microbiome metatranscriptome profiles of the common marmoset and performed gene co-expression network analysis. The algorithm we proposed for extracting gene modules in the gene co-expression network enabled the identification of dense, functionally related host-microbiome co-expression modules, which was challenging using the conventional module extraction algorithms. The identified gene modules provided new insights into relationships between host genes and intestinal microbiota and microbial genes bridging interactions between the host and bacteria. Our findings underscore that interactions between the host and microbiome are not uniform throughout the colon but are localized, indicating spatial variation in host-microbiome interactions along the colon. These insights could be crucial in understanding mechanisms behind localized gut diseases, such as colorectal cancer and inflammatory bowel disease, and in developing localized therapies (5861).

MATERIALS AND METHODS

Animal experiments

Common marmosets were housed at the Central Institute for Experimental Animals (Kawasaki, Japan). Five marmosets were euthanized with intravenous administration of pentobarbital overdose, and the digestive tract was isolated (Table S7, Supplemental Note Section 1). Three gastrointestinal site samples (cecum, transverse colon, and rectum), including the mucosal layer, were used for dual RNA-seq. These three sites were targeted at the beginning, middle, and end of the colon, which has an abundant microbiome.

RNA extraction and dual RNA sequencing of host and microbiome

RNA libraries were prepared after removing rRNA from the extracted high-quality Total RNA (RIN scores 8.6; Table S1). Illumina NovaSeq 6000 v1.5 sequencing yielded a total of 472 Gnt of paired-end reads (100 nt × 2). This data set included an average of 314.9 M reads per sample (Tables S2 and S8; Supplemental Note Section 2).

Construction of host-microbiome co-expression network and identification of gene modules

In this study, we utilized gene co-expression network analysis to identify interacting gene modules of host and microbiome (Supplemental Note Section 7). We established three types of networks: host-host, host-microbiome, and microbiome-microbiome, wherein genes were considered as nodes and co-expressing genes were interconnected (Fig. 1D). Networks were defined by an adjacency matrix, where the adjacency aij between gene i and gene j was defined as follows based on co-expression similarity sij:
aij={1, if sij>α0, otherwise
The co-expression similarity sij was computed using the Spearman’s correlation coefficient calculated for genes i and j represented by vectors of TPM values for host-host and microbiome-microbiome networks and absolute values of Spearman’s correlation coefficients for host-microbiome networks. The adjacency threshold α was set at 0.80. These three types of networks were then integrated into one.
We developed an algorithm to identify densely interacting gene modules from the host-microbiome co-expression network. This algorithm, based on clique finding, progressively merges cliques (Fig. 1E). In graph theory, a “clique” refers to a subgraph within the network where all nodes are fully connected. In the context of a gene co-expression network, all genes within a clique indicate a strong co-expression (correlation) among them. In this study, we focus only on networks and cliques that include gene nodes from both the host and microbiome, encompassing three types of networks: host-host, host-microbiome, and microbiome-microbiome. Our algorithm identified modules of interacting genes in the gene co-expression network as aggregates of overlapping cliques. It was implemented in three steps. (i) Cliques within the network were found. (ii) Beginning with the largest cliques, we examined their overlap with any other cliques. If overlap occurred, we computed the cliqueness scores independently for each of the three types of networks (host-host, host-microbiome, and microbiome-microbiome networks) upon merging the overlapping cliques. The cliqueness scores for the host-host, host-microbiome, and microbiome-microbiome networks were denoted as Chh, Chm, and Cmm, respectively. (iii) Cliques were merged if they satisfied the condition: Chh>τ, Cmm>τ, and Cmm>τ. The threshold τ for cliqueness was set at 0.60 based on parameter determination (Supplemental Note Section 7). If there were multiple cliques with minimum cliqueness scores exceeding the threshold, the clique with the highest cliqueness score was selected. Steps ii and iii were repeated until no cliques could be merged.
For the host-host networks, the cliqueness score Chh between cliques A and B was defined as follows:
Chh=E12 N (N1)
Here, N is defined as the number of host gene nodes in the union of cliques A and B, represented as N=|NANB|. NA and NB denote the number of host gene nodes within clique A and clique B, respectively. E corresponds to the total number of edges between the host nodes included in cliques A and/or B, formulated as E=EA+EB+EAB. Here, EA and EB represent the number of edges between host gene nodes within clique A and clique B, respectively, while EAB denotes the number of edges connecting across host gene nodes within clique A and clique B. The cliqueness score for microbiome-microbiome networks Cmm was calculated in the same way.
For the host-microbiome network, the cliqueness score Chm was defined as follows:
Chm=ENhost Nmicrobiome
where Nhost and Nmicrobiome represent the number of host and microbiome gene nodes, respectively, in the union of cliques A and B. E corresponds to the total number of edges between the host or microbiome gene nodes included in cliques A and/or B.

ACKNOWLEDGMENTS

We thank the scientists of the Central Institute for Experimental Animals for providing support for animal experiments and the members of the Sakakibara Lab at Keio University for helpful discussions.
This work was supported by grants from the Japan Agency for Medical Research and Development (AMED PRIME) Grant Number JP19gm6010006 and JST, CREST Grant Number JPMJCR20S3, Japan. M.U. has received funding from JSPS KAKENHI Grant Number JP20J21477.
M.U. performed the experiments, conducted the bioinformatics analysis, and co-wrote the paper; T.I. and E.S. provided common marmoset samples; M.U. and S.H. performed RNA extraction; A.T. performed deep sequencing with high-throughput sequencer; Y.S. designed and supervised the research, analyzed the data, and co-wrote the paper. All authors have read and approved the manuscript.
The authors declare no competing interests.

SUPPLEMENTAL MATERIAL

Supplemental material - msystems.01405-23-s0001.docx
Supplemental text, tables, and figures.
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

1.
Yoon HS, Cho CH, Yun MS, Jang SJ, You HJ, Kim J-H, Han D, Cha KH, Moon SH, Lee K, Kim Y-J, Lee S-J, Nam T-W, Ko G. 2021. Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat Microbiol 6:563–573.
2.
Vatanen T, Kostic AD, d’Hennezel E, Siljander H, Franzosa EA, Yassour M, Kolde R, Vlamakis H, Arthur TD, Hämäläinen A-M, et al. 2016. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell 165:842–853.
3.
Song X, Sun X, Oh SF, Wu M, Zhang Y, Zheng W, Geva-Zatorsky N, Jupp R, Mathis D, Benoist C, Kasper DL. 2020. Microbial bile acid metabolites modulate gut RORγ+ regulatory T cell homeostasis. Nature 577:410–415.
4.
Zhang L, Forst CV, Gordon A, Gussin G, Geber AB, Fernandez PJ, Ding T, Lashua L, Wang M, Balmaseda A, Bonneau R, Zhang B, Ghedin E. 2020. Characterization of antibiotic resistance and host-microbiome interactions in the human upper respiratory tract during influenza infection. Microbiome 8:39.
5.
Fujiogi M, Raita Y, Pérez-Losada M, Freishtat RJ, Celedón JC, Mansbach JM, Piedra PA, Zhu Z, Camargo CA, Hasegawa K. 2022. Integrated relationship of nasopharyngeal airway host response and microbiome associates with bronchiolitis severity. Nat Commun 13:4970.
6.
Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, Andrews E, Ajami NJ, Bonham KS, Brislawn CJ, et al. 2019. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569:655–662.
7.
Priya S, Burns MB, Ward T, Mars RAT, Adamowicz B, Lock EF, Kashyap PC, Knights D, Blekhman R. 2022. Identification of shared and disease-specific host gene–microbiome associations across human diseases using multi-omic integration. Nat Microbiol 7:780–795.
8.
Cremer J, Arnoldini M, Hwa T. 2017. Effect of water flow and chemical environment on microbiota growth and composition in the human colon. Proc Natl Acad Sci USA 114:6438–6443.
9.
Dupont HL, Jiang ZD, Dupont AW, Utay NS. 2020. The intestinal microbiome in human health and disease. Trans Am Clin Climatol Assoc 131:178–197.
10.
Hu S, Wang Y, Lichtenstein L, Tao Y, Musch MW, Jabri B, Antonopoulos D, Claud EC, Chang EB. 2010. Regional differences in colonic mucosa-associated microbiota determine the physiological expression of host heat shock proteins. Am J Physiol Gastrointest Liver Physiol 299:G1266–75.
11.
Yasuda K, Oh K, Ren B, Tickle TL, Franzosa EA, Wachtman LM, Miller AD, Westmoreland SV, Mansfield KG, Vallender EJ, Miller GM, Rowlett JK, Gevers D, Huttenhower C, Morgan XC. 2015. Biogeography of the intestinal mucosal and lumenal microbiome in the rhesus macaque. Cell Host Microbe 17:385–391.
12.
Kozik AJ, Nakatsu CH, Chun H, Jones-Hall YL. 2019. Comparison of the fecal, cecal, and mucus microbiome in male and female mice after TNBS-induced colitis. PLoS One 14:e0225079.
13.
Uehara M, Inoue T, Kominato M, Hase S, Sasaki E, Toyoda A, Sakakibara Y. 2022. Intraintestinal analysis of the functional activity of microbiomes and its application to the common marmoset intestine. mSystems 7:e00520-22.
14.
Newman MEJ. 2004. Detecting community structure in networks. Eur Phys J B 38:321–330.
15.
Blondel VD, Guillaume JL, Lambiotte R, Lefebvre E. 2008. Fast unfolding of communities in large networks. J Stat Mech 2008:10008.
16.
Traag VA, Waltman L, van Eck NJ. 2019. From Louvain to Leiden: guaranteeing well-connected communities. Sci Rep 9:5233.
17.
Langfelder P, Horvath S. 2008. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9:1–3.
18.
Hugenholtz F, de Vos WM. 2018. Mouse models for human intestinal microbiota research: a critical evaluation. Cell Mol Life Sci 75:149–160.
19.
Kishi N, Sato K, Sasaki E, Okano H. 2014. Common marmoset as a new model animal for neuroscience research and genome editing technology. Dev Growth Differ 56:53–62.
20.
Inoue T, Okahara N, Okahashi N, Sato K, Ueda M, Atarashi K, Isayama J, Yoshimasa A, Kawashima Y, Lee C-Y, Ueno M, Nozu R, Kurotaki Y, Mineshige T, Yurimoto T, Itaya K, Tanoue T, Nakahata T, Shiota A, Arita M, Honda K, Sasaki E. 2021. Generation of germ-free common marmosets. Research Square.
21.
Stuart JM, Segal E, Koller D, Kim SK. 2003. A gene-coexpression network for global discovery of conserved genetic modules. Science 302:249–255.
22.
Wang K, Wei G, Liu D. 2012. CD19: a biomarker for B cell development, lymphoma diagnosis and therapy. Exp Hematol Oncol 1:1–7.
23.
Huse K, Bai B, Hilden VI, Bollum LK, Våtsveen TK, Munthe LA, Smeland EB, Irish JM, Wälchli S, Myklebust JH. 2022. Mechanism of CD79A and CD79B support for IgM+ B cell fitness through B cell receptor surface expression. J Immunol 209:2042–2053.
24.
Clark EA, Giltiay NV. 2018. CD22: a regulator of innate and adaptive B cell responses and autoimmunity. Front Immunol 9:2235.
25.
Pan C, Baumgarth N, Parnes JR. 1999. CD72-deficient mice reveal nonredundant roles of CD72 in B cell development and activation. Immunity 11:495–506.
26.
Alegre ML, Frauwirth KA, Thompson CB. 2001. T-cell regulation by CD28 and CTLA-4. Nat Rev Immunol 1:220–228.
27.
Yang Y, Zang Y, Zheng C, Li Z, Gu X, Zhou M, Wang Z, Xiang J, Chen Z, Zhou Y. 2020. CD3D is associated with immune checkpoints and predicts favorable clinical outcome in colon cancer. Immunotherapy 12:25–35.
28.
Hubbard N, Hagin D, Sommer K, Song Y, Khan I, Clough C, Ochs HD, Rawlings DJ, Scharenberg AM, Torgerson TR. 2016. Targeted gene editing restores regulated CD40L function in X-linked hyper-IgM syndrome. Blood 127:2513–2522.
29.
Sobhani N, Tardiel-Cyril DR, Davtyan A, Generali D, Roudi R, Li Y. 2021. CTLA-4 in regulatory T cells for cancer immunotherapy. Cancers 13:1–18.
30.
He Y, Hua R, Li B, Gu H, Sun Y, Li Z. 2020. Loss of FBP1 promotes proliferation, migration, and invasion by regulating fatty acid metabolism in esophageal squamous cell carcinoma. Aging 13:4986–4998.
31.
Hirata H, Sugimachi K, Komatsu H, Ueda M, Masuda T, Uchi R, Sakimura S, Nambara S, Saito T, Shinden Y, Iguchi T, Eguchi H, Ito S, Terashima K, Sakamoto K, Hirakawa M, Honda H, Mimori K. 2016. Decreased expression of fructose-1,6-bisphosphatase associates with glucose metabolism and tumor progression in hepatocellular carcinoma. Cancer Res 76:3265–3276.
32.
Dong Y, Huaying S, Danying W, Chihong Z, Ruibin J, Xiaojiang S, Jianguo F. 2018. Significance of methylation of FBP1 gene in non-small cell lung cancer. Biomed Res Int 2018:3726091.
33.
Li H, Qi Z, Niu Y, Yang Y, Li M, Pang Y, Liu M, Cheng X, Xu M, Wang Z. 2021. FBP1 regulates proliferation, metastasis, and chemoresistance by participating in C-MYC/STAT3 signaling axis in ovarian cancer. Oncogene 40:5938–5949.
34.
Hou Y, Wei W, Guan X, Liu Y, Bian G, He D, Fan Q, Cai X, Zhang Y, Wang G, Zheng X, Hao H. 2021. A diet-microbial metabolism feedforward loop modulates intestinal stem cell renewal in the stressed gut. Nat Commun 12:271.
35.
Mardinoglu A, Shoaie S, Bergentall M, Ghaffari P, Zhang C, Larsson E, Bäckhed F, Nielsen J. 2015. The gut microbiota modulates host amino acid and glutathione metabolism in mice. Mol Syst Biol 11:834.
36.
Medina DA, Pinto F, Ovalle A, Thomson P, Garrido D. 2017. Prebiotics mediate microbial interactions in a consortium of the infant gut microbiome. Int J Mol Sci 18:2095.
37.
Cheng J, Hu J, Geng F, Nie S. 2022. Bacteroides utilization for dietary polysaccharides and their beneficial effects on gut health. Food Sci Hum Wellness11:1101–1110.
38.
Rosser EC, Piper CJM, Matei DE, Blair PA, Rendeiro AF, Orford M, Alber DG, Krausgruber T, Catalan D, Klein N, Manson JJ, Drozdov I, Bock C, Wedderburn LR, Eaton S, Mauri C. 2020. Microbiota-derived metabolites suppress arthritis by amplifying aryl-hydrocarbon receptor activation in regulatory B cells. Cell Metab 31:837–851.
39.
Ding X, Bin P, Wu W, Chang Y, Zhu G. 2020. Tryptophan metabolism, regulatory T cells, and inflammatory bowel disease: a mini review. Mediators Inflamm 2020:9706140.
40.
Ezeji JC, Sarikonda DK, Hopperton A, Erkkila HL, Cohen DE, Martinez SP, Cominelli F, Kuwahara T, Dichosa AEK, Good CE, Jacobs MR, Khoretonenko M, Veloo A, Rodriguez-Palacios A. 2021. Parabacteroides distasonis: intriguing aerotolerant gut anaerobe with emerging antimicrobial resistance and pathogenic and probiotic roles in human health. Gut Microbes 13:1922241.
41.
Cekanaviciute E, Yoo BB, Runia TF, Debelius JW, Singh S, Nelson CA, Kanner R, Bencosme Y, Lee YK, Hauser SL, Crabtree-Hartman E, Sand IK, Gacias M, Zhu Y, Casaccia P, Cree BAC, Knight R, Mazmanian SK, Baranzini SE. 2017. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proc Natl Acad Sci USA 114:10713–10718.
42.
Łaniewski P, Herbst-Kralovetz MM. 2021. Bacterial vaginosis and health-associated bacteria modulate the immunometabolic landscape in 3D model of human cervix. NPJ Biofilms Microbiomes 7:88.
43.
Tomkovich S, Gharaibeh RZ, Dejea CM, Pope JL, Jiang J, Winglee K, Gauthier J, Newsome RC, Yang Y, Fodor AA, Schmittgen TD, Sears CL, Jobin C. 2020. Human colon mucosal biofilms and murine host communicate via altered mRNA and microRNA expression during cancer. mSystems 5:e00451-19.
44.
Cornick S, Tawiah A, Chadee K. 2015. Roles and regulation of the mucus barrier in the gut. Tissue Barriers 3:e982426.
45.
González-Morelo KJ, Vega-Sagardía M, Garrido D. 2020. Molecular insights into O-linked glycan utilization by gut microbes. Front Microbiol 11:591568.
46.
Png CW, Lindén SK, Gilshenan KS, Zoetendal EG, McSweeney CS, Sly LI, McGuckin MA, Florin THJ. 2010. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol 105:2420–2428.
47.
Zurkinden L, Sviridov D, Vogt B, Escher G. 2020. Downregulation of Cyp7a1 by cholic acid and chenodeoxycholic acid in Cyp27a1/ApoE double knockout mice: differential cardiovascular outcome. Front Endocrinol11:586980.
48.
Biagioli M, Marchianò S, Carino A, Di Giorgio C, Santucci L, Distrutti E, Fiorucci S. 2021. Bile acids activated receptors in inflammatory bowel disease. Cells 10:1281.
49.
Badi SA, Motahhary A, Bahramali G, Masoumi M, Khalili SFS, Ebrahimzadeh N, Nouri P, Rahimi A, Masotti A, Moshiri A, Siadat SD. 2020. The regulation of Niemann-Pick C1-Like 1 (NPC1L1) gene expression in opposite direction by Bacteroides spp. and related outer membrane vesicles in Caco-2 cell line. J Diabetes Metab Disord 19:415–422.
50.
Wang J, Thingholm LB, Skiecevičienė J, Rausch P, Kummen M, Hov JR, Degenhardt F, Heinsen F-A, Rühlemann MC, Szymczak S, et al. 2016. Genome-wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota. Nat Genet 48:1396–1406.
51.
Qin Y, Havulinna AS, Liu Y, Jousilahti P, Ritchie SC, Tokolyi A, Sanders JG, Valsta L, Brożyńska M, Zhu Q, Tripathi A, Vázquez-Baeza Y, Loomba R, Cheng S, Jain M, Niiranen T, Lahti L, Knight R, Salomaa V, Inouye M, Méric G. 2022. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat Genet 54:134–142.
52.
Zhang Q, Linke V, Overmyer KA, Traeger LL, Kasahara K, Miller IJ, Manson DE, Polaske TJ, Kerby RL, Kemis JH, et al. 2023. Genetic mapping of microbial and host traits reveals production of immunomodulatory lipids by Akkermansia muciniphila in the murine gut. Nat Microbiol 8:424–440.
53.
Zeevi D, Korem T, Godneva A, Bar N, Kurilshikov A, Lotan-Pompan M, Weinberger A, Fu J, Wijmenga C, Zhernakova A, Segal E. 2019. Structural variation in the gut microbiome associates with host health. Nature 568:43–48.
54.
Wang Y, Baumdicker F, Schweiger P, Kuenzel S, Staubach F. 2021. Horizontal gene transfer-mediated bacterial strain variation affects host fitness in Drosophila. BMC Biol 19:187.
55.
Han B, Sivaramakrishnan P, Lin C-CJ, Neve IAA, He J, Tay LWR, Sowa JN, Sizovs A, Du G, Wang J, Herman C, Wang MC. 2017. Microbial genetic composition tunes host longevity. Cell 169:1249–1262.
56.
Zinnen AD, Vichich J, Metzger JM, Gambardella JC, Bondarenko V, Simmons HA, Emborg ME. 2022. Alpha-synuclein and tau are abundantly expressed in the ENS of the human appendix and monkey cecum. PLoS One 17:e0269190.
57.
Fisher RE. 2000. The primate appendix: a reassessment. Anat Rec 261:228–236.
58.
Debelius JW, Engstrand L, Matussek A, Brusselaers N, Morton JT, Stenmarker M, Olsen RS. 2023. The local tumor microbiome is associated with survival in late-stage colorectal cancer patients. Microbiol Spectr 11:e05066-22.
59.
Drewes JL, White JR, Dejea CM, Fathi P, Iyadorai T, Vadivelu J, Roslani AC, Wick EC, Mongodin EF, Loke MF, Thulasi K, Gan HM, Goh KL, Chong HY, Kumar S, Wanyiri JW, Sears CL. 2017. High-resolution bacterial 16S rRNA gene profile meta-analysis and biofilm status reveal common colorectal cancer consortia. NPJ Biofilms Microbiomes 3:34.
60.
Čipčić Paljetak H, Barešić A, Panek M, Perić M, Matijašić M, Lojkić I, Barišić A, Vranešić Bender D, Ljubas Kelečić D, Brinar M, Kalauz M, Miličević M, Grgić D, Turk N, Karas I, Čuković-Čavka S, Krznarić Ž, Verbanac D. 2022. Gut microbiota in mucosa and feces of newly diagnosed, treatment-naïve adult inflammatory bowel disease and irritable bowel syndrome patients. Gut Microbes 14:2083419.
61.
Yu L-H. 2018. Microbiota dysbiosis and barrier dysfunction in inflammatory bowel disease and colorectal cancers: exploring a common ground hypothesis. J Biomed Sci 25:79.

Information & Contributors

Information

Published In

cover image mSystems
mSystems
Volume 9Number 516 May 2024
eLocator: e01405-23
Editor: Mani Arumugam, Kobenhavns Universitet, Copenhagen, Denmark
PubMed: 38557130

History

Received: 4 January 2024
Accepted: 12 March 2024
Published online: 1 April 2024

Keywords

  1. gut microbiome
  2. metatranscriptome
  3. transcriptome
  4. network analysis
  5. host-microbiome interaction
  6. non-human primate
  7. common marmoset

Data Availability

All raw sequence data have been submitted to the DDBJ under project PRJDB16352 from the Ministry of Education, Culture, Sports, Science and Technology of Japan. The gene expression profiles of the host and microbiome are available at https://doi.org/10.5281/zenodo.8264866. The algorithm for module extraction is available at https://github.com/MikaUhr/MEIGN.git.

Contributors

Authors

Department of Biosciences and Informatics, Keio University, Yokohama, Kanagawa, Japan
Author Contributions: Formal analysis, Funding acquisition, Investigation, Methodology, Resources, Validation, Visualization, Writing – original draft, and Writing – review and editing.
Takashi Inoue
Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
Author Contributions: Resources and Writing – review and editing.
Sumitaka Hase
Department of Biosciences and Informatics, Keio University, Yokohama, Kanagawa, Japan
Author Contributions: Investigation and Writing – review and editing.
Erika Sasaki
Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
Author Contributions: Resources and Writing – review and editing.
Atsushi Toyoda
Department of Genomics and Evolutionary Biology, National Institute of Genetics, Mishima, Shizuoka, Japan
Author Contributions: Resources and Writing – review and editing.
Department of Biosciences and Informatics, Keio University, Yokohama, Kanagawa, Japan
Author Contributions: Conceptualization, Funding acquisition, Project administration, Supervision, Writing – original draft, and Writing – review and editing.

Editor

Mani Arumugam
Editor
Kobenhavns Universitet, Copenhagen, Denmark

Notes

The authors declare no conflict of interest.

Ethics Approval

The animal experiment protocol was approved by the CIEA Institutional Animal Care and Use Committee (approval no. 17031, 18032, and 21084). The study was conducted in accordance with the guidelines of CIEA that comply with the Guidelines for Proper Conduct of Animal Experiments published by the Science Council of Japan. Animal care was conducted in accordance with the Guide for the Care and Use of Laboratory Animals (Institute for Laboratory Animal Resources, 2011).

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures

Tables

Media

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy