Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Apr 10.
Published in final edited form as: Cell Host Microbe. 2024 Mar 12;32(4):506–526.e9. doi: 10.1016/j.chom.2024.02.012

Longitudinal profiling of the microbiome at four body sites reveals core stability and individualized dynamics during health and disease

Xin Zhou 1,2,3,4,17, Xiaotao Shen 1,2,17, Jethro S Johnson 4,5, Daniel J Spakowicz 4,6, Melissa Agnello 7, Wenyu Zhou 1,2, Monica Avina 1, Alexander Honkala 1,8,9, Faye Chleilat 1, Shirley Jingyi Chen 8, Kexin Cha 8, Shana Leopold 4,10, Chenchen Zhu 1, Lei Chen 4,11, Lin Lyu 11, Daniel Hornburg 1, Si Wu 1, Xinyue Zhang 1, Chao Jiang 1,12, Liuyiqi Jiang 12, Lihua Jiang 1, Ruiqi Jian 1, Andrew W Brooks 1, Meng Wang 1, Kévin Contrepois 1, Peng Gao 1, Sophia Miryam Schüssler-Fiorenza Rose 1, Thi Dong Binh Tran 4, Hoan Nguyen 4, Alessandra Celli 1, Bo-Young Hong 4,13, Eddy J Bautista 4,14, Yair Dorsett 4,15, Paula B Kavathas 10,16, Yanjiao Zhou 4,15, Erica Sodergren 4, George M Weinstock 4, Michael P Snyder 1,2,3,8,18
PMCID: PMC11022754  NIHMSID: NIHMS1978110  PMID: 38479397

SUMMARY

To understand dynamic interplay between the human microbiome and host during health and disease, we analyzed the microbial composition, temporal dynamics, and associations with host multi-omics, immune and clinical markers of microbiomes from four body sites in 86 participants over six years. We found that microbiome stability and individuality are body-site-specific and heavily influenced by the host. The stool and oral microbiome are more stable than the skin and nasal microbiomes, possibly due to their interaction with the host and environment. We identify individual-specific and commonly shared bacterial taxa, with individualized taxa showing greater stability. Interestingly, microbiome dynamics correlate across body sites, suggesting systemic dynamics influenced by host-microbial-environment interactions. Notably, insulin-resistant individuals show altered microbial stability and associations between microbiome, molecular markers, and clinical features, suggesting their disrupted interaction in metabolic disease. Our study offers comprehensive views of multi-site microbial dynamics and their relationship with host health and disease.

Keywords: microbiome stability, stool microbiome, skin microbiome, oral microbiome, nasal microbiome, precision medicine, longitudinal profiling

eTOC Blurb:

Zhou et al. explore the complex interactions between human microbiomes and their hosts across different body sites, revealing that microbiome stability and its impact on health are influenced by site-specific host factors. Their findings underscore the systemic nature of host-microbiome relationships, with significant implications for understanding metabolic diseases.

Graphical Abstract:

graphic file with name nihms-1978110-f0001.jpg

Some of the icons in the summary figure were created with Biorender.com.

INTRODUCTION

The human microbiome comprises highly dynamic microbial communities inhabiting various body sites15, engaging in intricate host-microbial interactions that display territory-specific complexity610. Advancements in multi-omics technologies have catalyzed the elucidation of the molecular mechanisms underlying microbial ecology and their interactions with host, unveiling the critical roles of the microbiome in normal physiological processes such as aging1113 as well as diseases including inflammatory bowel disease (IBD)1416, cardiovascular disease1719, and type 2 diabetes mellitus (T2DM)2023.

The etiology and pathogenesis of insulin resistance and T2DM have been closely linked to the human microbiome2427. Patients with impaired insulin and glucose homeostasis exhibit shifted microbiome composition in the gut21,23,2529, skin3033, and other body sites3440, reflecting an ecological dysbiosis characterized by altered microbial alpha diversity25,41, decreased compositional stability42,43, and greater inter-individual variability25. Compromised mucosal and skin barrier integrity, often associated with insulin resistance, may potentiate microbial translocation, thereby exacerbating systemic inflammation4447. Although human microbiome studies are often, by necessity, observational, a causal relationship between microbiome dysbiosis and impaired glucose/insulin homeostasis has been demonstrated through human microbiome manipulation41,48,49.

While prior studies on the microbiome and glucose homeostasis have been informative, they exhibit certain limitations. Firstly, these studies21,50,51 often lack dense longitudinal sampling, essential for capturing stability features, thus limiting fundamental insights into host-microbe interactions5254. Secondly, they primarily focus on the microbiome from a single host site4,50,5558, overlooking the importance of simultaneous multi-region sampling for assessing microbiome site-specific dynamics, and their interplay across various host microenvironments9,5962. Lastly, most studies8,18,21,6366 do not concurrently measure host clinical and molecular phenotypes, impeding the exploration of the molecular relationships underpinning health and disease-related host-microbiome interactions67,68.

Collaborative initiatives like the Integrative Human Microbiome Project (iHMP) and Integrative Personal Omics Profiling (iPOP) offer avenues to surmount previous studies’ limitations by investigating well-characterized human longitudinal cohorts6971. In this study, we examined the relationships between multi-site microbiomes and host health in the context of prediabetes, through characterizing the microbiome collected from four body sites in 86 adults for over six years and examining their associations with host omics and clinical characteristics. We describe unique longitudinal trajectories for microbiomes across different body sites, demonstrating their responsiveness to both host-specific and environmental factors. We found that personalized microbiomes exhibit greater stability than communal microbes, highlighting the resilience of the individualized microbiome. Nonetheless, these systems remain vulnerable to disturbances like viral infections, which may precipitate dysbiosis linked to metabolic dysfunctions. Furthermore, our analysis revealed associations between site-specific microbiomes and clinical parameters such as cytokine profiles and insulin sensitivity, highlighting the intertwined nature of immune activity and metabolic well-being.

RESULTS

Description of the study design

We analyzed stool, skin, oral, and nasal microbiome from a human cohort of 86 participants for up to six years (1,126.6 ± 455.8 days). (Fig. 1A, see STAR Methods). The cohort comprised 41 males and 45 females, aged between 29 and 75 years old (55 ± 9.8 years old), with BMIs ranging from 19.1 to 40.8 kg/m^2 (28.31 ± 4.44 kg/m^2) (Table S1A). Sampling occurred quarterly, with an additional 3–7 samples collected within five weeks (12% of the total) during periods of stress, such as respiratory illness, vaccination, or antibiotic use. The 16S ribosomal RNA gene sequencing method employed in this study targeted a variable region to facilitate the detection of amplicon sequence variants (ASVs)72, enabling the identification and differentiation of most bacterial taxa at the genus and species levels73,74.

Figure 1. Longitudinal profiles of the microbiome at four body sites.

Figure 1.

A. Study Design. B. Overlap of sample numbers among different omics types. C. Proportion of stress, insulin resistant and healthy samples. D. UMAP of microbiome samples by body site. E. Density distribution of microbiome richness and evenness. F. Rank prevalence curve of microbiome genera with the 100 highest longitudinal prevalence at each body site.

A unique feature of this cohort is the multi-omics phenotyping of participants at each timepoint (Fig. 1B). Untargeted proteomics (302 proteins), untargeted metabolomics (724 annotated metabolic features), targeted lipidomics (846 annotated lipids), and 62 targeted cytokine and growth factor measurements were performed, along with 51 clinical markers, including C-reactive protein (CRP), fasting glucose (FG), hemoglobin A1C (HbA1C), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) from plasma samples. (Fig. 1A). Glucose control assessments, comprising an annual oral glucose tolerance test for all participants and a gold-standard steady-state plasma glucose (SSPG) measurement75 for 58 individuals, classified 28 individuals as insulin-sensitive (IS) and 30 as insulin-resistant (IR) (Fig 1C). Overall, we analyzed a total of 3,058 visits, 5,432 biological samples (1,467 plasma samples, 926 stool samples, 1,116 skin samples, 1,001 oral samples, and 922 nasal samples) generating a total of 118,124,374 measurements. The microbial and other data can be found on our data portal: https://portal.hmpdacc.org/.

Microbial Demographics and Personalization Across Body Sites: Unraveling the Impact of Diet and Environment

We initially examined the overall demographics of the microbiome at each of the four body sites (Fig. 1D) (Fig. S1A). Extending previous studies7,62,7678, we observed a separation between body sites, including a clear boundary of the skin and nasal samples, highlighting the pronounced territory specificity of each microbiome6,7,79.

Micro-biotypes, like enterotypes in the stool microbiome50,77,78, are present in all body sites, with their community structure predominantly influenced by specific taxa. The stool microbiome primarily exhibited a gradient of abundance distributions between Bacteroidetes and Firmicutes, except for a few samples with high Prevotella. The recently identified core genus Phocaeicola80 had minimal impact on the overall Bacteroidetes/Firmicutes gradient, but samples with high Phocaeicola and Bacteroides were clearly separated. (Fig. S1A) The oral microbiome was primarily composed of Prevotella, Streptococcus, Veillonella, Haemophilus, Neisseria, and Leptotrichia, as previously described8183. The skin and nasal microbiome samples jointly displayed a triangular distribution, primarily driven by three dominant genera: Cutibacterium, Corynebacterium, and Staphylococcus (Fig. 1D, Fig. S1A) 4,8,62,79. The distribution of microbial genera remained consistent with findings from other cohorts 8,59,84, irrespective of participant diversity in insulin sensitivity in our study. (Fig. S1B). Our study, however, extends these observations by longitudinally comparing inter- and intra-individual covariance across all four body sites within the same cohort.

Intraclass (intra-individual) correlation coefficient (ICC) analysis confirmed that microbial personalization is more pronounced at the ASV level than at broader taxonomic resolutions (Fig. S1C), highlighting stronger individualization with finer taxonomy resolution. Notably, despite similar ecological characteristics and dominant bacterial constituents, the nasal microbiome manifested greater personalization than the skin microbiome (Fig. S1D), presumably because nasal microbiome dynamics are more host dependent79.

While environmental factors like season and diet can influence the human microbiome, our results suggest that the impact of seasonality on the gut microbiome, despite mixed results in previous studies50,8587, is relatively modest. Likely due to its direct external exposure, the skin microbiome exhibited the largest seasonal dynamic. This was followed by the oral microbiome, which may be shaped by seasonal dietary changes51,88,89, as it explained more dietary variance than other sites (Fig. S1E). We also observed a significant fluctuation of the skin and oral microbiome evenness: these two communities both become more even during summer with an increased number of different microbes (Fig. S1F) possibly triggered by environmental changes such as temperature and humidity85,90. Based on two participants with accessible environmental and chemical exposure data90, we found stronger exposome-skin microbiome covariance than in other body sites (Fig. S1G) with noticeable environmental impacts on internal sites like oral and stool microbiomes91, supporting our earlier single-individual observation92. By modeling85 the microbial dynamics across seasons, we found more pronounced alterations in skin and nasal microbiomes than in stool and oral microbiomes. (Table S1B) These findings not only extend our knowledge of gut microbiome specificity and individuality56,93 to other body sites but also broaden our understanding of environmental influences on microbiomes, with diet shaping oral microbiomes and environmental exposure impacting skin and nasal microbiomes.

Microbiome from Distinct Body Sites are Ecologically Unique and Altered in Insulin Resistance

Beyond the influence of personal and environmental factors, we observed distinct ecological attributes among the microbiome at the four body sites (Fig. 1E). The stool microbiome exhibited the greatest richness and evenness, underscoring its essential complexity and functional significance. In contrast, the skin microbiome displayed a more skewed population due to its lower evenness compared to the nasal microbiome, despite their similar richness distributions. These ecological features, which often shift with disease progression, aligned with previous findings of IR-related gut dysbiosis25,70, characterized by a significant decrease in stool microbiome’s alpha diversity (t = 2.8462, p-value = 0.0067). Furthermore, shifts in the richness-evenness scatter plots for all four body sites (Fig. S1H) suggested systemic dysbiosis in IR individuals, highlighted by significantly higher skin microbiome richness (t = −2.9102, p-value = 0.0057) (Fig. S1H) and evenness (t = −2.4393, p-value = 0.019) (Fig. S1H). These shifts indicate that IR-associated dysbiosis extends beyond the gut microbiome.

Importantly, extending longitudinal observations from single body sites56,9496, we define a “core microbiome” (see STAR Methods) as microbes consistently present over time, representing potentially indispensable97,98 genera at each body sites. Interestingly, we found that stool and oral microbiomes maintain more than 25 highly prevalent core genera, whereas nasal and skin microbiomes only had three (Fig. 1F). Importantly, some core genera have low relative abundance, (i.e., Coprococcus Mean Prevalence = 80.75%; Mean Relative Abundance = 0.544%), demonstrating the potential significance of low-abundance strains (Fig. S1I). Intriguingly, the richness of core genera in stool and oral microbiome is negatively associated with steady-state plasma glucose (SSPG) (Spearman Rho = −0.52, p-value = 0.00047) and BMI (Spearman Rho = −0.40, p-value = 0.005), respectively (Fig. S1J), indicating that IR and obesity may be associated with a loss of core microbiomes at these sites.

We further explored the microbiome ecology variations between insulin resistant (IR) and insulin sensitive (IS) individuals. IR subjects had a significantly higher number of skin core genera (t = −2.5856, p-value = 0.014) and a lower number of stool core genera (t = 2.9659, p-value = 0.0051) compared to IS individuals (Fig. S1K). Notably, several butyrate-producing bacteria (i.e., Coprococcus, Parasutterella, and Butyricicoccus) were decreased in IR stool core microbiomes, whereas diabetes-related opportunistic skin pathogens (i.e., Finegoldia99101 and Acinetobacter102105) were enriched in the skin core microbiome of IR individuals (Table S1C). In addition, we found a clear divergence of the rank prevalence curves in stool and skin microbiome (Fig. S1L), demonstrating a global microbial prevalence shift in IR individuals at these two sites.

Additionally, several taxa differed significantly between IR and IS individuals in relative abundance. The stool microbiome of IR individuals showed an increase of genus Phocaeicola (LEfSe_effect size: 0.03; BH-adjusted p-value = 0.017) and a reduction of the genus Unclassified Ruminococcaceae (LEfSe_effect size: 0.017; BH-adjusted p-value = 0.0039), whereas the skin microbiome exhibited a decrease in the genus Cutibacterium (LEfSe_effect size: 0.069; BH-adjusted p-value = 0.007) and an increase of the genus Peptoniphilus (LEfSe_effect size: 0.0076; BH-adjusted p-value = 0.0022), which has been previously associated with diabetes-related skin dysbiosis100,106,107 and necrotizing infections108 (Fig. S1M; Table S1D). No such differences were observed in oral and nasal microbiomes. These findings further indicate that skin and stool microbiome stability is altered in IR individuals. Overall, our results reveal that IR participants exhibit a stool microbiome with reduced richness and butyrate-producing bacteria, and a skin microbiome more susceptible to opportunistic pathogens.

Distinct Taxon-Specific Stability and Individuality in Microbiomes Across Body Sites: A Potential Pathway for Personalized Interventions

We next examined microbiome stability at the genus level across body sites, hypothesizing stability is taxon- and site-specific. We defined a metric “Degree of Microbial Individuality” (DMI, see STAR Methods) for each genus, which quantifies similarity within an individual relative to the population; a high DMI means the microbes taxa is highly individual-specific. We also calculated a “Family Score” (FS, see STAR Methods) to evaluate microbial dissimilarity within households. Expanding observations of the stool microbiome54,70,109, skin, oral and nasal microbiome also demonstrated significantly less intra-individual and within-family variation compared to the variation observed between different individuals (Fig. 2A, Table S2A). Intra-individually, the rate of complete ASV turnover within a genus over time was highest in the nasal microbiome (35.5% of cases), followed by skin (24.4%), stool (24.2%), and oral sites (2.7%). This finding underscores significant intra-individual ecological dynamics at the sub-genus level within microbiomes, despite overall community stability.

Figure 2. The individuality of the microbiome differs significantly across genera and body sites.

Figure 2.

A. Bray Curtis dissimilarity comparisons within individuals, families, and between unrelated participants. B. DMI Scores (see STAR Methods). C. Average DMI Radar Plot by Body Site and Phylum, with significant Kruskal-Wallis test results for Actinobacteria, Bacteroidetes, Firmicutes, Proteobacteria, and Other phyla. D. DMI Cladogram representing stool, skin, oral, and nasal microbiomes. Significance indicated by asterisks: *p < 0.05, **p < 0.01, ***p < 0.001.

The DMI, irrespective of relative abundance, was high in the stool microbiome (Fig. 2B), particularly within the Bacteroidetes phylum (Fig. 2C, Table S2B), possibly due to its pronounced adaptive evolution110,111 and substantial colonization resistance112. Furthermore, the stool microbiome had the lowest FS, whereas oral and nasal microbiomes shared greater similarity within households (Fig. S2A), likely due to common living environments56,113 or direct microbiome exchanges114.

Our data revealed substantial DMI variance across body sites, potentially attributable to inherent niche-specific taxonomic complexities. (Fig. 2D, Table S2C). For example, Corynebacterium in nasal and Bacteroides in stool showed the highest DMI, respectively. These results suggest that specific microbial taxa, adapting to their respective niches, may exhibit enhanced individualization. Therefore, the microbial community at each body site is largely shaped by niche-specific interactions. In contrast, environmental genera such as Klebsiella115 and Haemophilus116 displayed uniformly low DMI across all examined body sites, indicating that external environmental factors exerts a relatively weaker influence on the individuality of the native host microbiome. Interestingly, after adjusting individuals’ collective DMI by each genera’s relative abundance, we found a notable increase of individuality in the stool microbiomes of IR individuals (Fig. S2B), likely due to the increased Bacteroidetes among IR individuals.

Overall, the DMI and FS metrics for each specific genus offer an overarching perspective on microbial host specificity. Meanwhile, they provide crucial insights into the taxonomic composition of the community and potential influences of environmental factors on the host’s microbiome. Additionally, the DMI measurements provide important ecological characteristics about micro-biotypes, including ‘enterotype’ in stool microbiome 78,117 or ‘cutotypes’ in skin microbiome63.

Interplay of Microbial Individuality and Stability: Insights from Longitudinal Multi-Site Microbiome Analysis

Prior studies4,9395,118120 demonstrated that microbiome stability is highly personalized. Consequently, we explored the relationship between microbial individuality and stability across body sites. We first examined the genus recolonization rate, measured by ASV consistency when a genus is detectable after being undetected in one or more consecutive samples. The overall recolonization rate (measured by 1 - Pairwise Jaccard Distance121,122) was significantly associated with DMI on all three body sites except for the oral microbiome (Fig. 3A), which intrinsically maintains a high recolonization rate. Such correlations were strongest in the nasal microbiome (Fig. 3A), potentially explaining the high ICC observed above. Surprisingly, no difference in recolonization rate was found between IR and IS individuals. (Fig. S3A). Our results suggest that highly individualized strains are more likely to recolonize, validating a hypothesis raised by fecal transplantation studies123,124 and implying its potential efficacy in treating IR-related dysbiosis.

Figure 3. Temporal stability of microbiomes associated with individuality and stress events.

Figure 3.

A. Correlations of taxa-recurrence with mean DMI for stool, skin, oral, and nasal samples. B. Linear regression between dissimilarity and collection date interval. C. Beta coefficient of individual-based correlation between sample pair’s BC distances and the collection date intervals. D. Correlations of microbiome abundances within and between body sites. E. DMI differences between correlated and non-correlated genera. F. Microbiome shifts during health and stress events over three months. Significance indicated by asterisks: *p < 0.05, **p < 0.01, ***p < 0.001.

The longitudinal data also enabled tracking of microbiome stability over time by quantifying the dissimilarity between sample pairs in relation to collection date-intervals, which was reported to be higher in IBD-related gut dysbiosis15. Our analysis revealed that the stool microbiome changed more slowly over time, with the nasal site exhibiting the fastest rate of change (p-value < 0.001) (Fig. 3B). Additionally, IR individuals showed significantly lower stability in stool and skin microbiomes than IS individuals, as evidenced by linear mixed models (Stool p-value: 1.82 × 10–06, Skin p-value: 2.84 × 10–12), corroborating our findings of greater microbial abundance disparities in these two body sites between IR and IS participants. (Fig. S1M).

Intra- and Inter-Individual Correlations of Microbiome Dynamics Across Body Sites: Implications for Microbial Interdependence and Territory Specificity

We next investigated whether microbiome dynamics were intra-individually co-associated across body sites, both at the community level and for individual taxa. Hierarchical clustering demonstrated a strong link in personal microbiome dynamics between the skin and nasal sites, whereas the dynamics of the stool microbiome was less correlated with other body sites. (Fig. 3C). The inter-site correlation of microbiome stability indicates systemic microbial coordination, potentially regulated by the common mucosal system125,126. Comparison between IS and IR individuals revealed that both groups had a strong skin-nasal link, but a skin-oral correlation was exclusive to IS individuals. (Fig. S3BS3C). This implies that IR status undermines the stability of the skin or oral microbiome, possibly due to the compromised host regulation of microbiome stability or due to impaired mucosal immunity related to IR127, a key factor for between-body-sites microbial interactions125,126,128,129.

Since the microbiome often operates as inter-dependent guilds130132, we questioned which body sites demonstrate the highest microbial inter-dependency. We found that 18.23% of all stool genus pairings (5,671) were significantly correlated within individuals, mostly Firmicutes (Fig. 3D). Surprisingly, co-association was highest in the oral microbiome (50.17%), followed by skin (47.51%) and nasal (38.36%), highlighting strong synergistic interactions among microbiome members at each body site. (Table S3A)

We further investigated the microbial crosstalk between body sites by searching for sample-wise microbial collinearity. Members from the skin and nasal sites were the most correlated (15.57% of all possible pairs) (Fig. 3D, Table S3A). However, remarkable territory specificity was still evident among core microbiomes of each body site (Table S3B). For instance, three predominant core genera - Corynebacterium, Staphylococcus, and Cutibacterium - exhibited no longitudinal correlation between skin and nasal sites. Similarly, oral Prevotella’s abundance did not correlate with its abundance in the stool microbiome, consistent with our observation of high FS in oral but not stool Prevotella (Table S2C). Although dysbiotic microbiome translocation between body sites has been reported133135, our results suggest these translocation cases likely do not involve the niche-specific core taxa. Moreover, genera that are more interdependent exhibit lower DMI compared to less interdependent genera (Fig 3E, Fig. S3D), indicating that highly individualized microbiomes exhibit greater temporal stability93,136. Expanding on our findings of micro-biotypes across body sites (Fig. S1A), we observed that dominant taxa driving these biotypes exhibit significant inter-individual correlations. Examples include significant correlations of Cutibacterium levels in one’s skin with their Cutibacterium level in nasal (beta = 0.56, BH-adjusted p-value = 0.0012), their Bacteroides in stool (beta = 0.52, BH-adjusted p-value = 0.0038), and their Leptotrichia in oral microbiomes (beta = 0.43, BH-adjusted p-value = 0.0088). Conversely, individuals with high Unclassified Ruminococcaceae in stool correlates with low Veillonella in oral microbiome (beta = −0.35, BH-adjusted p-value = 0.015) (Table S3C). These findings suggest that the establishment of micro-biotypes are regulated by personalized factors.

Acute Events Impact Microbiome Dynamics Across Body Sites and are Influenced by Insulin Resistance

Three strong perturbation events - infection, vaccination, and antibiotic usage - are known to cause stool microbiome disruptions70,76,125,137. Within a three-month period, the stool microbiome of IR individuals exhibited greater temporal shifts between healthy visits and perturbations than IS individuals (Fig. 3F). This pattern, however, was not consistently discernible across other body sites, suggesting a site-specific microbiome response to these events. Unlike IS individuals, IR individuals displayed reduced stool microbiome evenness and lacked the perturbation in nasal microbiome during respiratory infection (Fig. S3E), possibly due to higher IR-associated mucosal inflammation138140 masking or superseding infection-induced local microbiome changes observed in IS individuals. During the course of infection, we identified a transient increase in several genera such as Alistipes in the stool, Peptoniphilus on the skin, Unclassified Prevotellaceae in the oral cavity, and Oribacterium in the nasal cavity. Conversely, we noticed a decrease in the presence of Clostridium cluster IV in the stool, Unclassified Clostridia in the oral cavity, and Unclassified Neisseriales in the nasal cavity (Fig. S3FS3H, Table S3D). Although the extent to which transient microbial shifts contribute to IR-related dysbiosis is not clear, the notable increase of core microbiomes in IR skin samples (Fig. S1K) may suggest a correlation between acute perturbations and long-term changes in the core microbiome composition. Overall, our findings indicate that dysbiosis can manifest differently across body sites, potentially through site-specific mechanisms. For instance, IR-related temporary disruptions in the stool microbiome seem to be characterized by a loss of core microbiome species producing short chain fatty acids. In contrast, in less complex skin and nasal microbiomes, dysbiosis might involve the acquisition of opportunistic pathogenic species such as Peptoniphilus.

The Interplay Between Host Immune System and Microbiome Across Body Sites: Insights Into Insulin Resistance and Inflammation

The dynamics of microbial stability are likely linked to multiple host regulatory mechanisms, particularly the immune system44,129,141144. Using our customized bayes model145, we examined the longitudinal interplay between 62 host circulating cytokines/chemokines, growth factors and microbiome abundance at all four body sites. Based on credible interval, we identified 477 stool, 226 skin, 318 oral, and 221 nasal significant microbiome-cytokine associations that are body-site-specific (Fig. 4A, Table S4A). The stool and oral microbiome showed a significantly broader microbiome interaction spectrum than skin and nasal microbiome. (Fig. 4A). Interestingly, the cytokines associated with epithelial/endothelial growth and vascular inflammation (i.e., EGF, VCAM-1, IL-22), IL-1 family members (i.e., IL-1b, IL-1Ra), and leptin demonstrated the highest number of interactions with the microbiome. We further identified a subgroup of cytokines including IL-1b, IL-1Ra, MCP3(CCL-7), and IL-23 as the strongest correlative cytokines with the microbiome via effect size (Fig. S4A). The clear pattern of body-site-specific interactions may contribute to the taxa niche-specificity. For example, Moraxella shows a negative correlation with 23 cytokines on the skin, yet only with three in the nasal cavity (Table S4B). This reduced microbe-immune interaction in the nasal cavity may explain the higher prevalence of Moraxella in nasal7,146.

Figure 4. Systematic connections between circulating cytokines and microbiomes.

Figure 4.

A. Mixed-effects model outputs for cytokine-microbiome correlations; red lines for positive, blue for negative associations; circle size denotes significant correlation count. B. Cytokine-related genera percentages by phylum. C. Density plot of significant cytokine-microbiome correlation coefficients, compared by genera prevalence. D. Correlation coefficients by body site and phylum. p-values for positive versus negative associations were annotated in the middle. E. Hierarchical clustering of Spearman coefficients between cytokines and diverse genera. Significance indicated by asterisks: *p < 0.05, **p < 0.01, ***p < 0.001.

We next examined whether certain phyla are more frequently interacting with cytokines, potentially influencing their stability and individuality. Members of the Firmicutes phylum (primarily Clostridia) were significantly overrepresented among cytokine-correlated microbes across all body sites (Xstool2 = 19.343, Pstool = 1.092 × 10−5; Xskin2 = 10.418, Pskin = 0.0012; Xoral2 = 30.935, Poral = 2.668 × 10−8; Xnasal2= 31.396, Pnasal = 2.104 × 10−8) (Fig. 4B). This interaction may account for Firmicutes’ higher FS and lower DMI in the stool microbiome compared to other phyla. Additionally, an increase of Firmicutes is associated with conditions like obesity-related gut dysbiosis147, IBD-related oral dysbiosis148, and psoriasis-related skin dysbiosis149, which share inflammation as a common factor.

Interestingly, cytokines appear to play a pivotal role in shaping an individual’s core microbiome and in curbing the colonization of non-commensal bacteria, including many from the Proteobacteria phylum. At all four body sites we found that opportunistic microbes demonstrated a stronger correlation with cytokines than core microbiomes (see STAR Methods) (stool: W = 1,715, p-value = 1.181×10–12, skin: W = 223, p-value = 0.004041, oral: W = 1,055, p-value = 7.877×10–6, nasal: W = 482, p-value = 0.004691) (Fig. 4C). This correlation is largely driven by Proteobacteria rather than Firmicutes, as Proteobacteria consistently constitutes a larger segment of the opportunistic microbiome compared to the core microbiome (Fig. S4B). Proteobacteria often carry potent lipopolysaccharides (LPS) and instigate the downstream immune cascade150154. Previous studies125,155,156 have reported an increase in Proteobacteria abundance during inflammation. Contrarily, our study revealed that the correlations between cytokines and Proteobacteria abundance are mostly negative, except for Proteobacteria members in the nasal microbiome (Fig. 4D). The negative correlation of Proteobacteria is stronger in stool (W = 25,315, p-value = 0.04439), skin (W = 4,995, p-value = 0.005214), and oral sites (W = 9,443, p-value = 9.649×10–5), but weaker in nasal sites (W = 5,218, p-value = 0.2693). Furthermore, all cytokine correlations (n =10) from opportunistic stool Proteobacteria are negative, while many high prevalence Proteobacteria members exhibit positive correlations with cytokines (Fig. S4C). Our results suggest that inflammation might contribute to the adaptation of more prevalent Proteobacteria125,157.

The host response by cytokines and chemokines may be linked with the observed richness of bacterial genera, in addition to their relative abundance. We therefore examined the correlation between cytokines and the richness of the 20 most diverse genera per body site. The richness of several prevalent stool microbiome genera within the Bacteroidetes phylum, such as Prevotella, Phocaeicola, and Parabacteroides, form a cluster (Column 3, Fig. 4E) with primarily negative associations, echoing our finding that the relative abundances of Bacteroidetes members are more likely to be negatively correlated with cytokines (Fig. 4D). Also, leptin and GM-CSF, both strongly associated with BMI (Fig. S4D), show the strongest overall correlation with richness (Row Cluster F, Fig. 4E). This extends findings from numerous studies147,158160 on obesity’s influence on the gut microbiome diversity to additional body sites. Furthermore, we discovered that seven skin genera (Column Cluster 1, Fig. 4E) positively correlated with a cytokine cluster (Row Cluster A, Fig. 4E), indicating that the richness of specific skin genera (e.g., Rothia, Veillonella, Streptococcus) is positively associated with the level of plasma cytokines. Consequently, the observed increase in the skin microbiome richness in IR individuals (Fig. S6B) may suggest a diminished host selection of the skin microbiome during inflammatory periods.

Overall, our analysis elucidates the interplay between chronic low-grade inflammation and microbial dynamics across various body sites. It connects the immune system with microbiome composition and individualization, and shed lights on how these dynamics are both influenced by and contribute to insulin resistance.

The Microbiome is Highly Connected with Host Molecules: Unraveling the Role in Insulin Resistance and Inflammation

To comprehensively explore the relationship67,161,162 between the microbiome and internal host molecules and its role in IR, we analyzed the correlations between microbiome genera and plasma proteins, lipids, and metabolites in the host. We first modularized the lipidomics data to address its high collinearity (Fig. S5A, Table S5A), then applied a linear mixed model to residualize all omics data (see STAR Methods), reducing between-individual variation and emphasizing longitudinal intra-individual relationships.

Interestingly, the microbiome-host molecule interaction network partitions according to internal molecular composition rather than body sites of the microbiome (Fig. 5A), suggesting that certain taxa are primarily influenced by internal molecules interactions over influencing host molecular composition. Notably, three enterotypes driving taxa: Bacteroides, Prevotella, and Unclassified Ruminococcaceae, exhibit a clear preference for the lipidome, proteome, and metabolome regions, respectively (Fig. 5A, Table S5B). The close association between Prevotella and proteins has been previously documented163,164, as well as the relationship between Bacteroides and lipids165,166. However, our findings reinforce this understanding to include both additional taxa and multiple body sites, suggesting that these connections are not only site- and taxa-specific but also systemic and robust.

Figure 5. Interactions between plasma metabolites, lipids, proteomics, and microbiome over time.

Figure 5.

A. Correlation Network shows links between microbiome genera relative abundance and plasma analytics, color-coded by type (Microbiomes: Dark yellow, Blue, Dark red, Green; Plasma analytics: Dark blue, Orange, Red). B. Plasma analytics-microbiome relative abundance correlation summary of Fig. 5A. C. Correlations between genera and the metabolite ethyl glucuronide. D. Plasma analytics-microbiome relative richness summary of Fig. S6A. E. Correlations between genera and the metabolite p-Cresol glucuronide.

Many taxa-molecule interactions were consistent across body sites, validating the robustness of these relationships. The skin microbiome exhibited the broadest connectivity in the microbe-lipidome association, whereas the stool microbiome was mainly connected to the metabolome and proteome, as expected167,168. (Fig. 5B). Proteome pathway annotation further supported these connections and their potential functional implications. The skin microbiome-related proteins were enriched for pathways regulating lipid transport and metabolism, whereas the stool, nasal, and oral microbiomes were more strongly connected to the host immune response, including complement activation and humoral immune response. The oral microbiome exhibited significant associations with proteins linked with regulation of proteolysis, hydrolase activity, and enzyme, peptidase, and lipase inhibitor activity. (Fig. S5B). Strikingly, the complexity of the network between the stool microbiome and host molecular relationships was significantly reduced in individuals with IR (Fig. S5C), indicating a loss of balance in these individuals.

Our study reveals several metabolites, such as alcohol associated metabolite ethyl glucuronide169, interact with microbiomes across all four body sites, demonstrating a more global effects of alcohol beyond the gut and oral microbiome170172. Notably, skin Neisseria and Klebsiella, recognized for their acetaldehyde-173,174 and ethanol-producing175,176 potential, showed a positive correlation with plasma ethyl glucuronide level (Fig. 5C). Conversely, Faecalimonas, typically an acetate-producing bacteria177,178 and sensitive to alcohol179, exhibited a significant negative correlation with ethyl glucuronide. Furthermore, Desulfovibrio, a key genus promoting microbiome-related metabolic syndrome180,181, was positively correlated with ethyl glucuronide. These findings suggest that alcohol metabolism is associated with microbial dysbiosis across various body sites.

We also observed notable interactions between microbiome genera richness, a recognized indicator182,183 of host metabolic status, and internal plasma analytics. These interactions were most pronounced in the stool microbiome, likely due to its high richness. (Fig. 5D, Fig. S6A, Table S5C). Intriguingly, we discovered a positive association between p-Cresol glucuronide, a bacteriostatic184 metabolite produced exclusively by the anaerobic gut microbiome and linked to insulin resistance27,185, and the richness of Unclassified Ruminococcaceae and Oscillibacter in the stool. Conversely, it was negatively associated with genera typically considered metabolically beneficial, such as Roseburia186188. (Fig. 5E) These findings suggest that p-Cresol glucuronide may influence the development of insulin resistance-related dysbiosis by altering host tolerance to certain genera belonging to Clostridia.

In conclusion, our results not only validate several previous findings but also generate more host-microbial associations, enhancing our understanding of the complex interplay between the human microbiome, metabolites, and host health.

Elucidating Microbiome-Mediated Effects on Clinical Phenotypes: A Comprehensive Mediation Analysis across Four Body Sites

We next investigated potential causal linkages between microbes, host molecules and clinical phenotypes using mediation analysis189,190. This method measures the contribution of mediators, such as plasma omics or cytokines/chemokines, to the assumed191 microbiome-clinical phenotype (Table S6A) connection, providing confidence levels by inverting the roles of mediator and dependent variable56,189,192. Using this assumption, we identified 330 directionally significant mediation effects involving microbial taxa across all sites, with 207 and 164 of these mediation effects detected in IS and IR participants, respectively (Table S6B) These outcomes suggest potential microbiome-host causality exist and vary with IR status, though unaccounted confounders remain possible.

Compared to IS, we observed a significant decrease in cytokine-mediated effects of stool microbiome on hematologic parameters (p-value = 0.002) and metabolome-mediated effects of stool microbiome on immune phenotypes (p-value = 0.002) in IR individuals. Furthermore, there was an absence of lipidome-mediated associations between skin microbiome and host plasma lipids/cholesterol in IR participants (p-value = 0.031), suggesting a dysregulation in specific microbiome-metabolic interactions related to hematologic/immune and lipid/cholesterol homeostasis in IR. (Table S6C) In contrast, the oral microbiome mediated a large proportion of immune profiles via the modulation of lipidome (p-value = 0.035) and cytokines (p-value = 0.0001) in IR relative to IS participants, primarily through a negative relationship among major oral core microbiomes such as Veillonella (Table S6B), These causal relationships align with previous findings suggesting that diabetes-related oral dysbiosis may arise from a combination of the loss of core commensal oral taxa34,37,193 and an increase in the pathogenicity of resident oral bacteria during impaired glucose metabolism, as demonstrated by human observational studies194,195 and animal research36,40.

DISCUSSION

This study presents a systematic analysis of longitudinal multi-site microbiome ecology and host dynamics. With date-matched microbiome and host omics, we not only expand our knowledge of the stability and individuality of microbiome from various body sites, but also provide mechanism-generating hypotheses on host-microbiome interactions in the context of prediabetes. We demonstrate a number of interesting observations: 1) There is a “core” microbiome that is highly stable over time and an opportunistic microbiome that is highly variable and more potent to the immune system; these are niche-specific. 2) Correlations between microbiomes across body sites and extensive interactions with host factors indicate systemic coordination and interactions across the human body. 3) Highly individualized microbiomes are associated with distinct environmental factors (i.e., season, diet, chemical and biological exposome) in a niche-specific manner. However, these effects do not override the variance contributed by individuals, suggesting that the host is still the largest confounding factor for the variation observed in the microbiome. 4) Individuals with IR have a less stable microbiome with more diverse microbiome members on skin, possibly associated with upper respiratory infection, as well as significantly altered host-microbiome interactions.

Our study revealed that the stool and oral microbiomes exhibit the highest level of individualization, likely due to the unique influence of personalized dietary habits and host-specific factors, such as IR, impacting the digestive system196. Meanwhile, the skin and nasal microbiomes are less individualized, possibly owing more to individual environmental exposure90,113. We observed site-specific impact of environmental factors, such as season, on the microbiomes of these four body sites. For instance, a decrease in stool microbiome richness in late summer corresponded with previous findings of worsened insulin sensitivity during this period197,198. Similarly, we noted a decline in the richness and evenness of the oral microbiome from late summer through winter, suggesting a potential influence of environmental factors like the availability of fresh food and changes in sunlight durations. Meanwhile, changes in humidity from January to April might explain the richness increase in skin and decrease in nasal microbiome 90. These observations underscore the critical role of the host in modulating the microbiome in response to environmental fluctuations.

Leveraging dense longitudinal sampling, we were able to quantify the stability and degree of individuality of the microbiomes at different body sites. The strong correlation between microbiome stability and individuality suggests an active role of the host in the establishment of commensal bacterial populations. Notably, this individuality appears to be taxon-specific, pointing to the possibility that the stability of an individual’s microbiome may be influenced by the dominant bacterial taxa they carry. For instance, the higher DMI in stool Bacteroidetes links traditional Bacteroidetes/Firmicutes ratio or enterotypes to new insights in the context of metabolic disorders117,199. Furthermore, micro-biotypes across different body sites within the same individual correlate. Individuals with high levels of Bacteroides in the stool also have high Cutibacterium on the skin and Prevotella in the oral microbiome. Combined with our findings on the correlation between microbiome individuality and stability, the low-level engagement between the core microbiome and cytokines, and the clear host molecular type-specific patterns in the host-microbiome interactome, we propose that the colonization and adaptation of bacteria across multiple body sites is not random but precisely regulated by host factors.

Niche-specific traits often emerge through multi-site comparisons; for instance, we identified unique characteristics of the oral microbiome. The recurrence rate of oral bacteria does not increase with DMI, possibly due to a stable and conserved community structure within the oral microbiome 83,200. This, coupled with the oral microbiome’s high intra-site interdependence and minimal correlation with other body sites, underscores its specialization. The unique oral environment, shaped by factors such as saliva, teeth, gums, tongue, and distinct nutrient availability, likely contributes to this specialization201,202.

Microbial individuality and stability are closely related to the host immune system, which is well known to interact with microbes at multiple body sites141,203,204. This interaction modulates both the colonization of microbes, as well as their functional benefits (e.g., epithelium barrier integrity maintainance44,205). Our Bayesian model reveals that the interactions between the microbiome and cytokines, while present, are subtle. Certain genera exhibit an approximate 1.5-fold change in response to cytokine variations. The interaction between inflammatory cytokines and the microbiome demonstrated that low prevalence genera (i.e., stool Proteobacteria) are likely reduced during host inflammatory events. We also revealed a systematic relationship between cytokines and the genera complexity of the microbiome at each body site. Notably, the diversity within a subset of the skin microbiome positively correlates, while that within the stool microbiome negatively correlates with the same group of cytokines. Given the established association of IR with low-grade chronic inflammation206, the observed changes in diversity among IR individuals might be related to their unique cytokine profiles.

We also identified a list of interactions between host plasma biomolecules and microbiomes (Table S5B). Some of the relationships, such as alcohol metabolites and the gut microbiome, have been documented207,208. We also find many correlations across body sites with host factors; for example, the correlated Bacteroides in the stool and Cutibacterium in the skin are both closely related to lipid metabolism209213. Our multi-omics analyses suggest potential causality of host factors in these relationships, reinforcing the idea of host-driven systematic microbial coordination across body sites as proposed in gut-brain axis 214,215 and gut-lung axis216,217 theories. Since many of the metabolites, lipids and proteins are signaling molecules (e.g., chemokines, hormones, peptides), these molecules may play important roles in organismal communication across the entire host-microbiome ecosystem. Importantly, this interaction presumably occurs individually, and significantly altered at disease stage.

Intriguingly, we found Klebsiella on skin was positively associated with metabolites of alcohol. This indicates that alcohol intake may change the host into a Klebsiella-tolerant environment, resulting in the adaptation and expansion of pathogenic Klebsiella as previously described218. This observation supports previous findings in alcohol-associated pneumonia, where alcohol consumption and increased susceptibility to Klebsiella in the lungs may be a result of either intestinal Klebsiella-specific T-cell sequestration218,219 or alcohol-related impairment of tryptophan catabolite production/processing in the gut microbiome, which restricts pulmonary immune cell trafficking220,221.

Insulin resistance (IR) appears to disrupt the intricate balance between the host and microbiome, demonstrated by an unstable, dysbiotic microbiome in IR individuals. In our study, we discovered marked differences in the microbiome composition, diversity, and core members of the stool and skin microbiomes in IR individuals compared to their insulin-sensitive counterparts. Notably, the systematic shift in microbiome prevalence indicates an entire microbial community’s transformation instead of the abnormality of a few isolated members (Fig. S1L). This dysbiosis can potentially alter the complex host-microbiome interaction in IR subjects. Consistently, we detected reduced host-microbe coordination and a missing association between oral and skin microbiome stability. Further, our mediation analysis validates our result about the reduced gut microbiome-cytokine interaction in IR individuals70,145, and revealed heightened pro-inflammatory signals linked to the microbiomes of the upper respiratory tract (oral and nasal microbiome) in IR individuals. Our in-depth exploration of acute microbiome changes during infection provides additional substantiation for this theory. We observed a significant increase in IR-enriched skin genera such as Peptoniphilus and Intrasporangium (Fig. S3D) during the course of a respiratory viral infection, while a decline in specific genera in the stool microbiome known for butyrate production, like Clostridium_IV, Lawsonibacter, and Intestinimonas222225. While these findings do not explicitly validate the hypothetic link226 between respiratory viral infection, chronic metabolic dysregulation, and related complications, they suggest that infection-related microbial shifts may be associated with, or even cause, the dysbiosis and complications of metabolic syndrome as observed in animal models227.

Limitations of the Study

Caution is warranted in interpreting our results due to inherent limitations. The DMI and FS calculations primarily rely on the BC dissimilarity. Therefore, study design such as sample size, population representation, choice of dissimilarity metric, and genus internal complexity may impact the accuracy. Additionally, although DMI can provide a quantitative measure of microbiome individuality, understanding its biological or ecological significance can be challenging due to the potential influence of host genetics, environmental factors, and host regulation.

When the study began, 16S rRNA sequencing was highly efficient for analyzing microbiomes rich in human DNA, such as those in nasal and skin swabs. However, our team, along with others, later identified its limitations in accurately capturing specific bacterial genera in stool and skin microbiomes when focusing on only certain variable regions.73,228. Furthermore, unlike metagenomic sequencing, 16S analysis does not provide information on viruses and fungi within a microbiome community, nor does it reveal gene function potentials. Also, our study does not capture a collective profile of the entire skin or oral microbiome, given the distinct characteristics of species at various sites7,62,119. Instead, it represents the microbiome from a single swab taken from each indicated body site. Furthermore, the geographical and lifestyle constraints of our participants could restrict the broad applicability of our conclusions to more diverse and complex populations229,230. Moreover, we note that our analyses primarily determine associations or statistical mediation effects, using p-values as cut-offs. Despite the bolstered study power afforded by a longitudinal design231,232, some observations with a smaller sample or effect size may still be overlooked. Analysis of larger cohorts may reveal further insights. Additionally, the use of p-values has its limitations regardless of the cut off233, such as its dependence on sample size,243, the potential for misinterpretation234,235, and an inflated type 1 error rate with multiple tests236,237. Finally, while our study highlights notable associations and suggests potential causations, it does not establish causation. Our mediation analysis aims to infer directionality and potential causality, but unobserved factors could influence these findings. To confirm causative links, more targeted follow-up studies are essential.

In spite of these limitations our studies provide a number of observations concerning the individuality and stability of the microbiome across multiple body sites during health and disease and in individuals with different IR/IS status. These observations have important implications in modulation human molecular health using personalized prebiotics and probiotics. Our data also provides a valuable and unique resource for the general scientific community.

STAR METHODS

RESOURCE AVAILABILITY

Lead Contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Michael P. Snyder (mpsnyder@stanford.edu).

Materials availability

This study did not generate new unique reagents.

Data and code availability

The microbiome data are available at https://hmpdacc.org. Other omics data can be accessed through Stanford iPOP website at https://med.stanford.edu/ipop.html.

The microbiome data specific to this study can be accessed at GitHub Repository (https://github.com/xzhou7/iHMP). The metadata corresponding to these data files can be found at Stanford Data Repository (http://hmp2-data.stanford.edu/script.php?table=subject).

Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Human Cohort

Our study was conducted as part of the integrated Human Microbiome Project of the National Institutes of Health (NIH). This human study was approved by Stanford University’s Institutional Review Boards (#23602), ensuring ethical compliance and participant safety. Informed consent was obtained from all participants, who were either at risk for type 2 diabetes or voluntarily interested in diabetes-related research.

Eligible participants were either at risk for type 2 diabetes or voluntarily interested in diabetes-related research. Exclusion criteria encompassed hypertriglyceridemia > 4.0 mg/ml−1, uncontrolled hypertension, uncontrolled psychiatric disease, previous bariatric surgery, pregnancy or lactation, eating disorders (i.e., binge eating disorder, anorexia nervosa, or bulimia nervosa), alcohol use disorder, or failure to provide five consecutive samples from at least one body site (stool, skin, nasal, or oral). Consequently, data from 86 participants met the criteria for inclusion in this analysis.

Regarding the allocation of subjects to experimental groups, participants were not divided into separate experimental groups per se, but rather contributed samples for comprehensive microbiome analysis. This approach was designed to capture a broad spectrum of microbiome data across different body sites. In our analysis, we considered the impact of various factors, including biological age, gender, self-reported ethnicity on the study’s outcomes. These factors were included in our linear mixed model for intra class correlations. Members within the same household were compared for the family score computation.

METHOD DETAILS

Microbiome Sample Collection and Sequencing

Stool samples were self-collected by participants and other samples were collected by study coordinators following iPOP study standard operating procedures (SOP), as adapted from HMP_SOP corresponding sections (HMP_MOP_Version12_0_072910)70. Briefly, retroauricular areas were rubbed with pre-moistened swabs under pressure for skin sampling, anterior nares for nasal sampling, and rear of the oropharynx for oral sampling. Samples are stored at −80 C immediately after arrival. Stool and nasal samples were further processed and sequenced in-house at the Jackson Laboratory for Genomic Medicine (JAX-GM, Farmington, CT, USA) and detailed methods are described previously70, while oral and skin samples were sent to uBiome (uBiome, San Francisco, CA, USA) for further processing.

After 30 minutes of beads-beating lysis, skin and oral samples were processed using a silica-guanidinium thiocyanate-based nucleic acid isolation protocol242244 on a liquid-handling robot. The 16S rRNA variable region V4 was amplified by 35 cycles of PCR using the primer 515F (5’-GTGCCAGCMGCCGCGGTAA-3’) and 806R (5’-GGACTACHVGGGTWTC TAAT-3’)238. The DNA from each sample was barcoded and combined to create a sequencing library. The sequencing library was then purified using columns and microfluidic DNA fractionation245 to reduce unwanted DNA fragments. Bio-Rad MyiQ was used to quantify the DNA concentration of the library using the Kapa iCycler qPCR kit (Bio-Rad Laboratories, Hercules, CA, USA). Sequencing was performed on the Illumina NextSeq 500 Platform (Illumina, San Diego, CA, USA) via 2 * 150 bp paired-end sequencing protocol246.

Raw sequencing data from the stool samples and nasal samples are acquired from our previous publication70. Briefly, 16S rRNA gene from V1~V3 hyper-variable region was sequenced with primer pair of 27F (5′-AGAGTTTGATCCTGGCTCAG-3′) and 534R (5′- ATTACCGCGGCTGCTGG-3′) and being barcoded and sequenced on the Illumina MiSeq sequencing platform through a V3 2 × 300 sequencing protocol. The same cutoff used in skin and oral sequencing data was applied to stool and nasal sequencing data in demultiplexing. After demultiplexing, reads with Q-scores less than 35 and ambiguous bases (Ns) are trimmed for additional analysis.

Microbiome data processing

Demultiplexed sequenced samples were saved as FASTQ files using BCL2FASTQ software (Version 2.20, Illumina, CA, USA). Sequences with barcode mismatches, primer mismatches exceeding one, or Q-scores below 30 were excluded. Due to low overlap between forward and reverse reads according to FLASH (Version 1.2.11), only forward reads were selected for further processing.

Microbiome sequencing data from four body sites were combined and processed using the DADA2 R package (version 1.16)239. Sequences were filtered to remove ambiguous bases (maxN=0) and those with more than two expected errors (maxEE=2). After filtering, inter-sample composition analysis was performed based on the learned error rate. An amplicon sequence variant (ASV) table was constructed, and chimeras were removed using the DADA2 workflow consensus method. Reads passing all filters were aligned against a trained database of target 16S rRNA gene sequences and taxonomic annotations derived from Version 18 of The Ribosomal Database Project (RDP)247 Taxonomy release (Aug 14, 2020). Relative ASV abundance was determined by dividing the count associated with that taxon by the total number of filtered reads. Samples with depths below 1,000 reads were removed due to insufficient sequencing depths248 following the HMP consortium standard15,70. The Local Outlier Factor (LOF) of each point was calculated on a sequencing depth to richness (observed ASV) plot, and samples with a LOF greater than 3 (n=7) were removed due to an abnormal richness-sequencing depth relationship. The average sample sequencing depth after quality control was 23,554 for stool microbiome, 74,515 for skin microbiome, 132,912 for oral microbiome, and 24,899 for nasal microbiome. Batch effects were estimated using PERMANOVA analysis when both batch and subject ID were included, with results showing that the total variance explained by the 23 batches was 2% for stool, 3% for nasal, 0.6% for oral, and 3.6% for skin samples. The batch effects were considered small, and no correction for batch effects was applied in the analysis.

Lipidomics analyses

Lipid extraction and data generation were performed as previously described249251. Briefly, complex lipids were extracted from 40 μL of EDTA-plasma using a mixture of methyl tertiary-butyl ether, methanol, and water, followed by biphasic separation. Lipids were then analyzed using the Lipidyzer platform, which consists of a DMS device (SelexION Technology, Framingham, MA, USA) and a QTRAP 5500 (Sciex). Lipids were quantified using a mixture of 58 labeled internal standards provided with the platform (cat# 5040156, Sciex, Redwood City, CA, USA), and lipid abundances were reported in nmol/g.

To address the high collinearity of the lipidomic data, a customized clustering method was designed. Specifically, the lipidomics data were divided into six clusters using Fuzzy c-means clustering (R package “Mfuzz” (version 3.15)). For the lipids within each cluster, correlations were computed, and lipids with high correlative relationships (Spearman correlation > 0.8 and BH-adjusted p-values < 0.05) were grouped into the same module. Community analysis (‘fastgreedy.community’ function from R package “igraph” (v1.3.5)) was employed to detect the modules. For lipids not assigned to any of the modules, their original lipid species annotations were used for downstream analysis (Table S5A).

Metabolomics Analyses

Untargeted metabolic profiling was performed using a broad-spectrum LC-MS platform using a combination of reverse-phase liquid chromatography (RPLC) and hydrophilic interaction liquid chromatography (HILIC) separations and high-resolution MS70,252. Briefly, plasma metabolites were extracted following solvent precipitation using a mixture of ice-cold acetone, acetonitrile, and methanol (1:1:1, v/v). Hydrophilic metabolites were separated on a ZIC-HILIC (2.1 × 100 mm, 3.5 μm, 200 Å; Merck Millipore) while hydrophobic metabolites were separated on a Zorbax SBaq columns (2.1 × 50 mm, 1.7 μm, 100 Å; Agilent Technologies). Data was acquired on a Thermo Q Exactive plus mass spectrometer for HILIC and a Thermo Q Exactive mass spectrometer for RPLC. Raw data were processed using Progenesis QI (v2.3, Nonlinear Dynamics, Waters) and metabolites were formally identified by matching fragmentation spectra and retention time to analytical-grade standards or matching experimental MS/MS to fragmentation spectra in publicly available databases. A total of 726 annotated metabolites were retained for downstream analysis.

Proteomics Analyses

Plasma proteins were characterized using a TripleTOF 6600 system (Sciex) via liquid chromatography-mass spectrometry (LC-MS) with SWATH acquisition, following the methodology outlined in a previous study70. In every injection, 8-μg of tryptic peptides, derived from undepleted plasma, were loaded onto a ChromXP C18 column (0.3 × 150 mm, 3 μm, 120 Å, Sciex). The separation of peptides was achieved through a 43-minute gradient ranging from 4% to 32% B. High sensitivity MS/MS mode was utilized to construct variable Q1 window SWATH Acquisition methods (100 windows) with Analyst TF Software (v1.7). Scoring of peak groups was performed with PyProphet (v2.0.1)253 and alignment of peak groups with TRIC254, each adhering to stringent confidence thresholds (1% FDR at peptide level; 10% FDR at protein level). The abundance of proteins was calculated as the cumulative sum of the three most abundant peptides.

Luminex Multiplex Assays for Targeted Cytokine, Chemokine, and Growth Factors

The evaluation of circulating cytokines, chemokines, and growth factors was undertaken employing established procedures from the Stanford Human Immune Monitoring Center (HIMC). Specifically, EDTA-plasma was scrutinized using a Human 62-plex Luminex multiplex assay, consisting of conjugated antibodies (Affymetrix, Santa Clara, California). The raw data obtained from the assay were normalized against the median fluorescence intensity (MFI) value. Subsequently, variance stabilizing transformation (VST) was applied to the data to eradicate the batch effect, adhering to our previously outlined methodology145. Measurements featuring background noise (CHEX) exceeding five standard deviations from the mean (mean ± 5 × SD) were omitted from the data.

Exposome and Associated Environmental Analyses

As previously outlined90,255, the process of data collection for the exposome and associated environmental elements proceeded accordingly. The chemical exposome was sampled using the RTI MicroPEM V3.2 personal exposure monitor (RTI International, Research Triangle Park, NC, USA) for two participants. The MicroPEM, an active air sampling apparatus, operates by circulating air at a rate of 0.5 L/min. It was modified to house a customized cartridge containing 200 mg of zeolite adsorbent beads (Sigma 2–0304, Sigma-Aldrich Corp., St. Louis, MO USA) positioned at the airflow’s termination to gather both hydrophobic and hydrophilic compounds. Each sampling session spanned approximately five days. Post-session, the cartridge was detached and preserved at −80 °C until subsequent processing. Chemical extraction involved the resuspension of zeolite beads in a sterile Eppendorf LoBind tube filled with 1 mL of Mass Spec grade methanol. Following a 20-minute incubation period at room temperature, the samples were subjected to a 20-minute centrifugation process at 22,000 g, also at room temperature. The samples were then analyzed using a Waters UPLC-coupled Exactive Orbitrap Mass Spectrometer (Thermo, Waltham, MA, USA), yielding a collection of 158 exposome chemicals. Environmental data, on the other hand, were sourced from several origins. Parameters such as temperature, humidity, and sampling flow rate were directly recorded by the MicroPEM. GPS coordinates of the participants provided geographical data. Additional meteorological and demographic information was obtained from public data repositories including the Climate Data Online (CDO), the US Census Bureau, and local weather stations. This culminated in the collection of 10 environmental feature data points.

Dietary Analyses

A total of 25 food items were included in a questionnaire that was completed voluntarily by participants during their routine visit in the study, using a diet questionnaire hosted on https://www.projectredcap.org/ from our previous report145. The frequency of consuming each food item was scored in downstream analysis. For breads, biscuits, cakes, pies and pastries, we score the frequency from 0~4, with the associated frequency per day: 0) less than 1, 1)1 per day, 2)2~3 per day, 3)4~5 per day, 4)6 or more. For other foods we ask the participants to state the frequency from 6+ times per day to less than once per month.

Insulin-Suppression Test

A subset of eligible consenting participants (N=58) underwent an Insulin-suppression Test (IST), as a measure of insulin-mediated glucose uptake, to evaluate the insulin sensitivity status. Following a 12-hour overnight fast, participants were administered an infusion comprising 0.27 ug/m2 min of octreotide, 25m U/m2 min of insulin, and 240 mg/m2 min of glucose over a three-hour period during their visit to Stanford’s Clinical and Translational Research Unit (CTRU). Blood samples were procured at ten-minute intervals during the final half-hour of the infusion, resulting in a total of four blood draws. These samples were analyzed to determine plasma glucose and insulin levels. The mean value of the four steady-state plasma glucose (SSPG) and insulin concentrations were subsequently calculated. Participants were then categorized based on their SSPG values256: those with SSPG < 150 mg/dl were classified as insulin sensitive (IS) (n=28), while those with SSPG ≥ 150 mg/dl were classified as insulin resistant (IR) (n =30). Participants who were unable to provide measurements due to personal or medical circumstances were allocated to an indeterminate group (Unknown) (n= 28).

Clinical Lab Test

Clinical lab tests were performed at the Stanford Clinical Lab following its guideline of blood and urine collection and submission (https://stanfordlab.com/test-directory.html). The test includes a metabolic panel, complete blood count panel, glucose, HbA1C, insulin measurements, hsCRP, IgM, lipid panel, kidney panel, liver panel. Detailed measurements and annotations are provided as a supplementary table. (Table S6A)

QUANTIFICATION AND STATISTICAL ANALYSIS

UMAP for Microbiome Distribution

The distribution of the microbiome was visualized using Uniform Manifold Approximation and Projection (UMAP), facilitated by the R package “Seurat (Version 4.0)”. Prior to the application of UMAP, the count data were first normalized to relative abundance and scaled to represent one million reads per sample. A total of 1524 variable features were identified and encapsulated within a Seurat object. From this point, a distance matrix was produced through the utilization of the R package “Vegan (Version 2.6–2)”, employing Bray Curtis dissimilarity. This distance matrix was subsequently transformed via Principal Coordinate Analysis (PCoA). The first ten dimensions from the PCoA (out of a total of 1094 generated) were used to determine neighboring relationships. The UMAP projection was then calculated, with default settings employed. This projection was made possible by invoking Python’s UMAP via reticulate. The final UMAP results were visualized using the first two dimensions.

Intraclass Correlation

Intraclass correlation Coefficient (ICC) was calculated from Linear Mixed Models, in which we modeled random intercepts but a fixed slope, allowing different personal levels between individuals70. We first linearly transformed each analyte (when applicable) and standardized the total variation to 1 before applying ‘lmer’ function from R package “lme4 (V1.1–30)”, with the formula as:

Exp1+Days+(1SubjectID)

Where Exp was the linearly transformed and standardized values of each analyte, Days was the length of time individuals participated in the study, SubjectID was the subject ID associated with each participant.

We then used ICC as the proportion of total variation explained by subject structure in the cohort by VsubjectID/Vtotal, in which V was the variance from the corresponding component extracted by VarCorr and Vtotal was 1.

Permutation Test on Bray Curtis Distance Comparison

The Bray-Curtis (BC) distance was used to quantify the degree of similarity between two microbiome samples, with the ASV serving as the unit for calculating dissimilarity for the complete microbiome sample or for specific taxa. Similarity metrics were calculated pairwise for both intra-individual and inter-individual comparisons. A permutation test was employed to estimate the null distribution while accounting for the varying sample sizes of each participant. The null hypothesis being tested was that there is no difference between intra-individual and inter-individual distances.

For each microbial genus, a test statistic was calculated as the mean difference in BC distances between intra-individual and inter-individual comparisons. To estimate the null distribution, all sample labels were randomly permuted, and the BC distances were computed pairwise. This process was repeated 10,000 times, generating a null distribution of test statistics.

P-values were then calculated by determining the proportion of permuted test statistics that were at least as extreme as the observed test statistic. In the case of multiple comparisons, such as for different microbial genera, p-values were adjusted using the BH procedure to control the false discovery rate. Statistical significance was determined using a threshold of BH adjusted p-value < 0.1.

Degree of Microbial Individuality

Degree of Microbial Individuality (DMI) was measured as the mathematical difference between a given genus regarding their populational median of the inter-individual BC distance and the median of the intra-individual BC distance. To assess the robustness and variability of the DMI estimates, we employed a bootstrap resampling technique257.

First, we summarized the between-sample distance of genera with longitudinal prevalence greater than 10% at a given body site. The distance of sample pairs was then allocated into inter-individual or intra-individual groups. For each genus, we resampled the data with replacement and computed the DMI using the following formula:

DMIi=BCinter-individual-BCintra-individual

This resampling process was repeated multiple times to obtain a distribution of DMI estimates, which enabled us to assess the variability and robustness of our results.

By employing the bootstrap resampling technique, we aimed to gain insights into the reliability of our DMI calculations and understand how sensitive they were to potential variations in the data. This approach provided a more comprehensive understanding of the degree of microbial individuality across various body sites and taxa. To ensure the accuracy of our results, we considered a DMI to be reliable only if that DMI’s standard deviation (SD) of the bootstrapped distribution is less than 15 times of the mean DMI. This criterion helped to filter out any DMI estimates that might be influenced by high variability or uncertainty in the data. Detail of the bootstrap result was included in our supplementary materials (Table S2C), where we used column “confidence” to distinguish DMI values with high or low reliability. The total number of genera reported per region after bootstrapping included: 105 for stool, 35 for skin, 63 for oral, and 33 for nasal samples.

To quantify the cumulative DMI for each individual, the DMI score was multiplied by the average relative abundance of each genus for a given individual. This generated a weighted DMI (abundance_dmi) that represented the product of the DMI score and the genus’s relative abundance. The total DMI for each individual was computed by summing these weighted DMI values across all genera. This approach offered a comprehensive measure of the overall DMI per individual, accounting for the contribution of each genus weighted by its relative abundance in the individual’s microbiome.

Family Score

To assess the impact of a shared living environment on microbiome variability, we introduced a metric called the Family Score (FS). The FS represents the relative influence of a shared environment on the inter-individual dissimilarity of a given genus within cohabitating pairs. We began by excluding genera with a longitudinal prevalence of less than 10%, leaving 141 for stool, 119 for skin, 41 for oral, and 33 for nasal samples. For each remaining genus, we computed the “within-family” inter-individual Bray-Curtis (BC) distance between cohabiting pairs. We also calculated the median inter-individual BC distance (BC inter-individual) and intra-individual BC distance (BC intra-individual). Using these values, we computed the FS for a given genus ‘i ‘ with the following formula:

FamilyScorei=BCinter-individual-BCintra-family/BCinter-individual-BCintra-individual

This formula normalizes the FS to a scale that allows for comparisons across families and non-families. An FS of 0 indicates that the shared living environment has no impact on inter-individual dissimilarity, while an FS of 1 suggests that living in the same environment causes inter-individual dissimilarity to resemble intra-individual dissimilarity. We truncated the FS at 0 and 1 to maintain consistency in the comparison scale. Any FS values greater than or equal to 1 were assigned a value of 1, and values less than or equal to 0 were assigned a value of 0.

Classification of The Microbiome Genera by Their Longitudinal Prevalence

The microbiome genera were categorized as the core microbiome, opportunistic microbiome, and middle group based on their longitudinal prevalence258260. Calculation of prevalence was based on the presence or absence of reads from each sample. For each sample, the relative abundance of each genus was first transformed to 1 if it was greater than 0; then, the proportion of 1 for each genus in each participant was determined as the longitudinal prevalence. Then the genera were assigned to a group based on their longitudinal prevalence: core microbiome: longitudinal prevalence > 80%; middle group: 20% ≤ longitudinal prevalence ≤ 80%; opportunistic microbiome: longitudinal prevalence < 20%.

Body Site-Specific Longitudinal Model for Bray-Curtis distance

To estimate the effect of body site on the change in BC distance over time, we used a linear mixed effects regression with the BC distance as the response variable implemented in the “lme4” package in R261. The BC distance of all pairwise samples was transformed to a more normally distributed dataset. The transformation method used was the log-log transformation, which has been widely used in survival analysis studies. Specifically, the transformation was performed by applying the formula:

transformed_dist=log10(-log10(1-dist))

A total of eight different transformations were evaluated (variations of logarithmic, square root, arcsine, and ratio transformations of the original distance) to identify the most suitable transformation for our analysis. To assess the normality of the transformed distances, an Anderson-Darling test was performed, and the best method was selected by Anderson-Darling’s statistics A262.

Fixed effects included an interaction between the body site and time, and with a random intercept for each individual. Time was normalized to the days from the first sample for each individual. The model was optimized using the “nlopwrap” method263 and nested models were compared by likelihood ratio test using the “lmtest” package in R264. Differences in the slopes of the body sites over time were assessed by F-test with Satterthwaites’s degrees of freedom265. For the comparisons between body sites, the model was constructed as:

dist-1+diffdays*dataset+(1subject_id)

For the comparison between IR and IS participants, the model was constructed as:

dist-1+IRIS*diffdays+(1subject_id)+(1+IRISsubject_id)

Where dist was the pairwise BC distance, diffdays was the date interval between two samples, subject_id was the subject ID associated with each participant, and IRIS was the insulin sensitivity status of each participant.

Bayesian Mixed-Effects Model for Microbial Taxa and Cytokine Interactions

We utilized a Bayesian negative-binomial longitudinal mixed-effects model to analyze the interactions between microbial taxa and cytokines. This model choice addresses several key characteristics of our data: the compositional nature of microbiome data, the presence of zero-inflation and high skewness in cytokine measurements, and the repeated measurements of both cytokine and microbiome data in our study cohort (n=62 with longitudinal, date-matching measurements). The model’s capacity to handle the highly skewed cytokine data was a critical factor in its selection, especially given the dramatic range of cytokine surges observed in our dataset, ranging from a 10.8-fold increase for MIP1B to as much as a 618-fold increase for LEPTIN. Each genus’s reads count was modeled as a sparse-matrix response variable, with a plasma cytokine level MFI quantity and time as fixed effects and a random intercept for each individual, following the formula:

M-i=X-iβ+Z-ib-i+ε-i

Where Mi is a vector of the genus-level microbe relative abundances for each participant i,Xi is the design matrix for the fixed effects, β. Each row of matrix Xi contains the terms (1) time (days post–study start), Di, and (2) cytokine measurements, Yi, from 1 to n. Zi is the random effects design vector of 1’s denoting a random intercept, bi is a scalar for each participant, and εi is a zero-centered error term.

Posterior sampling was performed using four chains, 5,000 iterations per sample, and a 1,000-iteration burn-in of a No-U-Turn Sampler implemented in the “brms (Version 2.18.0)” package in R266269. The iteration plots and posterior predictive distributions were visually inspected for chain convergence. Microbe genera and cytokines above the limit of detection in less than 10% of the samples were excluded from the analysis. A microbe-cytokine association was considered significant if the 95% credible interval on the fit coefficient of the cytokine term did not include zero.

In our model, the beta represents the effect size. We estimated the variance explained by the model using the “bayes_R2” function from the brms package, and this information is included in the final table. While we provide p-values for each pair, as inferred from a method detailed previously270, and the subsequent BH-adjusted p-values, it’s important to note that these are supplementary. In our Bayesian analysis, the determination of significance is based on the commonly practiced use of credible intervals derived from Markov Chain Monte Carlo (MCMC) sampling.

Correlation Network Analysis.

Correlation network analysis was conducted to construct a network between the microbiome (from stool, skin, oral, and nasal samples) and internal multi-omics data (proteome, metabolome, and lipidome) from plasma, following a modified version of published methods70. Initially, time points with unmatched collection dates for each pair of microbiome and internal omics data were excluded. Subjects with fewer than five samples for a specific microbiome type were also removed from the corresponding correlation analysis.

The microbiome data, which included relative abundance or observed ASV richness at the genus level, was processed by retaining only genera detected in at least 10% of all samples. Centered log ratio (CLR) normalization was applied to address compositionality in microbiome data using the R package “compositions” (Version 2.0–4). Proteome, metabolome, and lipidome module data were log2 transformed.

To account for repeated sampling from the same subject, a linear mixed-effects model was utilized, incorporating subject ID as a random effect using the R package “lme4” (Version 1.1–30). Spearman correlations were then calculated, and p-values were adjusted using the Benjamini & Hochberg method. For correlation network construction, BH adjusted p-values < 0.2 were included. In the linear mixed model assessing longitudinal effects, the estimated correlation coefficient served as the measure of effect size (Fig. S6B).

To gauge the robustness of the point estimation for the proportion of microbiome genera that are significantly correlated, we employed a bootstrapping approach. Specifically, we determined the percentage of significantly correlated genera by dividing the number of significantly correlated pairs by the total possible pairs. We bootstrapped this procedure 20 times to obtain mean and standard deviation values for the percentage of significantly correlated microbiome genera. These values were then used to compare the scale of interdependency of microbiome across different body sites. To ascertain if one body site exhibits a significantly greater interdependency compared to another, we employed the two-sided Wilcoxon rank-sum test. The significance of these comparisons was determined using p-values adjusted for multiple testing using the BH correction.

The estimation of inter-individual microbiome-wide correlation was conducted utilizing the SparCC method (R package discordant, Version 1.25.0), a methodology specifically tailored for compositional data241,271. To prepare the microbiome relative abundance data for this analysis, the data were multiplied by 20000 and subsequently rounded, a process designed to convert proportions into data resembling counts. Following this, the SparCC method was employed to compute the correlation matrix. This matrix was then transformed into a long format data frame, with each row representing a pair of genera and their corresponding correlation coefficient. To assess the statistical significance of the observed correlations, a permutation test was implemented. For each pair of genera, the sample labels were randomly permuted, and the permuted SparCC correlation coefficient was calculated. We executed this procedure 10,000 times to produce a null distribution of SparCC correlation coefficients. Following this, we counted the instances where our observed correlation coefficient was encompassed within this null distribution. A finding was deemed “non-significant” if the correlation coefficient from any of the 10,000 permutations was more extreme than the coefficient from our original analysis, and subsequent raw p-value was computed based on the permutation results. Results with the BH adjusted p-value < 0.1 from this permutation was included in our final report.

The inter-omics correlation network was visualized using the R packages “ggraph” (Version 2.0.5), “igraph” (Version 1.3.2), and “tidygraph” (Version 1.2.1) under the “kk” layout.

Pathway Enrichment for Proteins Correlated with the Microbiome.

The enrichment of pathways corresponding to proteins associated with the microbiome from four different body sites was achieved via the R package “clusterProfiler (Version 3.15)”. The proteins in question served as input for pathway enrichment, specifically through Gene Ontology (GO) processes, enabling the identification of statistically significant pathways. The determination of these pathways was reliant on Fisher’s exact test.

GO terms of interest were isolated based on their Benjamini-Hochberg adjusted p-values; terms with p-values below 0.05 were retained for ensuing analyses. For the GO terms that demonstrated significant enrichment, the R package “simplifyEnrichment” was employed. This package facilitated the calculation of similarities between each pair of GO terms. The construction of a network only incorporated edges that exhibited similarities exceeding 0.70.

In order to discern modules within the correlation network, community analysis was undertaken utilizing the R package “igraph (Version 1.3.4)”. To encapsulate each module, only the GO term that presented with the smallest Benjamini-Hochberg adjusted p-value was preserved.

Mediation Analysis.

A mediation analysis was conducted to investigate the potential influence of microbiomes from stool, skin, oral, and nasal sources on phenotypes through internal multi-omics data, including proteome, metabolome, lipidome, and cytokine56,272,273. Phenotype data were obtained via clinical laboratory tests of plasma samples. The associations between the microbiome and phenotype (Direct Effect), as well as internal omics data (Indirect Effect), were initially determined as outlined in the ‘Correlation network analysis’ section. Only significant associations (BH adjusted p-values < 0.2) were considered for subsequent mediation analysis. The linear regression model from R package “mediation” was employed for the mediation analysis. Ultimately, pairs with significant Average Causal Mediation Effects (ACME, p-values < 0.05) were reported, representing the microbiome’s impact on phenotype measurements through internal multi-omics.

To control the false discovery rate (FDR), a reverse mediation analysis was performed by exchanging the mediator with effects (i.e., the microbiome influencing internal omics data via phenotype), and pairs with significant ACME in reverse mediation (p-values < 0.05) were excluded from the final results. Comparisons between body sites and insulin sensitivity statuses were conducted for each dataset using the Fisher exact test. Initially, it was hypothesized that the true meditative effect of each mediated pathway (e.g., genusi~cytokinej~immunek) was considerably large (n= 1 × 108). Subsequently, the presence of a significantly different meditative linkage between the designated comparison pairs was assessed.

Principal Variance Component Analysis (PVCA)

To assess the variation in microbiome data based on individual and season, the principal variance component analysis (PVCA)274 was performed via R package “pvca (Version 3.15)”. The PVCA is a combination of the principal component analysis and variance components analysis https://www.niehs.nih.gov/research/resources/software/biostatistics/pvca/index.cfm, which were originally employed to assess batch effects in microarray data275 and widely used for microbiome related variance decompositions276,277. For microbiome samples in each body site, the season was determined by subtracting the date of collection from the first day of the year (from 1–365 days). Each sample’s participant ID and season were then entered into the PVCA as variables. Then, the “ggtern (Version 3.3.5)” R package was used to visualize the data.

Deconvolute the Environmental Effect on the Microbiome

Exposome and Diet Data Analysis

To investigate the influence of exposome and diet data on the microbiome from different body sites, exposome data (chemical and environmental) were collected and processed as previously described255. Diet data were collected and detailed in the methods section above. As an example, the analysis process for exposome chemical data is described below.

Microbiome samples with matching exposome chemical data within a 3-day period were selected for subsequent analysis (NChemical = 8, NEnvironmental = 32 (NParticipant1 = 13; NParticipant2 = 19)). Microbiome data were normalized using the centered log ratio (CLR, “clr” function from R package “compositions”), and exposome data were log2-transformed and auto-scaled92. Principal component analysis (PCA) was performed on both microbiome and exposome data. Principal components (PCs) from the microbiome and exposome were further analyzed, with PCs accounting for over 80% of cumulative explained variation being included. A linear regression model was constructed using PCs of microbiome data as the dependent variable (Y) and corresponding exposome PCs as the independent variable (X). The R2 value was extracted to represent the exposome’s contribution to microbiome data. The same method was applied to evaluate the dietary effect on the microbiome from four body sites.

Seasonal Effects on Microbiome

Z-score normalized microbial data or microbial diversity was systematically analyzed using Generalized Additive Mixed Models (GAMMs)278. For each genus of interest, the model was formulated as:

genera/diversity~IRIS+s(Time,bs=cc)+(1|Subject_ID),knots=list(TimeOfYear=c(0,366)

In this model, the response variable ‘genera’ represents the z-score normalized microbial relative abundance. The fixed effects components include the status of insulin sensitivity (IRIS) and a cyclic cubic spline smoother for the Time variable, encapsulating potential cyclical patterns across the year (from 0 to 366). The term (1|Subject_ID) includes a random intercept for each subject, to account for within-subject correlation.

The model was fitted using the Restricted Maximum Likelihood (REML) method for robust estimation of smoothing parameters in a complex and unbalanced design and incorporated the use of ‘lmeControl’ function from the ‘nlme’ package in R to handle the optimization process of the mixed-effects models. This was conducted by specifying ‘optim’ as the optimizer for the model fit.

The resulting model provides insight into the temporal dynamics of gene expression and its relationship with insulin sensitivity status (IRIS), considering the random effects associated with each subject. The graphical representation of these models for each genus and p value for smooth terms were saved for further exploration.

The Effects of Infection on Microbiome

The analysis of the effects of infection on the microbiome involved genera that were significantly altered during infection periods. The methodology implemented was informed by a previous method for estimating infection processes based on self-reported symptoms70.

The infection status was classified into longitudinal categories: pre-healthy (−H) state, event early (EE) state, event late (EL) state, recovery (RE) state, and post-healthy (+H) state. The pre-healthy state comprised the healthy baselines observed within 186 days preceding the onset of the infection event. The EEs state was characterized by visits occurring between day 1 and day 6 of the event. The EL state spanned visits on days 7 to 14 since the onset of the event. The recovery state included visits within the 15–40-day period since the event’s inception, and the post-healthy state encompassed visits within the 186 days following the event.

The categorization of these states was designed as a continuous progression from the pre-healthy to post-healthy state, with the average duration of an infection event being 88 days. 58 infection events were detected among 32 participants (IS: 7 participants, IR:12 participants, Unknown:13 participants) in our study. Each event was assigned a unique identification consisting of the subject ID and the event number. The progression of infection states within an event was tracked in the order of pre-healthy, event early, event late, recovery, and post-healthy.

In order to assess the effects of these infection states and insulin sensitivity status on microbiome genera, GAMM was constructed:

genera~IRIS+s(Infection_status,bs=cc,k=5)+(1|event)

In this model, ‘genera’ represents the z-score normalized microbial relative abundance, IRIS indicates the insulin sensitivity status of each participant, and ‘Infection_status’ is a smoothing function of the longitudinal infection states with cyclic cubic regression splines. The term ‘(1|event)’ is a random intercept for each infection event.

For the evenness changes during infection among insulin resistant (IR) and insulin sensitive (IS) individuals, the model was reformulated as follows:

evenness~s(Infection_status,bs=cc,k=5)+(1|event)

In this adjusted model, ‘evenness’ denotes the outcome variable, specifically the Subject ID based Z-score transformed Pielou’s evenness measure of the microbiome sample, indicating the diversity of the microbial community. The remaining components of the model and their interpretations align with the previously described model.

Supplementary Material

1
2

Supplementary Table 1. Meta-Data, Statistical Analyses, and Taxa Abundance in IS and IR Individuals, related to Figure 1

S1A. Participants related metadata, related to Figure 1A. S1B. The p-value 1284 of smooth term in GAMM model for seasonal microbial variations, related to Figure S1E. S1C. Chi-Square Test result to determine if taxa are more likely a core taxon in either IS or IR individual, related to Figure 1F. S1D. LEFSE result (comparing the relative abundance of all taxa by individual mean value), related to Figure S1M.

Supplementary Table 2. Intra- and Interindividual BC Distance, DMI Comparisons by Phylum and Genera, related to Figure 2

S2A. Wilcoxon signed-rank test of BC distance intraindividual (within), interindividual (between) and within the same household (family), related to Figure 2A. S2B. The DMI comparison between body sites by phylum, related to Figure 2C. S2C. DMI and FS of all genera, related to Figure 2D.

Supplementary Table 3. Correlations and GAMM Analysis of Microbial Genera Across Body Sites and Infections, related to Figure 3

S3A. Comparison of the proportion of inter-correlated genera both within and between various body sites, related to Figure 3D. S3B. Intra individual correlations between microbiome genera, related to Figure 3D. S3C. Inter-individual correlation between microbial genera using SPARCC, related to Figure 3D. S3D. Generalized Additive Mixed Models (GAMM) on microbial perturbations during infection, related to Figure S3F, S3G, S3H.

Supplementary Table 4. Correlated Cytokine-Microbiome Pairs and Cytokine Correlations by Body Site, related to Figure 4

S4A. The number of significantly correlated pairs of cytokine and microbiome, based on cytokines and body sites related to Figure 4A. S4B. Pairwise cytokine correlations, from the BRMS model related to Figure 4A.

Supplementary Table 5. Lipid Annotations and LMM Results on Omics-Microbiome Correlations, related to Figure 5

S5A. Lipid module and their associated lipid species annotation, related to Figure 5A, Figure S6A. S5B. The result from LMM infers the internal omics and microbiome relative abundance longitudinal correlations, related to Figure 5A. S5C. The result from LMM infers the internal omics and microbiome richness longitudinal correlations, related to Figure 5C.

Supplementary Table 6. Clinical Lab Test Annotations and Mediation Analysis Outcomes, related to Figure 6

S6A. Annotation from the clinical lab test and their phenotype class, related to Figure 6A. S6B. The mediation analysis results, related to Figure 6A. S6C. The comparison between different mediate effects between IR and IS, related to Figure 6A.

Figure 6. Causal inference decodes microbiome-driven phenotypic dynamics mediated by internal molecules and cytokines.

Figure 6.

A. Microbiome and phenotype mediation analysis. Comparisons between IS and IR regarding each mediated effect were performed using a Fisher exact t test. B. Akkermansia’s Mediation Effect on Blood A1C Level via Plasma IL-15. C. Akkermansia’s Mediation Effect on Blood A1C Level in Insulin Sensitive Individuals. D. Haemophilus’s Mediation Effect on Plasma Triglycerides Level. E. Haemophilus’s Mediation Effect on Plasma Triglycerides Level in Insulin Sensitive Individuals.

KEY RESOURCES TABLE.

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
N/A
Bacterial and virus strains
N/A
Biological samples
EDTA-plasma and PBMCs prepared from intravenous whole blood collection this manuscript N/A
Stool Sample this manuscript N/A
Skin Swaps this manuscript N/A
Oral Swaps this manuscript N/A
Nasal Swaps this manuscript N/A
Chemicals, peptides, and recombinant proteins
MO BIO’s PowerSoil DNA Isolation Kit Qiagen 47014
AllPrep DNA/RNA/Protein kit Qiagen 80004
Critical commercial assays
Insulin-Suppression Test Stanford’s Clinical and Translational Research Unit https://med.stanford.edu/ctru/services.html#laboratory-services
Luminex Multiplex Assays Stanford Human Immune Monitoring Center https://iti.stanford.edu/himc/immunoassays.html
Microbial 16S rRNA V4 sequencing uBiome Caporaso, et.al238
Clinical Lab Test Stanford’s Clinical and Translational Research Unit https://stanfordlab.com/test-directory.html
Deposited data
Raw 16S rRNA V4 sequencing data, skin and oral this manuscript https://hmpdacc.org
Raw 16S rRNA V1-3 sequencing data, stool and nasal Zhou, et.al.70 https://hmpdacc.org
Experimental models: Cell lines
N/A
Experimental models: Organisms/strains
N/A
Oligonucleotides
27F:5′-AGAGTTTGATCCTGGCTCAG-3′ this manuscript N/A
534R: 5′- ATTACCGCGGCTGCTGG-3′ this manuscript N/A
515F: 5’-GTGCCAGCMGCCGCGGTAA-3’ uBiome Caporaso, et.al238
806R: 5’-GGACTACHVGGGTWTC TAAT-’ uBiome Caporaso, et.al238
Recombinant DNA
N/A
Software and algorithms
BCL2FASTQ2 Illumina https://support.illumina.com/sequencing/sequencing_software/bcl2fastq-conversion-software.html
DADA2 Github239 https://benjjneb.github.io/dada2/
Phyloseq Bioconductor240 https://bioconductor.org/packages/release/bioc/html/phyloseq.html
SPARCC Github241 https://rdrr.io/github/siskac/discordant/src/R/SparCC.R
igraph Bioconductor https://igraph.org/
Other
Human Microbiome Project – Core Microbiome Sampling Protocol A HMP Protocol70 # 07-001, v12.0

Study Highlights:

  • The stability of the human microbiome varies among individuals and body sites.

  • Highly individualized microbial genera are more stable over time.

  • At each body site, coordinated microbial dynamics are observed in response to disease.

  • Microbiome stability and skin microbial composition are altered with insulin resistance.

ACKNOWLEDGMENT:

We extend our sincere gratitude to all research participants for their dedication and involvement. We are grateful for the valuable insights provided by the members of Snyder Lab (Stanford University) and Weinstock Lab (The Jackson Laboratory of Genomic Medicine). Our thanks also go to Mrs. Ada Yee Ki Chen and Mrs. Lisa Stainton for their research administrative support. Our appreciation extends to the Stanford Immune Monitoring Core and Ms. Yael Rosenberg-Hasson for their contribution to the cytokine Luminex Multiplex Assay. We also thank Dr. Victor Felix and Dr. Owen White (University of Maryland) for their assistance in uploading our raw sequencing data to the iHMP portal. We thank artist Lettie McGuire EdM for her help with the graphical abstract and cover design.

This work was funded by National Institutes of Health (NIH) Common Fund Human Microbiome Project U54_DE0237891, NIH U54_DK102556, NIH R01_DK110186, NIH S10_OD020141, NIH R01 AT010232-04, NIH UL1 TR001085, NIH P30_DK116074, S10_OD023452. We are also sincerely grateful for the generous support from Leona M. and Harry B. Helmsley Charitable Trust (Grant No. G-2004-03820), the Innovative Medicines Accelerator (Grant No. IMA-1051) grant and the RAMBAM-Stanford International Collaboration grant at Stanford University.

We acknowledge the fellowship support received by X. Zhou from the Stanford Aging and Ethnogeriatrics (SAGE) Research Center under NIH/NIA Resource Centers for Minority Aging Research grant P30AG059307, S.M.S.-F.R. from the NIH K08 ES028825, A.W.B. from 1F32DK126287, D.J.S. from NIA-K01AG070310, and J.S.J. from the Kennedy Trust for Rheumatology Research.

Inclusion And Diversity Statement

Multiple authors of this paper and study participants of the research identify themselves as an underrepresented ethnic minority. One or more authors of this paper identifies themselves as part of the LGBTQ+ community. Multiple authors of this paper received support from programs that are designed to increase minority representation in science.

Footnotes

DECLARATION OF INTEREST

M.P.S. is a co-founder and the scientific advisory board member of Personalis, Qbio, January, SensOmics, Filtricine, Akna, Protos, Mirvie, NiMo, Onza, Oralome, Marble Therapeutics and Iollo. He is also on the scientific advisory board of Danaher, Genapsys, and Jupiter. A.H. is a founder and shareholder of Arxeon. Y.Z. and G.M.W. are co-founders of General Biomics. No other potential conflicts of interest relevant to this article were reported.

Declaration of Generative AI and AI-assisted Technologies in the Writing Process

During the preparation of this work the author(s) used ChatGPT in order to improve the quality of scientific writing. After using this tool/service, the author(s) reviewed and edited the content as needed and take(s) full responsibility for the content of the publication.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCE

  • 1.Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. (2010). A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464, 59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gilbert JA, Blaser MJ, Caporaso JG, Jansson JK, Lynch SV, and Knight R (2018). Current understanding of the human microbiome. Nat Med 24, 392–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ursell LK, Metcalf JL, Parfrey LW, and Knight R (2012). Defining the human microbiome. Nutrition reviews 70 Suppl 1, S38–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Oh J, Byrd AL, Park M, Program NCS, Kong HH, and Segre JA (2016). Temporal Stability of the Human Skin Microbiome. Cell 165, 854–866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Roswall J, Olsson LM, Kovatcheva-Datchary P, Nilsson S, Tremaroli V, Simon MC, Kiilerich P, Akrami R, Kramer M, Uhlen M, et al. (2021). Developmental trajectory of the healthy human gut microbiota during the first 5 years of life. Cell host & microbe 29, 765–776 e763. [DOI] [PubMed] [Google Scholar]
  • 6.Zhou Y, Gao H, Mihindukulasuriya KA, La Rosa PS, Wylie KM, Vishnivetskaya T, Podar M, Warner B, Tarr PI, Nelson DE, et al. (2013). Biogeography of the ecosystems of the healthy human body. Genome biology 14, R1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Human Microbiome Project C (2012). Structure, function and diversity of the healthy human microbiome. Nature 486, 207–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zhou Y, Mihindukulasuriya KA, Gao H, La Rosa PS, Wylie KM, Martin JC, Kota K, Shannon WD, Mitreva M, Sodergren E, and Weinstock GM (2014). Exploration of bacterial community classes in major human habitats. Genome biology 15, R66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.DiGiulio DB, Callahan BJ, McMurdie PJ, Costello EK, Lyell DJ, Robaczewska A, Sun CL, Goltsman DS, Wong RJ, Shaw G, et al. (2015). Temporal and spatial variation of the human microbiota during pregnancy. Proceedings of the National Academy of Sciences of the United States of America 112, 11060–11065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, Mujagic Z, Vila AV, Falony G, Vieira-Silva S, et al. (2016). Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 352, 565–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wilmanski T, Diener C, Rappaport N, Patwardhan S, Wiedrick J, Lapidus J, Earls JC, Zimmer A, Glusman G, Robinson M, et al. (2021). Gut microbiome pattern reflects healthy ageing and predicts survival in humans. Nat Metab 3, 274–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bana B, and Cabreiro F (2019). The Microbiome and Aging. Annu Rev Genet 53, 239–261. [DOI] [PubMed] [Google Scholar]
  • 13.Zhou X, WANG B, Demkowicz PC, Johnson JS, Chen Y, Spakowicz D, Zhou Y, Dorsett Y, Chen L, and Sodergren E Exploratory studies of oral and fecal microbiome in healthy human aging. Frontiers in Aging, 110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zhang Y, Bhosle A, Bae S, McIver LJ, Pishchany G, Accorsi EK, Thompson KN, Arze C, Wang Y, Subramanian A, et al. (2022). Discovery of bioactive microbial gene products in inflammatory bowel disease. Nature 606, 754–760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fornelos N, Franzosa EA, Bishai J, Annand JW, Oka A, Lloyd-Price J, Arthur TD, Garner A, Avila-Pacheco J, Haiser HJ, et al. (2020). Growth effects of N-acylethanolamines on gut bacteria reflect altered bacterial abundances in inflammatory bowel disease. Nat Microbiol 5, 486–497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Nemet I, Saha PP, Gupta N, Zhu W, Romano KA, Skye SM, Cajka T, Mohan ML, Li L, Wu Y, et al. (2020). A Cardiovascular Disease-Linked Gut Microbial Metabolite Acts via Adrenergic Receptors. Cell 180, 862–877 e822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Walker RL, Vlamakis H, Lee JWJ, Besse LA, Xanthakis V, Vasan RS, Shaw SY, and Xavier RJ (2021). Population study of the gut microbiome: associations with diet, lifestyle, and cardiometabolic disease. Genome Med 13, 188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Fromentin S, Forslund SK, Chechi K, Aron-Wisnewsky J, Chakaroun R, Nielsen T, Tremaroli V, Ji B, Prifti E, Myridakis A, et al. (2022). Microbiome and metabolome features of the cardiometabolic disease spectrum. Nat Med 28, 303–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Qi Q, Li J, Yu B, Moon JY, Chai JC, Merino J, Hu J, Ruiz-Canela M, Rebholz C, Wang Z, et al. (2022). Host and gut microbial tryptophan metabolism and type 2 diabetes: an integrative analysis of host genetics, diet, gut microbiome and circulating metabolites in cohort studies. Gut 71, 1095–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wu H, Tremaroli V, Schmidt C, Lundqvist A, Olsson LM, Kramer M, Gummesson A, Perkins R, Bergstrom G, and Backhed F (2020). The Gut Microbiota in Prediabetes and Diabetes: A Population-Based Cross-Sectional Study. Cell metabolism 32, 379–390 e373. [DOI] [PubMed] [Google Scholar]
  • 22.Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, Prifti E, Vieira-Silva S, Gudmundsdottir V, Pedersen HK, et al. (2017). Corrigendum: Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 545, 116. [DOI] [PubMed] [Google Scholar]
  • 23.Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, Jensen BA, Forslund K, Hildebrand F, Prifti E, Falony G, et al. (2016). Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 535, 376–381. [DOI] [PubMed] [Google Scholar]
  • 24.Ruuskanen MO, Erawijantari PP, Havulinna AS, Liu Y, Meric G, Tuomilehto J, Inouye M, Jousilahti P, Salomaa V, Jain M, et al. (2022). Gut Microbiome Composition Is Predictive of Incident Type 2 Diabetes in a Population Cohort of 5,572 Finnish Adults. Diabetes Care 45, 811–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chen Z, Radjabzadeh D, Chen L, Kurilshikov A, Kavousi M, Ahmadizar F, Ikram MA, Uitterlinden AG, Zhernakova A, Fu J, et al. (2021). Association of Insulin Resistance and Type 2 Diabetes With Gut Microbial Diversity: A Microbiome-Wide Analysis From Population Studies. JAMA Netw Open 4, e2118811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Deng K, Shuai M, Zhang Z, Jiang Z, Fu Y, Shen L, Zheng JS, and Chen YM (2022). Temporal relationship among adiposity, gut microbiota, and insulin resistance in a longitudinal human cohort. BMC Med 20, 171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Khan MT, Nieuwdorp M, and Backhed F (2014). Microbial modulation of insulin sensitivity. Cell metabolism 20, 753–760. [DOI] [PubMed] [Google Scholar]
  • 28.Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, Prifti E, Vieira-Silva S, Gudmundsdottir V, Pedersen HK, et al. (2015). Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 528, 262–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Al-Muhanna FA, Dowdell AK, Al Eleq AH, Albaker WI, Brooks AW, Al-Sultan AI, Al-Rubaish AM, Alkharsah KR, Sulaiman RM, Al-Quorain AA, et al. (2022). Gut microbiota analyses of Saudi populations for type 2 diabetes-related phenotypes reveals significant association. BMC microbiology 22, 301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gardiner M, Vicaretti M, Sparks J, Bansal S, Bush S, Liu M, Darling A, Harry E, and Burke CM (2017). A longitudinal study of the diabetic skin and wound microbiome. PeerJ 5, e3543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Redel H, Gao Z, Li H, Alekseyenko AV, Zhou Y, Perez-Perez GI, Weinstock G, Sodergren E, and Blaser MJ (2013). Quantitation and composition of cutaneous microbiota in diabetic and nondiabetic men. The Journal of infectious diseases 207, 1105–1114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tai J, Han MS, Kwak J, and Kim TH (2021). Association Between Microbiota and Nasal Mucosal Diseases in terms of Immunity. Int J Mol Sci 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.De Pessemier B, Grine L, Debaere M, Maes A, Paetzold B, and Callewaert C (2021). Gut-Skin Axis: Current Knowledge of the Interrelationship between Microbial Dysbiosis and Skin Conditions. Microorganisms 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Besnard P, Christensen JE, Bernard A, Simoneau-Robin I, Collet X, Verges B, and Burcelin R (2020). Identification of an oral microbiota signature associated with an impaired orosensory perception of lipids in insulin-resistant patients. Acta Diabetol 57, 1445–1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Demmer RT, Trinh P, Rosenbaum M, Li G, LeDuc C, Leibel R, Gonzalez A, Knight R, Paster B, Colombo PC, et al. (2019). Subgingival Microbiota and Longitudinal Glucose Change: The Oral Infections, Glucose Intolerance and Insulin Resistance Study (ORIGINS). J Dent Res 98, 1488–1496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Xiao E, Mattos M, Vieira GHA, Chen S, Correa JD, Wu Y, Albiero ML, Bittinger K, and Graves DT (2017). Diabetes Enhances IL-17 Expression and Alters the Oral Microbiome to Increase Its Pathogenicity. Cell host & microbe 22, 120–128 e124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Almeida-Santos A, Martins-Mendes D, Gaya-Vidal M, Perez-Pardal L, and Beja-Pereira A (2021). Characterization of the Oral Microbiome of Medicated Type-2 Diabetes Patients. Frontiers in microbiology 12, 610370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Negrini TC, Carlos IZ, Duque C, Caiaffa KS, and Arthur RA (2021). Interplay Among the Oral Microbiome, Oral Cavity Conditions, the Host Immune Response, Diabetes Mellitus, and Its Associated-Risk Factors-An Overview. Front Oral Health 2, 697428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Demmer RT, Jacobs DR Jr., Singh R, Zuk A, Rosenbaum M, Papapanou PN, and Desvarieux M (2015). Periodontal Bacteria and Prediabetes Prevalence in ORIGINS: The Oral Infections, Glucose Intolerance, and Insulin Resistance Study. J Dent Res 94, 201S–211S. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Watanabe K, Katagiri S, Takahashi H, Sasaki N, Maekawa S, Komazaki R, Hatasa M, Kitajima Y, Maruyama Y, Shiba T, et al. (2021). Porphyromonas gingivalis impairs glucose uptake in skeletal muscle associated with altering gut microbiota. FASEB J 35, e21171. [DOI] [PubMed] [Google Scholar]
  • 41.Vrieze A, Van Nood E, Holleman F, Salojarvi J, Kootte RS, Bartelsman JF, Dallinga-Thie GM, Ackermans MT, Serlie MJ, Oozeer R, et al. (2012). Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 143, 913–916 e917. [DOI] [PubMed] [Google Scholar]
  • 42.Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, Casella G, Drew JC, Ilonen J, Knip M, Hyoty H, et al. (2011). Toward defining the autoimmune microbiome for type 1 diabetes. The ISME journal 5, 82–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Frost F, Kacprowski T, Ruhlemann M, Pietzner M, Bang C, Franke A, Nauck M, Volker U, Volzke H, Dorr M, et al. (2021). Long-term instability of the intestinal microbiome is associated with metabolic liver disease, low microbiota diversity, diabetes mellitus and impaired exocrine pancreatic function. Gut 70, 522–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kawano Y, Edwards M, Huang Y, Bilate AM, Araujo LP, Tanoue T, Atarashi K, Ladinsky MS, Reiner SL, Wang HH, et al. (2022). Microbiota imbalance induced by dietary sugar disrupts immune-mediated protection from metabolic syndrome. Cell 185, 3501–3519 e3520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Massier L, Chakaroun R, Tabei S, Crane A, Didt KD, Fallmann J, von Bergen M, Haange SB, Heyne H, Stumvoll M, et al. (2020). Adipose tissue derived bacteria are associated with inflammation in obesity and type 2 diabetes. Gut 69, 1796–1806. [DOI] [PubMed] [Google Scholar]
  • 46.Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermudez-Humaran LG, Smirnova N, Berge M, Sulpice T, Lahtinen S, et al. (2011). Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Mol Med 3, 559–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Linh HT, Iwata Y, Senda Y, Sakai-Takemori Y, Nakade Y, Oshima M, Nakagawa-Yoneda S, Ogura H, Sato K, Minami T, et al. (2022). Intestinal Bacterial Translocation Contributes to Diabetic Kidney Disease. J Am Soc Nephrol 33, 1105–1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.de Groot P, Scheithauer T, Bakker GJ, Prodan A, Levin E, Khan MT, Herrema H, Ackermans M, Serlie MJM, de Brauw M, et al. (2020). Donor metabolic characteristics drive effects of faecal microbiota transplantation on recipient insulin sensitivity, energy expenditure and intestinal transit time. Gut 69, 502–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Balaich J, Estrella M, Wu G, Jeffrey PD, Biswas A, Zhao L, Korennykh A, and Donia MS (2021). The human microbiome encodes resistance to the antidiabetic drug acarbose. Nature 600, 110–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Gacesa R, Kurilshikov A, Vich Vila A, Sinha T, Klaassen MAY, Bolte LA, Andreu-Sanchez S, Chen L, Collij V, Hu S, et al. (2022). Environmental factors shaping the gut microbiome in a Dutch population. Nature 604, 732–739. [DOI] [PubMed] [Google Scholar]
  • 51.Asnicar F, Berry SE, Valdes AM, Nguyen LH, Piccinno G, Drew DA, Leeming E, Gibson R, Le Roy C, Khatib HA, et al. (2021). Microbiome connections with host metabolism and habitual diet from 1,098 deeply phenotyped individuals. Nat Med 27, 321–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Fassarella M, Blaak EE, Penders J, Nauta A, Smidt H, and Zoetendal EG (2021). Gut microbiome stability and resilience: elucidating the response to perturbations in order to modulate gut health. Gut 70, 595–605. [DOI] [PubMed] [Google Scholar]
  • 53.Price ND, Magis AT, Earls JC, Glusman G, Levy R, Lausted C, McDonald DT, Kusebauch U, Moss CL, Zhou Y, et al. (2017). A wellness study of 108 individuals using personal, dense, dynamic data clouds. Nat Biotechnol 35, 747–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tebani A, Gummesson A, Zhong W, Koistinen IS, Lakshmikanth T, Olsson LM, Boulund F, Neiman M, Stenlund H, Hellstrom C, et al. (2020). Integration of molecular profiles in a longitudinal wellness profiling cohort. Nat Commun 11, 4487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Olsson LM, Boulund F, Nilsson S, Khan MT, Gummesson A, Fagerberg L, Engstrand L, Perkins R, Uhlen M, Bergstrom G, et al. (2022). Dynamics of the normal gut microbiota: A longitudinal one-year population study in Sweden. Cell host & microbe 30, 726–739 e723. [DOI] [PubMed] [Google Scholar]
  • 56.Chen L, Wang D, Garmaeva S, Kurilshikov A, Vich Vila A, Gacesa R, Sinha T, Lifelines Cohort S, Segal E, Weersma RK, et al. (2021). The long-term genetic stability and individual specificity of the human gut microbiome. Cell 184, 2302–2315 e2312. [DOI] [PubMed] [Google Scholar]
  • 57.Li J, Li Y, Ivey KL, Wang DD, Wilkinson JE, Franke A, Lee KH, Chan A, Huttenhower C, Hu FB, et al. (2022). Interplay between diet and gut microbiome, and circulating concentrations of trimethylamine N-oxide: findings from a longitudinal cohort of US men. Gut 71, 724–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zhou W, Spoto M, Hardy R, Guan C, Fleming E, Larson PJ, Brown JS, and Oh J (2020). Host-Specific Evolutionary and Transmission Dynamics Shape the Functional Diversification of Staphylococcus epidermidis in Human Skin. Cell 180, 454–470 e418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, and Knight R (2009). Bacterial community variation in human body habitats across space and time. Science 326, 1694–1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Larson PJ, Chong D, Fleming E, and Oh J (2021). Challenges in Developing a Human Model System for Skin Microbiome Research. J Invest Dermatol 141, 228–231 e224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Selway CA, Sudarpa J, and Weyrich LS (2022). Moving beyond the gut microbiome: Combining systems biology and multi-site microbiome analyses to combat non-communicable diseases. Medicine in Microecology 12, 100052. [Google Scholar]
  • 62.Oh J, Byrd AL, Deming C, Conlan S, Program NCS, Kong HH, and Segre JA (2014). Biogeography and individuality shape function in the human skin metagenome. Nature 514, 59–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Li Z, Xia J, Jiang L, Tan Y, An Y, Zhu X, Ruan J, Chen Z, Zhen H, Ma Y, et al. (2021). Characterization of the human skin resistome and identification of two microbiota cutotypes. Microbiome 9, 47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.David LA, Materna AC, Friedman J, Campos-Baptista MI, Blackburn MC, Perrotta A, Erdman SE, and Alm EJ (2014). Host lifestyle affects human microbiota on daily timescales. Genome biology 15, R89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Oh J, Freeman AF, Program NCS, Park M, Sokolic R, Candotti F, Holland SM, Segre JA, and Kong HH (2013). The altered landscape of the human skin microbiome in patients with primary immunodeficiencies. Genome research 23, 2103–2114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Caporaso JG, Lauber CL, Costello EK, Berg-Lyons D, Gonzalez A, Stombaugh J, Knights D, Gajer P, Ravel J, Fierer N, et al. (2011). Moving pictures of the human microbiome. Genome biology 12, R50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Olofsson LE, and Backhed F (2022). The Metabolic Role and Therapeutic Potential of the Microbiome. Endocr Rev 43, 907–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wozniak JM, Mills RH, Olson J, Caldera JR, Sepich-Poore GD, Carrillo-Terrazas M, Tsai CM, Vargas F, Knight R, Dorrestein PC, et al. (2020). Mortality Risk Profiling of Staphylococcus aureus Bacteremia by Multi-omic Serum Analysis Reveals Early Predictive and Pathogenic Signatures. Cell 182, 1311–1327 e1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chen R, Mias GI, Li-Pook-Than J, Jiang L, Lam HY, Chen R, Miriami E, Karczewski KJ, Hariharan M, Dewey FE, et al. (2012). Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell 148, 1293–1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zhou W, Sailani MR, Contrepois K, Zhou Y, Ahadi S, Leopold SR, Zhang MJ, Rao V, Avina M, Mishra T, et al. (2019). Longitudinal multi-omics of host-microbe dynamics in prediabetes. Nature 569, 663–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Integrative, H.M.P.R.N.C. (2019). The Integrative Human Microbiome Project. Nature 569, 641–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Callahan BJ, McMurdie PJ, and Holmes SP (2017). Exact sequence variants should replace operational taxonomic units in marker-gene data analysis. The ISME journal 11, 2639–2643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Johnson JS, Spakowicz DJ, Hong BY, Petersen LM, Demkowicz P, Chen L, Leopold SR, Hanson BM, Agresta HO, Gerstein M, et al. (2019). Evaluation of 16S rRNA gene sequencing for species and strain-level microbiome analysis. Nat Commun 10, 5029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Schloss PD (2021). Amplicon Sequence Variants Artificially Split Bacterial Genomes into Separate Clusters. mSphere 6, e0019121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Muniyappa R, Lee S, Chen H, and Quon MJ (2008). Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usage. Am J Physiol Endocrinol Metab 294, E15–26. [DOI] [PubMed] [Google Scholar]
  • 76.Schussler-Fiorenza Rose SM, Contrepois K, Moneghetti KJ, Zhou W, Mishra T, Mataraso S, Dagan-Rosenfeld O, Ganz AB, Dunn J, Hornburg D, et al. (2019). A longitudinal big data approach for precision health. Nat Med 25, 792–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto JM, et al. (2011). Enterotypes of the human gut microbiome. Nature 473, 174–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Costea PI, Hildebrand F, Arumugam M, Backhed F, Blaser MJ, Bushman FD, de Vos WM, Ehrlich SD, Fraser CM, Hattori M, et al. (2018). Enterotypes in the landscape of gut microbial community composition. Nat Microbiol 3, 8–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Oh J, Conlan S, Polley EC, Segre JA, and Kong HH (2012). Shifts in human skin and nares microbiota of healthy children and adults. Genome Med 4, 77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Garcia-Lopez M, Meier-Kolthoff JP, Tindall BJ, Gronow S, Woyke T, Kyrpides NC, Hahnke RL, and Goker M (2019). Analysis of 1,000 Type-Strain Genomes Improves Taxonomic Classification of Bacteroidetes. Frontiers in microbiology 10, 2083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kilian M, Chapple IL, Hannig M, Marsh PD, Meuric V, Pedersen AM, Tonetti MS, Wade WG, and Zaura E (2016). The oral microbiome - an update for oral healthcare professionals. Br Dent J 221, 657–666. [DOI] [PubMed] [Google Scholar]
  • 82.Aas JA, Paster BJ, Stokes LN, Olsen I, and Dewhirst FE (2005). Defining the normal bacterial flora of the oral cavity. J Clin Microbiol 43, 5721–5732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Mark Welch JL, Rossetti BJ, Rieken CW, Dewhirst FE, and Borisy GG (2016). Biogeography of a human oral microbiome at the micron scale. Proceedings of the National Academy of Sciences of the United States of America 113, E791–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lahti L, Salojarvi J, Salonen A, Scheffer M, and de Vos WM (2014). Tipping elements in the human intestinal ecosystem. Nat Commun 5, 4344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sailani MR, Metwally AA, Zhou W, Rose SMS, Ahadi S, Contrepois K, Mishra T, Zhang MJ, Kidzinski L, Chu TJ, and Snyder MP (2020). Deep longitudinal multiomics profiling reveals two biological seasonal patterns in California. Nat Commun 11, 4933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Smits SA, Leach J, Sonnenburg ED, Gonzalez CG, Lichtman JS, Reid G, Knight R, Manjurano A, Changalucha J, Elias JE, et al. (2017). Seasonal cycling in the gut microbiome of the Hadza hunter-gatherers of Tanzania. Science 357, 802–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Koliada A, Moseiko V, Romanenko M, Piven L, Lushchak O, Kryzhanovska N, Guryanov V, and Vaiserman A (2020). Seasonal variation in gut microbiota composition: cross-sectional evidence from Ukrainian population. BMC microbiology 20, 100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wastyk HC, Fragiadakis GK, Perelman D, Dahan D, Merrill BD, Yu FB, Topf M, Gonzalez CG, Van Treuren W, Han S, et al. (2021). Gut-microbiota-targeted diets modulate human immune status. Cell 184, 4137–4153 e4114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lancaster SM, Lee-McMullen B, Abbott CW, Quijada JV, Hornburg D, Park H, Perelman D, Peterson DJ, Tang M, Robinson A, et al. (2022). Global, distinctive, and personal changes in molecular and microbial profiles by specific fibers in humans. Cell host & microbe 30, 848–862 e847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Jiang C, Wang X, Li X, Inlora J, Wang T, Liu Q, and Snyder M (2018). Dynamic Human Environmental Exposome Revealed by Longitudinal Personal Monitoring. Cell 175, 277–291 e231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Fouladi F, Bailey MJ, Patterson WB, Sioda M, Blakley IC, Fodor AA, Jones RB, Chen Z, Kim JS, Lurmann F, et al. (2020). Air pollution exposure is associated with the gut microbiome as revealed by shotgun metagenomic sequencing. Environ Int 138, 105604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Gao P, Shen X, Zhang X, Jiang C, Zhang S, Zhou X, Schussler-Fiorenza Rose SM, and Snyder M (2022). Precision environmental health monitoring by longitudinal exposome and multi-omics profiling. Genome research 32, 1199–1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sunagawa S, Schloissnig S, Arumugam M, Forslund K, Mitreva M, Tap J, Zhu A, Waller A, Mende DR, Kultima JR, et al. (2013). Individuality and temporal stability of the human gut microbiome. Cent Asian J Glob Health 2, 120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Mehta RS, Abu-Ali GS, Drew DA, Lloyd-Price J, Subramanian A, Lochhead P, Joshi AD, Ivey KL, Khalili H, Brown GT, et al. (2018). Stability of the human faecal microbiome in a cohort of adult men. Nat Microbiol 3, 347–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC, Program NCS, Bouffard GG, Blakesley RW, Murray PR, et al. (2009). Topographical and temporal diversity of the human skin microbiome. Science 324, 1190–1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gajer P, Brotman RM, Bai G, Sakamoto J, Schutte UM, Zhong X, Koenig SS, Fu L, Ma ZS, Zhou X, et al. (2012). Temporal dynamics of the human vaginal microbiota. Sci Transl Med 4, 132ra152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Lemanceau P, Blouin M, Muller D, and Moenne-Loccoz Y (2017). Let the Core Microbiota Be Functional. Trends Plant Sci 22, 583–595. [DOI] [PubMed] [Google Scholar]
  • 98.Priya S, and Blekhman R (2019). Population dynamics of the human gut microbiome: change is the only constant. Genome biology 20, 150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Scapaticci M, Marchetto S, Nardi A, Zoppelletto M, and Bartolini A (2018). A case of necrotizing fasciitis caused by Finegoldia magna in a patient with type 2 diabetes mellitus. Infez Med 26, 359–363. [PubMed] [Google Scholar]
  • 100.Jneid J, Cassir N, Schuldiner S, Jourdan N, Sotto A, Lavigne JP, and La Scola B (2018). Exploring the Microbiota of Diabetic Foot Infections With Culturomics. Front Cell Infect Microbiol 8, 282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Arencibia-Perez B, Benet-Munoz O, Roque-Castellano C, and Marchena-Gomez J (2020). Gluteal Abscess due to Finegoldia Magna in a Patient with Diabetes. Actas Dermosifiliogr (Engl Ed) 111, 527–528. [DOI] [PubMed] [Google Scholar]
  • 102.Castellanos N, Nakanouchi J, Yuzen DI, Fung S, Fernandez JS, Barberis C, Tuchscherr L, and Ramirez MS (2019). A Study on Acinetobacter baumannii and Staphylococcus aureus Strains Recovered from the Same Infection Site of a Diabetic Patient. Curr Microbiol 76, 842–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Perera D, Kleinstein SE, Hanson B, Hasturk H, Eveloff R, Freire M, and Ramsey M (2021). Impaired host response and the presence of Acinetobacter baumannii in the serum microbiome of type-II diabetic patients. iScience 24, 101941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Leung CH, and Liu CP (2019). Diabetic status and the relationship of blood glucose to mortality in adults with carbapenem-resistant Acinetobacter baumannii complex bacteremia. J Microbiol Immunol Infect 52, 654–662. [DOI] [PubMed] [Google Scholar]
  • 105.Henig O, Pogue JM, Martin E, Hayat U, Ja’ara M, Kilgore PE, Cha R, Dhar S, and Kaye KS (2020). The Impact of Multidrug-Resistant Organisms on Outcomes in Patients With Diabetic Foot Infections. Open Forum Infect Dis 7, ofaa161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Min KR, Galvis A, Baquerizo Nole KL, Sinha R, Clarke J, Kirsner RS, and Ajdic D (2020). Association between baseline abundance of Peptoniphilus, a Gram-positive anaerobic coccus, and wound healing outcomes of DFUs. PloS one 15, e0227006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Smith K, Collier A, Townsend EM, O’Donnell LE, Bal AM, Butcher J, Mackay WG, Ramage G, and Williams C (2016). One step closer to understanding the role of bacteria in diabetic foot ulcers: characterising the microbiome of ulcers. BMC microbiology 16, 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Brown K, Church D, Lynch T, and Gregson D (2014). Bloodstream infections due to Peptoniphilus spp.: report of 15 cases. Clin Microbiol Infect 20, O857–860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.i, M.C.E.a.s.b.u.e., and i, M.C. (2022). Gut microbiome of multiple sclerosis patients and paired household healthy controls reveal associations with disease risk and course. Cell 185, 3467–3486 e3416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Zhao S, Lieberman TD, Poyet M, Kauffman KM, Gibbons SM, Groussin M, Xavier RJ, and Alm EJ (2019). Adaptive Evolution within Gut Microbiomes of Healthy People. Cell host & microbe 25, 656–667 e658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Carrow HC, Batachari LE, and Chu H (2020). Strain diversity in the microbiome: Lessons from Bacteroides fragilis. PLoS pathogens 16, e1009056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Lee SM, Donaldson GP, Mikulski Z, Boyajian S, Ley K, and Mazmanian SK (2013). Bacterial colonization factors control specificity and stability of the gut microbiota. Nature 501, 426–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem T, Zeevi D, Costea PI, Godneva A, Kalka IN, Bar N, et al. (2018). Environment dominates over host genetics in shaping human gut microbiota. Nature 555, 210–215. [DOI] [PubMed] [Google Scholar]
  • 114.Kort R, Caspers M, van de Graaf A, van Egmond W, Keijser B, and Roeselers G (2014). Shaping the oral microbiota through intimate kissing. Microbiome 2, 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Rocha J, Henriques I, Gomila M, and Manaia CM (2022). Common and distinctive genomic features of Klebsiella pneumoniae thriving in the natural environment or in clinical settings. Scientific reports 12, 10441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Musher DM (1996). Haemophilus Species. In Medical Microbiology, th, and Baron S, eds. [PubMed] [Google Scholar]
  • 117.Vieira-Silva S, Falony G, Belda E, Nielsen T, Aron-Wisnewsky J, Chakaroun R, Forslund SK, Assmann K, Valles-Colomer M, Nguyen TTD, et al. (2020). Statin therapy is associated with lower prevalence of gut microbiota dysbiosis. Nature 581, 310–315. [DOI] [PubMed] [Google Scholar]
  • 118.Byrd AL, Liu M, Fujimura KE, Lyalina S, Nagarkar DR, Charbit B, Bergstedt J, Patin E, Harrison OJ, Quintana-Murci L, et al. (2021). Gut microbiome stability and dynamics in healthy donors and patients with non-gastrointestinal cancers. J Exp Med 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Byrd AL, Belkaid Y, and Segre JA (2018). The human skin microbiome. Nat Rev Microbiol 16, 143–155. [DOI] [PubMed] [Google Scholar]
  • 120.Flores GE, Caporaso JG, Henley JB, Rideout JR, Domogala D, Chase J, Leff JW, Vazquez-Baeza Y, Gonzalez A, Knight R, et al. (2014). Temporal variability is a personalized feature of the human microbiome. Genome biology 15, 531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Ontiveros VJ, Capitan JA, Casamayor EO, and Alonso D (2021). The characteristic time of ecological communities. Ecology 102, e03247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Real R, and Vargas JM (1996). The Probabilistic Basis of Jaccard’s Index of Similarity. Systematic Biology 45, 380–385. [Google Scholar]
  • 123.Li SS, Zhu A, Benes V, Costea PI, Hercog R, Hildebrand F, Huerta-Cepas J, Nieuwdorp M, Salojarvi J, Voigt AY, et al. (2016). Durable coexistence of donor and recipient strains after fecal microbiota transplantation. Science 352, 586–589. [DOI] [PubMed] [Google Scholar]
  • 124.Russell BJ, Brown SD, Siguenza N, Mai I, Saran AR, Lingaraju A, Maissy ES, Dantas Machado AC, Pinto AFM, Sanchez C, et al. (2022). Intestinal transgene delivery with native E. coli chassis allows persistent physiological changes. Cell 185, 3263–3277 e3215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Bartley JM, Zhou X, Kuchel GA, Weinstock GM, and Haynes L (2017). Impact of Age, Caloric Restriction, and Influenza Infection on Mouse Gut Microbiome: An Exploratory Study of the Role of Age-Related Microbiome Changes on Influenza Responses. Front Immunol 8, 1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Czerkinsky C, Prince SJ, Michalek SM, Jackson S, Russell MW, Moldoveanu Z, McGhee JR, and Mestecky J (1987). IgA antibody-producing cells in peripheral blood after antigen ingestion: evidence for a common mucosal immune system in humans. Proceedings of the National Academy of Sciences of the United States of America 84, 2449–2453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Winer DA, Luck H, Tsai S, and Winer S (2016). The Intestinal Immune System in Obesity and Insulin Resistance. Cell metabolism 23, 413–426. [DOI] [PubMed] [Google Scholar]
  • 128.Wang J, Li F, Wei H, Lian ZX, Sun R, and Tian Z (2014). Respiratory influenza virus infection induces intestinal immune injury via microbiota-mediated Th17 cell-dependent inflammation. J Exp Med 211, 2397–2410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Oh J, and Unutmaz D (2019). Immune cells for microbiota surveillance. Science 366, 419–420. [DOI] [PubMed] [Google Scholar]
  • 130.Zhao L, Zhang F, Ding X, Wu G, Lam YY, Wang X, Fu H, Xue X, Lu C, Ma J, et al. (2018). Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 359, 1151–1156. [DOI] [PubMed] [Google Scholar]
  • 131.Wu G, Zhao N, Zhang C, Lam YY, and Zhao L (2021). Guild-based analysis for understanding gut microbiome in human health and diseases. Genome Med 13, 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Guo M, Wu G, Tan Y, Li Y, Jin X, Qi W, Guo X, Zhang C, Zhu Z, and Zhao L (2023). Guild-Level Microbiome Signature Associated with COVID-19 Severity and Prognosis. mBio 14, e0351922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chen X, Li P, Liu M, Zheng H, He Y, Chen MX, Tang W, Yue X, Huang Y, Zhuang L, et al. (2020). Gut dysbiosis induces the development of pre-eclampsia through bacterial translocation. Gut 69, 513–522. [DOI] [PubMed] [Google Scholar]
  • 134.Marchetti G, Tincati C, and Silvestri G (2013). Microbial translocation in the pathogenesis of HIV infection and AIDS. Clinical microbiology reviews 26, 2–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Qin N, Yang F, Li A, Prifti E, Chen Y, Shao L, Guo J, Le Chatelier E, Yao J, Wu L, et al. (2014). Alterations of the human gut microbiome in liver cirrhosis. Nature 513, 59–64. [DOI] [PubMed] [Google Scholar]
  • 136.Schloissnig S, Arumugam M, Sunagawa S, Mitreva M, Tap J, Zhu A, Waller A, Mende DR, Kultima JR, Martin J, et al. (2013). Genomic variation landscape of the human gut microbiome. Nature 493, 45–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Lynn DJ, Benson SC, Lynn MA, and Pulendran B (2022). Modulation of immune responses to vaccination by the microbiota: implications and potential mechanisms. Nat Rev Immunol 22, 33–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Muller M, and Betlejewski S (2003). [Nasal mucosa in patients with diabetes mellitus]. Otolaryngol Pol 57, 361–364. [PubMed] [Google Scholar]
  • 139.Lee TK, Jeon YJ, and Jung SJ (2021). Bi-directional association between allergic rhinitis and diabetes mellitus from the national representative data of South Korea. Scientific reports 11, 4344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Nam JS, Roh YH, Kim J, Chang SW, Ha JG, Park JJ, Fahad WA, Yoon JH, Kim CH, and Cho HJ (2022). Association between diabetes mellitus and chronic rhinosinusitis with nasal polyps: A population-based cross-sectional study. Clin Otolaryngol 47, 167–173. [DOI] [PubMed] [Google Scholar]
  • 141.Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, Ter Horst R, Jansen T, Jacobs L, Bonder MJ, et al. (2016). Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity. Cell 167, 1125–1136 e1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Geva-Zatorsky N, Sefik E, Kua L, Pasman L, Tan TG, Ortiz-Lopez A, Yanortsang TB, Yang L, Jupp R, Mathis D, et al. (2017). Mining the Human Gut Microbiota for Immunomodulatory Organisms. Cell 168, 928–943 e911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Mendes V, Galvao I, and Vieira AT (2019). Mechanisms by Which the Gut Microbiota Influences Cytokine Production and Modulates Host Inflammatory Responses. J Interferon Cytokine Res 39, 393–409. [DOI] [PubMed] [Google Scholar]
  • 144.Bolte LA, Vich Vila A, Imhann F, Collij V, Gacesa R, Peters V, Wijmenga C, Kurilshikov A, Campmans-Kuijpers MJE, Fu J, et al. (2021). Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut 70, 1287–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Zhou X, Johnson JS, Spakowicz D, Zhou W, Zhou Y, Sodergren E, Snyder M, and Weinstock GM (2020). Longitudinal Analysis of Serum Cytokine Levels and Gut Microbial Abundance Links IL-17/IL-22 With Clostridia and Insulin Sensitivity in Humans. Diabetes 69, 1833–1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Lloyd-Price J, Mahurkar A, Rahnavard G, Crabtree J, Orvis J, Hall AB, Brady A, Creasy HH, McCracken C, Giglio MG, et al. (2017). Strains, functions and dynamics in the expanded Human Microbiome Project. Nature 550, 61–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, et al. (2009). A core gut microbiome in obese and lean twins. Nature 457, 480–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Somineni HK, Weitzner JH, Venkateswaran S, Dodd A, Prince J, Karikaran A, Sauer CG, Abramowicz S, Zwick ME, Cutler DJ, et al. (2021). Site- and Taxa-Specific Disease-Associated Oral Microbial Structures Distinguish Inflammatory Bowel Diseases. Inflamm Bowel Dis 27, 1889–1900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Gao Z, Tseng CH, Strober BE, Pei Z, and Blaser MJ (2008). Substantial alterations of the cutaneous bacterial biota in psoriatic lesions. PloS one 3, e2719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Shin NR, Whon TW, and Bae JW (2015). Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol 33, 496–503. [DOI] [PubMed] [Google Scholar]
  • 151.Bradley PH, and Pollard KS (2017). Proteobacteria explain significant functional variability in the human gut microbiome. Microbiome 5, 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.d’Hennezel E, Abubucker S, Murphy LO, and Cullen TW (2017). Total Lipopolysaccharide from the Human Gut Microbiome Silences Toll-Like Receptor Signaling. mSystems 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Pither MD, Illiano A, Pagliuca C, Jacobson A, Mantova G, Stornaiuolo A, Colicchio R, Vitiello M, Pinto G, and Silipo A (2022). Bacteroides thetaiotaomicron rough-type lipopolysaccharide: The chemical structure and the immunological activity. Carbohydrate Polymers 297, 120040. [DOI] [PubMed] [Google Scholar]
  • 154.Vatanen T, Kostic AD, d’Hennezel E, Siljander H, Franzosa EA, Yassour M, Kolde R, Vlamakis H, Arthur TD, Hamalainen AM, et al. (2016). Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell 165, 842–853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Deriu E, Boxx GM, He X, Pan C, Benavidez SD, Cen L, Rozengurt N, Shi W, and Cheng G (2016). Influenza Virus Affects Intestinal Microbiota and Secondary Salmonella Infection in the Gut through Type I Interferons. PLoS pathogens 12, e1005572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Winter SE, and Baumler AJ (2014). Why related bacterial species bloom simultaneously in the gut: principles underlying the ‘Like will to like’ concept. Cell Microbiol 16, 179–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Scales BS, Dickson RP, and Huffnagle GB (2016). A tale of two sites: how inflammation can reshape the microbiomes of the gut and lungs. J Leukoc Biol 100, 943–950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, and Gordon JI (2006). An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 1027–1031. [DOI] [PubMed] [Google Scholar]
  • 159.Maruvada P, Leone V, Kaplan LM, and Chang EB (2017). The Human Microbiome and Obesity: Moving beyond Associations. Cell host & microbe 22, 589–599. [DOI] [PubMed] [Google Scholar]
  • 160.Yang D, Johnson J, Zhou X, Deych E, Shands B, Hanson B, Sodergren E, Weinstock G, and Shannon WD (2019). New statistical method identifies cytokines that distinguish stool microbiomes. Scientific reports 9, 20082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Mallick H, Franzosa EA, McLver LJ, Banerjee S, Sirota-Madi A, Kostic AD, Clish CB, Vlamakis H, Xavier RJ, and Huttenhower C (2019). Predictive metabolomic profiling of microbial communities using amplicon or metagenomic sequences. Nat Commun 10, 3136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Park SY, Rao C, Coyte KZ, Kuziel GA, Zhang Y, Huang W, Franzosa EA, Weng JK, Huttenhower C, and Rakoff-Nahoum S (2022). Strain-level fitness in the gut microbiome is an emergent property of glycans and a single metabolite. Cell 185, 513–529 e521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Petersen LM, Bautista EJ, Nguyen H, Hanson BM, Chen L, Lek SH, Sodergren E, and Weinstock GM (2017). Community characteristics of the gut microbiomes of competitive cyclists. Microbiome 5, 98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Patra AK, and Yu Z (2022). Genomic Insights into the Distribution of Peptidases and Proteolytic Capacity among Prevotella and Paraprevotella Species. Microbiol Spectr 10, e0218521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Lamichhane S, Sen P, Alves MA, Ribeiro HC, Raunioniemi P, Hyotylainen T, and Oresic M (2021). Linking Gut Microbiome and Lipid Metabolism: Moving beyond Associations. Metabolites 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Flint HJ, and Duncan S (2014). Bacteroides and prevotella. [Google Scholar]
  • 167.Visconti A, Le Roy CI, Rosa F, Rossi N, Martin TC, Mohney RP, Li W, de Rinaldis E, Bell JT, Venter JC, et al. (2019). Interplay between the human gut microbiome and host metabolism. Nat Commun 10, 4505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Diener C, Dai CL, Wilmanski T, Baloni P, Smith B, Rappaport N, Hood L, Magis AT, and Gibbons SM (2022). Genome-microbiome interplay provides insight into the determinants of the human blood metabolome. Nat Metab 4, 1560–1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Song B-J, Akbar M, Jo I, Hardwick JP, and Abdelmegeed MA (2015). Translational implications of the alcohol-metabolizing enzymes, including cytochrome P450–2E1, in alcoholic and nonalcoholic liver disease. Advances in Pharmacology 74, 303–372. [DOI] [PubMed] [Google Scholar]
  • 170.Engen PA, Green SJ, Voigt RM, Forsyth CB, and Keshavarzian A (2015). The Gastrointestinal Microbiome: Alcohol Effects on the Composition of Intestinal Microbiota. Alcohol Res 37, 223–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Kosnicki KL, Penprase JC, Cintora P, Torres PJ, Harris GL, Brasser SM, and Kelley ST (2019). Effects of moderate, voluntary ethanol consumption on the rat and human gut microbiome. Addict Biol 24, 617–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Fan X, Peters BA, Jacobs EJ, Gapstur SM, Purdue MP, Freedman ND, Alekseyenko AV, Wu J, Yang L, Pei Z, et al. (2018). Drinking alcohol is associated with variation in the human oral microbiome in a large study of American adults. Microbiome 6, 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Yokoyama S, Takeuchi K, Shibata Y, Kageyama S, Matsumi R, Takeshita T, and Yamashita Y (2018). Characterization of oral microbiota and acetaldehyde production. J Oral Microbiol 10, 1492316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Tagaino R, Washio J, Abiko Y, Tanda N, Sasaki K, and Takahashi N (2019). Metabolic property of acetaldehyde production from ethanol and glucose by oral Streptococcus and Neisseria. Scientific reports 9, 10446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Yuan J, Chen C, Cui J, Lu J, Yan C, Wei X, Zhao X, Li N, Li S, Xue G, et al. (2019). Fatty Liver Disease Caused by High-Alcohol-Producing Klebsiella pneumoniae. Cell metabolism 30, 675–688 e677. [DOI] [PubMed] [Google Scholar]
  • 176.Li NN, Li W, Feng JX, Zhang WW, Zhang R, Du SH, Liu SY, Xue GH, Yan C, Cui JH, et al. (2021). High alcohol-producing Klebsiella pneumoniae causes fatty liver disease through 2,3-butanediol fermentation pathway in vivo. Gut Microbes 13, 1979883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Sakamoto M, Ikeyama N, Yuki M, and Ohkuma M (2018). Draft Genome Sequence of Faecalimonas umbilicata JCM 30896(T), an Acetate-Producing Bacterium Isolated from Human Feces. Microbiol Resour Announc 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Spector M (2009). Metabolism, central (intermediary). [Google Scholar]
  • 179.Tsuruya A, Kuwahara A, Saito Y, Yamaguchi H, Tsubo T, Suga S, Inai M, Aoki Y, Takahashi S, Tsutsumi E, et al. (2016). Ecophysiological consequences of alcoholism on human gut microbiota: implications for ethanol-related pathogenesis of colon cancer. Scientific reports 6, 27923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Petersen C, Bell R, Klag KA, Lee SH, Soto R, Ghazaryan A, Buhrke K, Ekiz HA, Ost KS, Boudina S, et al. (2019). T cell-mediated regulation of the microbiota protects against obesity. Science 365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Lin YC, Lin HF, Wu CC, Chen CL, and Ni YH (2022). Pathogenic effects of Desulfovibrio in the gut on fatty liver in diet-induced obese mice and children with obesity. Journal of gastroenterology. [DOI] [PubMed] [Google Scholar]
  • 182.Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, Almeida M, Arumugam M, Batto JM, Kennedy S, et al. (2013). Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541–546. [DOI] [PubMed] [Google Scholar]
  • 183.Wilmanski T, Rappaport N, Earls JC, Magis AT, Manor O, Lovejoy J, Omenn GS, Hood L, Gibbons SM, and Price ND (2019). Blood metabolome predicts gut microbiome alpha-diversity in humans. Nat Biotechnol 37, 1217–1228. [DOI] [PubMed] [Google Scholar]
  • 184.Passmore IJ, Letertre MPM, Preston MD, Bianconi I, Harrison MA, Nasher F, Kaur H, Hong HA, Baines SD, Cutting SM, et al. (2018). Para-cresol production by Clostridium difficile affects microbial diversity and membrane integrity of Gram-negative bacteria. PLoS pathogens 14, e1007191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Koppe L, Alix PM, Croze ML, Chambert S, Vanholder R, Glorieux G, Fouque D, and Soulage CO (2017). p-Cresyl glucuronide is a major metabolite of p-cresol in mouse: in contrast to p-cresyl sulphate, p-cresyl glucuronide fails to promote insulin resistance. Nephrol Dial Transplant 32, 2000–2009. [DOI] [PubMed] [Google Scholar]
  • 186.Kasahara K, Krautkramer KA, Org E, Romano KA, Kerby RL, Vivas EI, Mehrabian M, Denu JM, Backhed F, Lusis AJ, and Rey FE (2018). Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat Microbiol 3, 1461–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.La Rosa SL, Leth ML, Michalak L, Hansen ME, Pudlo NA, Glowacki R, Pereira G, Workman CT, Arntzen MO, Pope PB, et al. (2019). The human gut Firmicute Roseburia intestinalis is a primary degrader of dietary beta-mannans. Nat Commun 10, 905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Nie K, Ma K, Luo W, Shen Z, Yang Z, Xiao M, Tong T, Yang Y, and Wang X (2021). Roseburia intestinalis: A Beneficial Gut Organism From the Discoveries in Genus and Species. Front Cell Infect Microbiol 11, 757718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.VanderWeele TJ (2016). Mediation Analysis: A Practitioner’s Guide. Annu Rev Public Health 37, 17–32. [DOI] [PubMed] [Google Scholar]
  • 190.Jiang Z, Zhuo LB, He Y, Fu Y, Shen L, Xu F, Gou W, Miao Z, Shuai M, Liang Y, et al. (2022). The gut microbiota-bile acid axis links the positive association between chronic insomnia and cardiometabolic diseases. Nat Commun 13, 3002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Takeuchi T, Kubota T, Nakanishi Y, Tsugawa H, Suda W, Kwon AT, Yazaki J, Ikeda K, Nemoto S, Mochizuki Y, et al. (2023). Gut microbial carbohydrate metabolism contributes to insulin resistance. Nature 621, 389–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Lee H, Cashin AG, Lamb SE, Hopewell S, Vansteelandt S, VanderWeele TJ, MacKinnon DP, Mansell G, Collins GS, Golub RM, et al. (2021). A Guideline for Reporting Mediation Analyses of Randomized Trials and Observational Studies: The AGReMA Statement. JAMA 326, 1045–1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Demmer RT, Breskin A, Rosenbaum M, Zuk A, LeDuc C, Leibel R, Paster B, Desvarieux M, Jacobs DR Jr., and Papapanou PN (2017). The subgingival microbiome, systemic inflammation and insulin resistance: The Oral Infections, Glucose Intolerance and Insulin Resistance Study. J Clin Periodontol 44, 255–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Demmer RT, Squillaro A, Papapanou PN, Rosenbaum M, Friedewald WT, Jacobs DR Jr., and Desvarieux M (2012). Periodontal infection, systemic inflammation, and insulin resistance: results from the continuous National Health and Nutrition Examination Survey (NHANES) 1999–2004. Diabetes Care 35, 2235–2242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Li X, Kolltveit KM, Tronstad L, and Olsen I (2000). Systemic diseases caused by oral infection. Clinical microbiology reviews 13, 547–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Daskova N, Modos I, Krbcova M, Kuzma M, Pelantova H, Hradecky J, Heczkova M, Bratova M, Videnska P, Splichalova P, et al. (2023). Multi-omics signatures in new-onset diabetes predict metabolic response to dietary inulin: findings from an observational study followed by an interventional trial. Nutr Diabetes 13, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Berglund L, Berne C, Svardsudd K, Garmo H, Melhus H, and Zethelius B (2012). Seasonal variations of insulin sensitivity from a euglycemic insulin clamp in elderly men. Ups J Med Sci 117, 35–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Chen SH, Chuang SY, Lin KC, Tsai ST, and Chou P (2008). Community-based study on summer-winter difference in insulin resistance in Kin-Chen, Kinmen, Taiwan. J Chin Med Assoc 71, 619–627. [DOI] [PubMed] [Google Scholar]
  • 199.Wilmanski T, Kornilov SA, Diener C, Conomos MP, Lovejoy JC, Sebastiani P, Orwoll ES, Hood L, Price ND, Rappaport N, et al. (2022). Heterogeneity in statin responses explained by variation in the human gut microbiome. Med 3, 388–405 e386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Ding T, and Schloss PD (2014). Dynamics and associations of microbial community types across the human body. Nature 509, 357–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Adler CJ, Dobney K, Weyrich LS, Kaidonis J, Walker AW, Haak W, Bradshaw CJ, Townsend G, Soltysiak A, Alt KW, et al. (2013). Sequencing ancient calcified dental plaque shows changes in oral microbiota with dietary shifts of the Neolithic and Industrial revolutions. Nat Genet 45, 450–455, 455e451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Asangba AE, Mugisha L, Rukundo J, Lewis RJ, Halajian A, Cortes-Ortiz L, Junge RE, Irwin MT, Karlson J, Perkin A, et al. (2022). Large Comparative Analyses of Primate Body Site Microbiomes Indicate that the Oral Microbiome Is Unique among All Body Sites and Conserved among Nonhuman Primates. Microbiol Spectr 10, e0164321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Lv L, Jiang H, Yan R, Xu D, Wang K, Wang Q, Chen X, and Li L (2021). The Salivary Microbiota, Cytokines, and Metabolome in Patients with Ankylosing Spondylitis Are Altered and More Proinflammatory than Those in Healthy Controls. mSystems 6, e0117320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Elias AE, McBain AJ, and O’Neill CA (2021). The role of the skin microbiota in the modulation of cutaneous inflammation-Lessons from the gut. Exp Dermatol 30, 1509–1516. [DOI] [PubMed] [Google Scholar]
  • 205.Harris-Tryon TA, and Grice EA (2022). Microbiota and maintenance of skin barrier function. Science 376, 940–945. [DOI] [PubMed] [Google Scholar]
  • 206.Wu H, and Ballantyne CM (2020). Metabolic Inflammation and Insulin Resistance in Obesity. Circ Res 126, 1549–1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Vujkovic-Cvijin I, Sklar J, Jiang L, Natarajan L, Knight R, and Belkaid Y (2020). Host variables confound gut microbiota studies of human disease. Nature 587, 448–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Zhu L, Baker SS, Gill C, Liu W, Alkhouri R, Baker RD, and Gill SR (2013). Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 57, 601–609. [DOI] [PubMed] [Google Scholar]
  • 209.Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R, et al. (2011). Linking long-term dietary patterns with gut microbial enterotypes. Science 334, 105–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Gribbon EM, Cunliffe WJ, and Holland KT (1993). Interaction of Propionibacterium acnes with skin lipids in vitro. J Gen Microbiol 139, 1745–1751. [DOI] [PubMed] [Google Scholar]
  • 211.Mayslich C, Grange PA, and Dupin N (2021). Cutibacterium acnes as an Opportunistic Pathogen: An Update of Its Virulence-Associated Factors. Microorganisms 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Brown EM, Ke X, Hitchcock D, Jeanfavre S, Avila-Pacheco J, Nakata T, Arthur TD, Fornelos N, Heim C, Franzosa EA, et al. (2019). Bacteroides-Derived Sphingolipids Are Critical for Maintaining Intestinal Homeostasis and Symbiosis. Cell host & microbe 25, 668–680 e667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Kenny DJ, Plichta DR, Shungin D, Koppel N, Hall AB, Fu B, Vasan RS, Shaw SY, Vlamakis H, Balskus EP, and Xavier RJ (2020). Cholesterol Metabolism by Uncultured Human Gut Bacteria Influences Host Cholesterol Level. Cell host & microbe 28, 245–257 e246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Cryan JF, and Mazmanian SK (2022). Microbiota-brain axis: Context and causality. Science 376, 938–939. [DOI] [PubMed] [Google Scholar]
  • 215.Needham BD, Funabashi M, Adame MD, Wang Z, Boktor JC, Haney J, Wu WL, Rabut C, Ladinsky MS, Hwang SJ, et al. (2022). A gut-derived metabolite alters brain activity and anxiety behaviour in mice. Nature 602, 647–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Budden KF, Gellatly SL, Wood DL, Cooper MA, Morrison M, Hugenholtz P, and Hansbro PM (2017). Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol 15, 55–63. [DOI] [PubMed] [Google Scholar]
  • 217.Marsland BJ, Trompette A, and Gollwitzer ES (2015). The Gut-Lung Axis in Respiratory Disease. Ann Am Thorac Soc 12 Suppl 2, S150–156. [DOI] [PubMed] [Google Scholar]
  • 218.Atarashi K, Suda W, Luo C, Kawaguchi T, Motoo I, Narushima S, Kiguchi Y, Yasuma K, Watanabe E, Tanoue T, et al. (2017). Ectopic colonization of oral bacteria in the intestine drives TH1 cell induction and inflammation. Science 358, 359–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Samuelson DR, Shellito JE, Maffei VJ, Tague ED, Campagna SR, Blanchard EE, Luo M, Taylor CM, Ronis MJJ, Molina PE, and Welsh DA (2017). Alcohol-associated intestinal dysbiosis impairs pulmonary host defense against Klebsiella pneumoniae. PLoS pathogens 13, e1006426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Shellito JE, quan Zheng M, Ye P, Ruan S, Shean MK, and Kolls J (2001). Effect of alcohol consumption on host release of interleukin-17 during pulmonary infection with Klebsiella pneumoniae. Alcohol Clin Exp Res 25, 872–881. [PubMed] [Google Scholar]
  • 221.Samuelson DR, Gu M, Shellito JE, Molina PE, Taylor CM, Luo M, and Welsh DA (2021). Pulmonary immune cell trafficking promotes host defense against alcohol-associated Klebsiella pneumonia. Commun Biol 4, 997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Louis P, and Flint HJ (2009). Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol Lett 294, 1–8. [DOI] [PubMed] [Google Scholar]
  • 223.Sakamoto M, Iino T, Yuki M, and Ohkuma M (2018). Lawsonibacter asaccharolyticus gen. nov., sp. nov., a butyrate-producing bacterium isolated from human faeces. Int J Syst Evol Microbiol 68, 2074–2081. [DOI] [PubMed] [Google Scholar]
  • 224.Klaring K, Hanske L, Bui N, Charrier C, Blaut M, Haller D, Plugge CM, and Clavel T (2013). Intestinimonas butyriciproducens gen. nov., sp. nov., a butyrate-producing bacterium from the mouse intestine. Int J Syst Evol Microbiol 63, 4606–4612. [DOI] [PubMed] [Google Scholar]
  • 225.Bui TPN, Troise AD, Nijsse B, Roviello GN, Fogliano V, and de Vos WM (2020). Intestinimonas-like bacteria are important butyrate producers that utilize Nε-fructosyllysine and lysine in formula-fed infants and adults. Journal of functional foods 70, 103974. [Google Scholar]
  • 226.GREENE JA, and KEOHEN GF (1943). INSULIN RESISTANCE DUE TO INFECTION IN DIABETES MELLITUS IN MAN. Journal of the American Medical Association 121, 173–176. [Google Scholar]
  • 227.Sestan M, Marinovic S, Kavazovic I, Cekinovic D, Wueest S, Turk Wensveen T, Brizic I, Jonjic S, Konrad D, Wensveen FM, and Polic B (2018). Virus-Induced Interferon-gamma Causes Insulin Resistance in Skeletal Muscle and Derails Glycemic Control in Obesity. Immunity 49, 164–177 e166. [DOI] [PubMed] [Google Scholar]
  • 228.Meisel JS, Hannigan GD, Tyldsley AS, SanMiguel AJ, Hodkinson BP, Zheng Q, and Grice EA (2016). Skin Microbiome Surveys Are Strongly Influenced by Experimental Design. J Invest Dermatol 136, 947–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.He Y, Wu W, Zheng HM, Li P, McDonald D, Sheng HF, Chen MX, Chen ZH, Ji GY, Zheng ZD, et al. (2018). Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat Med 24, 1532–1535. [DOI] [PubMed] [Google Scholar]
  • 230.Gaulke CA, and Sharpton TJ (2018). The influence of ethnicity and geography on human gut microbiome composition. Nat Med 24, 1495–1496. [DOI] [PubMed] [Google Scholar]
  • 231.Rast P, and Hofer SM (2014). Longitudinal design considerations to optimize power to detect variances and covariances among rates of change: simulation results based on actual longitudinal studies. Psychol Methods 19, 133–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Taris TW, and Kompier MA (2006). Games researchers play--extreme-groups analysis and mediation analysis in longitudinal occupational health research. Scand J Work Environ Health 32, 463–472. [DOI] [PubMed] [Google Scholar]
  • 233.Amrhein V, and Greenland S (2018). Remove, rather than redefine, statistical significance. Nat Hum Behav 2, 4. [DOI] [PubMed] [Google Scholar]
  • 234.Wasserstein RL, and Lazar NA (2016). The ASA statement on p-values: context, process, and purpose. Taylor & Francis. [Google Scholar]
  • 235.Gruber S, and Tchetgen Tchetgen E (2016). Limitations of empirical calibration of p-values using observational data. Stat Med 35, 3869–3882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Benjamin DJ, Berger JO, Johannesson M, Nosek BA, Wagenmakers EJ, Berk R, Bollen KA, Brembs B, Brown L, Camerer C, et al. (2018). Redefine statistical significance. Nat Hum Behav 2, 6–10. [DOI] [PubMed] [Google Scholar]
  • 237.McShane BB, Gal D, Gelman A, Robert C, and Tackett JL (2019). Abandon statistical significance. The American Statistician 73, 235–245. [Google Scholar]
  • 238.Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, Fierer N, and Knight R (2011). Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proceedings of the National Academy of Sciences of the United States of America 108 Suppl 1, 4516–4522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, and Holmes SP (2016). DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods 13, 581–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.McMurdie PJ, and Holmes S (2013). phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PloS one 8, e61217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Friedman J, and Alm EJ (2012). Inferring correlation networks from genomic survey data. PLoS Comput Biol 8, e1002687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Hummel W, and Kula M-R (1989). Simple method for small-scale disruption of bacteria and yeasts. Journal of microbiological methods 9, 201–209. [Google Scholar]
  • 243.Cady NC, Stelick S, and Batt CA (2003). Nucleic acid purification using microfabricated silicon structures. Biosens Bioelectron 19, 59–66. [DOI] [PubMed] [Google Scholar]
  • 244.Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, Owens SM, Betley J, Fraser L, Bauer M, et al. (2012). Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. The ISME journal 6, 1621–1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Minalla A, Dubrow RS, and Bousse LJ (2001). Feasibility of high-resolution oligonucleotide separation on a microchip. [Google Scholar]
  • 246.Almonacid DE, Kraal L, Ossandon FJ, Budovskaya YV, Cardenas JP, Bik EM, Goddard AD, Richman J, and Apte ZS (2017). 16S rRNA gene sequencing and healthy reference ranges for 28 clinically relevant microbial taxa from the human gut microbiome. PloS one 12, e0176555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Cole JR, Wang Q, Fish JA, Chai B, McGarrell DM, Sun Y, Brown CT, Porras-Alfaro A, Kuske CR, and Tiedje JM (2014). Ribosomal Database Project: data and tools for high throughput rRNA analysis. Nucleic acids research 42, D633–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Kuczynski J, Liu Z, Lozupone C, McDonald D, Fierer N, and Knight R (2010). Microbial community resemblance methods differ in their ability to detect biologically relevant patterns. Nat Methods 7, 813–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Contrepois K, Wu S, Moneghetti KJ, Hornburg D, Ahadi S, Tsai MS, Metwally AA, Wei E, Lee-McMullen B, Quijada JV, et al. (2020). Molecular Choreography of Acute Exercise. Cell 181, 1112–1130 e1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Contrepois K, Mahmoudi S, Ubhi BK, Papsdorf K, Hornburg D, Brunet A, and Snyder M (2018). Cross-Platform Comparison of Untargeted and Targeted Lipidomics Approaches on Aging Mouse Plasma. Scientific reports 8, 17747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Hornburg D, Wu S, Moqri M, Zhou X, Contrepois K, Bararpour N, Traber GM, Su B, Metwally AA, Avina M, et al. (2023). Dynamic lipidome alterations associated with human health, disease and ageing. Nat Metab 5, 1578–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Contrepois K, Jiang L, and Snyder M (2015). Optimized Analytical Procedures for the Untargeted Metabolomic Profiling of Human Urine and Plasma by Combining Hydrophilic Interaction (HILIC) and Reverse-Phase Liquid Chromatography (RPLC)-Mass Spectrometry. Mol Cell Proteomics 14, 1684–1695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Rost HL, Schmitt U, Aebersold R, and Malmstrom L (2014). pyOpenMS: a Python-based interface to the OpenMS mass-spectrometry algorithm library. Proteomics 14, 74–77. [DOI] [PubMed] [Google Scholar]
  • 254.Rost HL, Liu Y, D’Agostino G, Zanella M, Navarro P, Rosenberger G, Collins BC, Gillet L, Testa G, Malmstrom L, and Aebersold R (2016). TRIC: an automated alignment strategy for reproducible protein quantification in targeted proteomics. Nat Methods 13, 777–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Jiang C, Zhang X, Gao P, Chen Q, and Snyder M (2021). Decoding personal biotic and abiotic airborne exposome. Nat Protoc 16, 1129–1151. [DOI] [PubMed] [Google Scholar]
  • 256.Contreras PH, Salgado AM, Bernal YA, and Vigil P (2019). A Simple and Improved Predictor of Insulin Resistance Extracted From the Oral Glucose Tolerance Test: The I0*G60. J Endocr Soc 3, 1154–1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Kulesa A, Krzywinski M, Blainey P, and Altman N (2015). Sampling distributions and the bootstrap. Nat Methods 12, 477–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Shade A, and Handelsman J (2012). Beyond the Venn diagram: the hunt for a core microbiome. Environ Microbiol 14, 4–12. [DOI] [PubMed] [Google Scholar]
  • 259.Risely A (2020). Applying the core microbiome to understand host-microbe systems. J Anim Ecol 89, 1549–1558. [DOI] [PubMed] [Google Scholar]
  • 260.Salonen A, Salojarvi J, Lahti L, and de Vos WM (2012). The adult intestinal core microbiota is determined by analysis depth and health status. Clin Microbiol Infect 18 Suppl 4, 16–20. [DOI] [PubMed] [Google Scholar]
  • 261.Bates D, Mächler M, Bolker B, and Walker S (2015). Fitting Linear Mixed-Effects Models Using lme4. Journal of Statistical Software 67, 1 – 48. [Google Scholar]
  • 262.Nelson LS (1998). The Anderson-Darling Test for Normality. Journal of Quality Technology 30, 298–299. [Google Scholar]
  • 263.Johnson SG (2011). The NLopt nonlinear-optimization package. [Google Scholar]
  • 264.Zeileis A, and Hothorn T (2002). Diagnostic checking in regression relationships. [Google Scholar]
  • 265.Satterthwaite FE (1946). An approximate distribution of estimates of variance components. Biometrics 2, 110–114. [PubMed] [Google Scholar]
  • 266.Bürkner P-C (2017). brms: An R package for Bayesian multilevel models using Stan. Journal of statistical software 80, 1–28. [Google Scholar]
  • 267.Bürkner P-C (2017). Advanced Bayesian Multilevel Modeling with the R Package brms. arXiv preprint arXiv:1705.11123. [Google Scholar]
  • 268.Bürkner P-C (2021). Bayesian Item Response Modeling in R with brms and Stan. Journal of Statistical Software 100, 1 – 54. [Google Scholar]
  • 269.Hoffman MD, and Gelman A (2014). The No-U-turn sampler: adaptively setting path lengths in Hamiltonian Monte Carlo. Journal of Machine Learning Research 15, 1593–1623. [Google Scholar]
  • 270.Altman DG, and Bland JM (2011). How to obtain the P value from a confidence interval. BMJ 343, d2304. [DOI] [PubMed] [Google Scholar]
  • 271.Fang H, Huang C, Zhao H, and Deng M (2015). CCLasso: correlation inference for compositional data through Lasso. Bioinformatics 31, 3172–3180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Hughes DA, Bacigalupe R, Wang J, Ruhlemann MC, Tito RY, Falony G, Joossens M, Vieira-Silva S, Henckaerts L, Rymenans L, et al. (2020). Genome-wide associations of human gut microbiome variation and implications for causal inference analyses. Nat Microbiol 5, 1079–1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Koh A, and Backhed F (2020). From Association to Causality: the Role of the Gut Microbiota and Its Functional Products on Host Metabolism. Mol Cell 78, 584–596. [DOI] [PubMed] [Google Scholar]
  • 274.Fu W (2019). Statistical issues in microbiome data analysis: batch effects and multi-omics analysis. [Google Scholar]
  • 275.Schaum N, Lehallier B, Hahn O, Palovics R, Hosseinzadeh S, Lee SE, Sit R, Lee DP, Losada PM, Zardeneta ME, et al. (2020). Ageing hallmarks exhibit organ-specific temporal signatures. Nature 583, 596–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Boedigheimer MJ, Wolfinger RD, Bass MB, Bushel PR, Chou JW, Cooper M, Corton JC, Fostel J, Hester S, and Lee JS (2008). Sources of variation in baseline gene expression levels from toxicogenomics study control animals across multiple laboratories. BMC genomics 9, 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Poore GD, Kopylova E, Zhu Q, Carpenter C, Fraraccio S, Wandro S, Kosciolek T, Janssen S, Metcalf J, and Song SJ (2020). Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature 579, 567–574. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 278.Wood SN (2004). Stable and efficient multiple smoothing parameter estimation for generalized additive models. Journal of the American Statistical Association 99, 673–686. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2

Supplementary Table 1. Meta-Data, Statistical Analyses, and Taxa Abundance in IS and IR Individuals, related to Figure 1

S1A. Participants related metadata, related to Figure 1A. S1B. The p-value 1284 of smooth term in GAMM model for seasonal microbial variations, related to Figure S1E. S1C. Chi-Square Test result to determine if taxa are more likely a core taxon in either IS or IR individual, related to Figure 1F. S1D. LEFSE result (comparing the relative abundance of all taxa by individual mean value), related to Figure S1M.

Supplementary Table 2. Intra- and Interindividual BC Distance, DMI Comparisons by Phylum and Genera, related to Figure 2

S2A. Wilcoxon signed-rank test of BC distance intraindividual (within), interindividual (between) and within the same household (family), related to Figure 2A. S2B. The DMI comparison between body sites by phylum, related to Figure 2C. S2C. DMI and FS of all genera, related to Figure 2D.

Supplementary Table 3. Correlations and GAMM Analysis of Microbial Genera Across Body Sites and Infections, related to Figure 3

S3A. Comparison of the proportion of inter-correlated genera both within and between various body sites, related to Figure 3D. S3B. Intra individual correlations between microbiome genera, related to Figure 3D. S3C. Inter-individual correlation between microbial genera using SPARCC, related to Figure 3D. S3D. Generalized Additive Mixed Models (GAMM) on microbial perturbations during infection, related to Figure S3F, S3G, S3H.

Supplementary Table 4. Correlated Cytokine-Microbiome Pairs and Cytokine Correlations by Body Site, related to Figure 4

S4A. The number of significantly correlated pairs of cytokine and microbiome, based on cytokines and body sites related to Figure 4A. S4B. Pairwise cytokine correlations, from the BRMS model related to Figure 4A.

Supplementary Table 5. Lipid Annotations and LMM Results on Omics-Microbiome Correlations, related to Figure 5

S5A. Lipid module and their associated lipid species annotation, related to Figure 5A, Figure S6A. S5B. The result from LMM infers the internal omics and microbiome relative abundance longitudinal correlations, related to Figure 5A. S5C. The result from LMM infers the internal omics and microbiome richness longitudinal correlations, related to Figure 5C.

Supplementary Table 6. Clinical Lab Test Annotations and Mediation Analysis Outcomes, related to Figure 6

S6A. Annotation from the clinical lab test and their phenotype class, related to Figure 6A. S6B. The mediation analysis results, related to Figure 6A. S6C. The comparison between different mediate effects between IR and IS, related to Figure 6A.

Data Availability Statement

The microbiome data are available at https://hmpdacc.org. Other omics data can be accessed through Stanford iPOP website at https://med.stanford.edu/ipop.html.

The microbiome data specific to this study can be accessed at GitHub Repository (https://github.com/xzhou7/iHMP). The metadata corresponding to these data files can be found at Stanford Data Repository (http://hmp2-data.stanford.edu/script.php?table=subject).

Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

RESOURCES