Skip to main content
JID Innovations logoLink to JID Innovations
. 2022 Mar 10;2(3):100115. doi: 10.1016/j.xjidi.2022.100115

Multiomic Analysis of the Gut Microbiome in Psoriasis Reveals Distinct Host‒Microbe Associations

Hsin-Wen Chang 1, Di Yan 2, Rasnik Singh 3, Audrey Bui 4, Kristina Lee 1, Alexa Truong 5, Jeffrey M Milush 6, Ma Somsouk 7, Wilson Liao 1,
PMCID: PMC9214347  PMID: 35757783

Abstract

Psoriasis is a chronic, inflammatory skin disease that affects 2‒3% of the global population. Besides skin manifestations, patients with psoriasis have increased susceptibility to a number of comorbidities, including psoriatic arthritis, cardiovascular disease, and inflammatory bowel disease. To understand the systemic component of psoriasis pathogenesis, we performed a pilot study to examine the fecal metagenome, host colonic transcriptome, and host peripheral blood immune profiles of patients with psoriasis and healthy controls. Our study showed increased functional diversity in the gut microbiome of patients with psoriasis. In addition, we identified microbial species that preferentially associate with patients with psoriasis and which have been previously found to associate with other autoimmune diseases. Intriguingly, our data revealed three psoriasis subgroups that have distinct microbial and host features. Integrating these features revealed host‒microbe associations that are specific to psoriasis or particular psoriasis subgroups. Our findings provide insight into the factors that may affect the development of comorbidities in patients with psoriasis and may hold diagnostic potential for early identification of patients with psoriasis at risk for these comorbidities.

Abbreviations: IBD, inflammatory bowel disease; RNA-seq, RNA sequencing

Introduction

Psoriasis is a prevalent immune-mediated disease characterized by inflamed skin lesions and epidermal hyperproliferation. A total of 2‒3% of the global population is affected by psoriasis, and the disease is known to be heterogeneous and multifactorial. It has been well established that psoriasis has different subtypes on the basis of distinct disease characteristics. Plaque psoriasis (psoriasis vulgaris) is the most prevalent psoriasis subtype, with others being guttate, inverse, erythrodermic, and pustular. Psoriasis pathogenesis is multifactorial with a strong genetic component (Elder et al., 2010). Recent studies have suggested that the microbiome, diet, and other environmental factors may also have a role in psoriasis pathogenesis (Afifi et al., 2017; Benhadou et al., 2018; Chang et al., 2018; Codoñer et al., 2018; Fahlén et al., 2012; Fyhrquist et al., 2019; Gao et al., 2008; Loesche et al., 2018; Takemoto et al., 2015; Tett et al., 2017; Yan et al., 2017). At the molecular level, aberrant activation of IL-17 signaling pathway is one of the major contributors to the disease (Brembilla et al., 2018). Therapeutically blocking components in IL-17 signaling pathway usually controls skin inflammation. However, the effectiveness of psoriasis treatments varies across patients, further highlighting the heterogeneous nature of psoriasis.

In addition to the skin manifestations, patients with psoriasis are at higher risk of developing a number of other comorbidities. Psoriatic arthritis is one of the most prevalent comorbidities among patients with psoriasis because up to one third of patients with psoriasis transition into psoriatic arthritis (Ritchlin et al., 2017). Patients with psoriasis also have a 2.5-fold higher risk of developing Crohn’s disease and a 1.7-fold higher risk of developing ulcerative colitis, linking psoriasis to inflammatory bowel disease (IBD) (Fu et al., 2018). Other common psoriasis comorbidities include cardiovascular disease (Prodanovich et al., 2009) and type 2 diabetes (Wan et al., 2018). Although the association between psoriasis and its comorbidities is firmly established, the underlying cause and triggers are yet to be elucidated. Developing these comorbidities in patients with psoriasis not only increases the disease burden for the patients but also complicates strategies in treatment and diagnosis. The ability to risk stratify patients with psoriasis for the development of certain comorbidities would greatly enhance strategies toward prevention, early detection, and treatment.

Although the skin microbiome has been the focus for cutaneous autoimmune disease, dysbiosis in the gut microbiome has been observed in psoriasis (Codoñer et al., 2018; Eppinga et al., 2016; Hidalgo-Cantabrana et al., 2019; Scher et al., 2015; Tan et al., 2018). Moreover, gut microbiome dysbiosis has been implicated in many psoriasis comorbidities such as psoriatic arthritis (Scher et al., 2015), IBD (Gevers et al., 2014; Knights et al., 2014; Lloyd-Price et al., 2019; Morgan et al., 2012), and type 2 diabetes (Zhou et al., 2019). Together, these findings suggest that the gut microbiome might be an important contributing factor for psoriasis pathogenesis and the emergence of psoriasis comorbidities. To better understand the systemic component of pathogenesis associated with psoriasis, we performed a detailed multiomics analysis with a cohort of patients with psoriasis and healthy individuals. We utilized shotgun metagenomic sequencing to profile both the taxonomic composition and functional capacity of the gut microbiome. In addition, we carried out RNA sequencing (RNA-seq) to profile the host intestinal transcriptome. Host peripheral blood was also collected to measure both systemic immune populations and their cytokine-producing capacity. Our study revealed several microbial features associated with psoriasis. Next, using clustering analysis, we identified three psoriasis subgroups, each with distinct microbial features that may predispose to certain psoriasis comorbidities. Finally, we performed multiomics analysis by integrating all our omics data and revealed disease and subgroup-specific host‒microbe associations. Our work highlights the heterogeneity of psoriasis and the potential role of microbial and host‒microbe associations in psoriasis pathogenesis and comorbidities. Moreover, this pilot study provides an analytical framework that can be applied to study host‒microbe association in other diseases.

Results

Study design and cohort summary

We recruited a cohort of 33 subjects with psoriasis not on systemic therapy and 15 age- and sex-matched healthy individuals (Table 1) to study the gut microbial features associated with psoriasis and their potential contribution to psoriasis pathogenesis. All patients with psoriasis were clinically diagnosed with psoriasis at the University of California San Francisco Psoriasis and Skin Treatment Center (San Francisco, CA) and had a mean PASI of 14.2, representing moderate-to-severe disease. To characterize microbial composition, we collected stool samples and subjected each sample to shotgun metagenomic sequencing that provides both taxonomic composition and functional capacity. For the subsequent analyses, we focused on bacterial species, microbial UniRef90 gene families, and microbial MetaCyc pathways. To link the changes of microbial features in psoriasis gut to the host response, we collected biopsies from the sigmoid colon and subjected these samples to RNA-seq. We also isolated PBMCs from blood samples to measure immune cell profiles and cytokine production capacity. Together, our study design (Figure 1) provided a comprehensive survey on both host biology and microbiome capacity in a cohort of patients with psoriasis in comparison with those in the healthy controls.

Table 1.

Demographic Information of Metagenomics Cohort

Characteristic Healthy Psoriasis P-Value
Sample size 15 33 NA
Sex (% female) 47% 52% 1
Age (mean age), y 45.8 ± 13.9 43.2 ± 14.6 0.5654
Mean PASI score NA 14.2 ± 13.3 NA
Median PASI score NA 10.1 NA

Abbreviation: NA, not applicable.

Figure 1.

Figure 1

Multiomic study design. In this study, we collected six different datasets for multiomic analysis: shotgun metagenomic sequencing from stool samples generated profiles of (i) microbial species, (ii) microbial gene families, and (iii) microbial gene pathways. RNA-seq from sigmoid colon biopsies generated (iv) host colonic transcriptome data. Flow cytometry analyses from PBMCs generated (v) immune population profiles and (vi) cytokine production profiles. The datasets measuring microbial features are in the green box, and the datasets measuring host features are in the yellow box. Stool samples were collected from 15 healthy subjects and 33 patients with psoriasis. Sigmoid colon biopsies and PBMCs were collected from 16 healthy subjects and 26 patients with psoriasis. A total of 14 healthy subjects and 26 patients with psoriasis had six fully complete datasets. RNA-seq, RNA sequencing.

Microbial diversity and community structure between psoriasis and healthy gut microbiome

Gut microbiome dysbiosis has been previously associated with decreased microbial diversity. Low microbial diversity has been observed in several human diseases, including IBD, obesity, and autism (Gevers et al., 2014; Hsiao et al., 2013; Turnbaugh et al., 2009). It has been hypothesized that losing microbial diversity rise from a missing group of beneficial microbes in the gut microbiome, which can lead to many detrimental consequences such as loss of control in the growth of opportunistic pathogens and lack of production of beneficial microbial-derived compounds. Alpha diversities assess the microbial diversity within a community by calculating richness (numbers of species) and evenness (even distribution of each species within a community) (Lozupone and Knight, 2005). To compare the microbial diversity in patients with psoriasis with that in healthy subjects, we calculated different alpha diversity indices to estimate the overall diversity (Shannon), evenness (Simpson), and richness (chao1) of each community. We observed higher evenness of microbial functional diversities associated with patients with psoriasis, whereas similar taxonomical diversity was observed between patients with psoriasis and healthy controls (Figure 2a and Supplementary Figure S1a and b). The overall microbial community structures were similar between psoriasis samples and healthy samples because no distinct clusters were observed in principal coordinate analysis plots for taxonomic and functional profiles (Supplementary Figure S1c and d). All patients with psoriasis in our cohort had a normal-appearing lower gastrointestinal endoscopic examination, so drastic differences in diversity and community structure in the psoriasis microbiome as observed with other gastrointestinal diseases might not be expected.

Figure 2.

Figure 2

Microbial features associated with PSO and healthy subjects. Boxplots compare alpha diversities of gut microbiome in patients with PSO and those in the healthy subjects. Alpha diversity was measured by Shannon index, Simpson diversity index, and chao1 estimation for (a) microbial Uniref90 gene families. Statistical significance was determined by Wilcoxon test. (b) Dot plot summary of DA microbial species identified by DEseq2. Each dot represents a DA microbial species with dot size‒present adjusted P-value, and x-axis represents log2 fold change. (c) Boxplots of select DA microbial species. Blue boxes represent PSO sample, and red boxes represent healthy samples. ∗∗P < 0.01. adj, adjusted; DA, differential abundant; n.s., not significant; PSO, psoriasis.

Supplementary Figure S1.

Supplementary Figure S1

Microbial diversity metrics. (a) Boxplot comparing the alpha diversity of gut microbiome in patients with PSO and healthy subjects. Alpha diversity was measured by Shannon index, Simpson diversity index, and chao1 estimation for microbial species. PCoA of the microbial community structures based on Bray‒Curtis distance matrix for (b) microbial species and (c) microbial gene families. (d) PCA of host transcriptome by top 5,000 most variable genes. In all plots, red denotes healthy samples, and blue denotes PSO samples. n.s., not significant; PC, principal component; PCA, principal component analysis; PCoA, principal coordinate analysis; PSO, psoriasis; RNA-seq, RNA sequencing.

Identification of the microbial features associated with the psoriasis gut microbiome

We hypothesized that even though the gut microbiome from patients with psoriasis has seemingly normal overall microbial community structure and diversity, the differences between psoriasis and healthy microbiome may be in specific microbial features. To identify the microbial features that are differentially abundant between gut microbiome from patients with psoriasis and healthy individuals, we performed differentially abundance analysis using DEseq2 (Love et al., 2014), which is designed for RNA-seq analysis but is widely adapted for microbiome data (McMurdie and Holmes, 2014). We estimated differential abundant features using a negative binomial model after controlling for known confounding factors for gut microbiome such as sex, age, and experimental batch. Our analysis revealed bacterial species and microbial gene families and pathways that are differentially abundant between microbiome in patients with psoriasis and healthy individuals (Figure 2b and Supplementary Table S1, Supplementary Table S2, Supplementary Table S3). Among bacterial species that are differentially abundant between psoriasis and healthy gut microbiome, we found an increase of Bacteroides vulgatus and Parasutterella excrementihominis and a decrease of Phascolarctobacterium succinatutens (Figure 2c).

Microbial gene family analysis reveals three psoriasis subgroups with distinct microbial and host features

We then performed a hierarchical clustering analysis on the microbial gene families differentially abundant between patients with psoriasis and healthy controls and identified three distinct groups in our cohort (Figure 3a). To confirm that these subgroups were not observed by chance, we performed bootstrapped gap statistics on both the Euclidean distance and Bray‒Curtis distance, which confirmed the presence of the three distinct groups in the cohort (Supplementary Figure S2a and b). Among the three groups identified by clustering, group 1 consists of a mixture of healthy and psoriasis samples (14 healthy samples and 15 psoriasis samples), group 2 consists of all patients with psoriasis (nine psoriasis samples), and group 3 consists of almost all patients with psoriasis (one healthy sample and nine psoriasis samples) (Figure 3b). For the subsequent investigations of these psoriasis subgroups, we termed the subjects with psoriasis from these subgroups PSO1, PSO2, and PSO3. The clustering was not confounded by body mass index, age, and sex or diet (Supplementary Figure S3a‒c and g and Supplementary Table S4). All psoriasis subgroups had similar disease severity, disease duration, and age of disease onset (Supplementary Figure S3d‒f). However, we found that each psoriasis subgroup had a number of distinct microbial and host features (Figure 3c). Some of the interesting features associated with each psoriasis subgroup are highlighted below.

Figure 3.

Figure 3

PSO subgroups identified by a differential abundance of microbial gene families. (a) The hierarchical cluster dendrogram shows the membership of all samples in this cohort. The colored boxes represent the grouping of each sample into three distinct groups. The red box depicts group 1, the blue box depicts group 2, and the green box depicts group 3. The red dotted line represents where the tree is cut to derive the three subgroups. The AU bootstrap confidence scores and BP values are represented in red and green, respectively, at the major branches. (b) The stacked bar plot represents the distribution of disease status of the three groups identified by cluster analysis. The height of each bar represents the size of each group, and the color represents the disease status, with red for healthy subjects and blue for PSO samples. (c) Heatmap of microbial and host features associated with each PSO subgroup. Columns represent PSO subgroups, and rows represents microbial features identified from shotgun metagenomics or host features from colonic RNA-seq. Differential abundance microbial features with nonzero counts for at least 10 samples were plotted on the heatmap to exclude features with high dropout rates. The color of each cell represents the average abundance and is scaled by means of the features. The data type of each feature is indicated in the side bar: pink represents microbial species, blue represents microbial pathway, and yellow represents host GEx. AU, arbitrary unit; BP, bootstrap probability; GEx, gene expression; PSO, psoriasis; RNA-seq, RNA sequencing.

Supplementary Figure S2.

Supplementary Figure S2

Bray-Curtis analysis. (a) Gap statistics calculated using Bray‒Curtis and Euclidean dissimilar matrix of the DA UniRef90 gene families with bootstrapping for 1,000 times. (b) PCoA plot shows the Bray‒Curtis dissimilarity matrix of the DA UniRef90 gene families grouped the cohort into three groups that are similar to the grouping defined by hierarchical clustering (as represented by the color of each point). The shapes represent the disease status (round circles depict healthy samples, and triangles depict PSO samples). DA, differentially abundant; DE, differentially expressed; maxSE, maximum numeric vector of function values; PCoA, principal coordinate analysis; PSO, psoriasis.

Supplementary Figure S3.

Supplementary Figure S3

Comparisons of metadata and microbial diversity in the three subgroups identified in the cohort. Only subjects with a complete dataset from the three measurement types are included. (a‒c) Comparisons of BMI, age, and gender in the three groups. (d‒f) Comparisons of PASI, disease onset, and disease duration in each PSO subgroup. (g, h) Comparison of diet scores and self-reported joint pain or swelling in the three groups. Comparisons of observed microbial, Shannon index, and Simpson diversity index in the three groups for (i) microbial species, (j) UniRef90 gene families, and (k) MetaCyc pathway. BMI, body mass index; Dz, disease; F, female; M, male; n.s., not significant; PSO, psoriasis; y, year.

Both PSO2 and PSO3 are dominated by subjects with psoriasis, suggesting that these are two distinct psoriasis-specific subgroups. We identified several common microbial features shared by these psoriasis subgroups as well as some microbial features that are distinct to each subgroup (Figure 3c and Supplementary Table S5, Supplementary Table S6, Supplementary Table S7). Although psoriasis samples have a lower abundance in P. succinatutens than healthy controls (Figure 2c), the reduced abundance of P. succinatutens is specific to PSO2 and PSO3 but not to PSO1 (Figure 3c). In addition, samples in PSO2 and PSO3 are less abundant with Turicibacter sanguinis and unclassified Turicibacter species (Figure 3c). Samples in PSO2 are more abundant with Bacteroides xylanisolvens and less abundant with Prevotella copri, Streptococcus thermophilus, and Coprococcus sp ART55 1 (Figure 3c). On the contrary, samples in PSO3 have a lower abundance in Ruminococcaceae bacterium D16 and Lachnospiraceae bacterium 1 1 57FAA (Figure 3c). In addition to distinct taxonomic features, PSO2 has a distinct profile in microbial functions, especially in microbial pathways. Both abundant and depleted pathways were found in PSO2 relative to those in other psoriasis subgroups, suggesting a shift of microbial functions in PSO2 (Figure 3c). Microbial communities in PSO2 have lower abundance in the arginine and polyamine biosynthesis superfamily, suggesting a lower capacity for polyamine production (Figure 3c). In contrast to PSO2, we did not identify microbial pathways that are uniquely associated with PSO1 and PSO3.

From the perspective of host response, PSO2 and PSO3 patients have distinct intestinal transcriptomic signatures (Figure 3c and Supplementary Table S8). Gut biopsies from PSO2 patients have increased expression in ATP13A5 and PDE9A and reduced expression in sulfotransferases, SULT1C2 and SULT1C3. In contrast, most of the transcriptomic signatures associated with PSO3 have increased expression compared with those associated with other psoriasis samples (Figure 3c). To gain more insight into the host intestinal immune response, we deconvoluted the immune cell composition in sigmoid colon bulk RNA-seq using the digital cytometry framework CIBERSORTx (Newman et al., 2019). As part of the CIBERSORTx framework, we first defined gene signatures of various immune cell populations in the sigmoid colon using single-cell RNA-seq generated from healthy sigmoid colon (James et al., 2020). We then used CIBERSORTx to apply the gene signature matrix to the bulk RNA-seq to infer the composition of cell populations. Sigmoid colon from patients in PSO3 has more abundant CD8+ T cells and less abundant NK cells and activated CD4+ T cells than that from other psoriasis subgroups (Figure 4a and Supplementary Table S9).

Figure 4.

Figure 4

PSO subgroup‒specific microbial and host features. (a) Boxplots show differences in sigmoid colon immune cell compositions deconvoluted by CIBERSORTx between different PSO subgroups. (b) Boxplots showing circulating host immune responses measured by flow cytometry. The statistical significance in boxplots was determined by Wilcox test on pairwise comparison of each group of interest. P-values were depicted by symbols: ∗∗∗∗P < 0.0001, ∗∗∗P < 0.001, ∗∗P < 0.01, and P < 0.05. Comparisons with P > 0.05 are not shown. PSO, psoriasis; Teff, effector T cell.

In addition to colonic transcriptomic signatures, flow cytometry analysis revealed distinct immune features in circulatory blood between the two psoriasis-dominant groups. Patients in PSO3 have higher activated memory CD4+ effector T cells than those in PSO2, whereas the frequency of memory CD4+ effector T cells and total CD4+ effector T cells are comparable between these two psoriasis subgroups (Figure 4b). Similarly, memory CD8+ T cells have a more activated population in PSO3 patients (Figure 4b). Despite lower T-cell activation, patients in PSO2 have a higher capacity to produce IL-22 by circulating CD8+ T cells and TNF-α by circulating γδ T cells (Figure 4b) than healthy controls or those in PSO1. Together, our data reveal differential underlying circulatory immune responses associated with PSO2 and PSO3. PSO2 patients have a higher capacity for proinflammatory cytokine production, whereas PSO3 patients have higher baseline T-cell activation.

Distinct correlations between psoriasis severity with microbial features in each psoriasis subgroups

To further understand the relationship between the psoriasis subgroups and psoriasis heterogeneity, we tested whether microbial features in these psoriasis subgroups are significantly correlated with psoriasis parameters. In PSO1 patients, disease severity is positively correlated with the microbial pathways involved in purine degradation (Figure 5). Disease severity in PSO2 patients is positively correlated with several microbial pathways, including pentose phosphate biosynthesis and L-arginine biosynthesis (Figure 5). The microbial L-rhamnose degradation pathway is positively correlated with disease duration in PSO2 (Figure 5). The microbial L-isoleucine biosynthesis pathway is correlated with disease duration positively in PSO1 patients and negatively in PSO2 patients (Figure 5). No significant correlations between microbial features and disease information were observed in PSO3.

Figure 5.

Figure 5

Correlations between microbial features and psoriasis-related clinical features. Dot plot summarizes the correlation between microbial features and disease parameters of PASI scores and disease duration. Each dot represents a significant correlation between a disease parameter and a microbial feature. The direction and strength of the correlation are presented by dot color (red represents positive correlation, and blue represents negative correlation). The dot size represents false discovery rate‒adjusted P-values. adj, adjusted; corr., correlation value; Dz, disease.

Multiomic analysis in patients with psoriasis and subgroups reveals distinct host‒microbe associations

To gain a comprehensive view of the crosstalk between the gut microbiome and host biology, we constructed multiomic correlation networks by integrating microbial and host features across different measurements (Supplementary Table S10). A subset of 40 subjects from our cohort with complete measurements from shotgun metagenomic sequencing, gut RNA-seq, and circulating immune profiling were included in the multiomic analysis. This multiomic cohort consists of 26 subjects with psoriasis and 14 age- and sex-matched healthy subjects (Table 2). Multiomic networks were constructed within each disease status and within each psoriasis subgroup to reveal disease-specific and subgroup-specific host‒microbe relationships. The resulting multiomic networks consist of nodes that represent host or microbial features and edges that represent significant correlations between nodes. The network in patients with psoriasis is denser and consists of more edges than the multiomic network in healthy subjects (Figure 6a). The final multiomic network consists of 73 significant correlations within healthy subjects and 97 significant correlations within patients with psoriasis (Table 3). Intriguingly, the multiomic network in each psoriasis subgroup displayed a distinct network structure. Multiomic networks in PSO1 and PSO2 were more interconnected than the network in PSO3 (Figure 6a). It is interesting that PSO3 had fewer edges and nodes than other psoriasis subgroups and all psoriasis samples combined (Table 3). Overall, our data revealed distinct multiomic network structures in patients with psoriasis and healthy controls as well as among different psoriasis subgroups.

Table 2.

Demographic Information of Subjects with Full Set of Multiomics Data

Characteristic Healthy Psoriasis P-Value
Sample size 14 26 NA
Sex (% female) 43% 58% 0.5096
Age (mean age), y 44.8 ± 13.8 44.7 ± 15.5 0.9789
Mean PASI score NA 11 ±8.9 NA
Median PASI score NA 7.7 NA

Abbreviation: NA, not applicable.

Figure 6.

Figure 6

Figure 6

Multiomic networks associated with PSO and PSO-specific subgroups. (a) Overview of multiomic networks within healthy subjects, patients with PSO, and each PSO subgroup. Each node represents a microbial or host feature, and each edge represents a significant association between the two nodes. The color of nodes represents the measurement type of the node. Host‒microbe modules are identified using greedy optimization of modularity. (b) PSO-specific modules associate microbial features with circulating IL-17 production. (c‒e) PSO subgroup‒specific modules are also identified in each PSO-specific subgroup. For each module, the color of nodes represents the measurement type of the node, and the color of edges indicates the direction of the correlation (red edges represent positive associations, and blue edges represent negative associations). DE, differentially expressed; PSO, psoriasis; RNA-seq, RNA sequencing.

Table 3.

Summary of Multiomic Networks

Characteristic Healthy Psoriasis PSO1 PSO2 PSO3
Number of subjects 14 26 12 8 6
Number of edges 73 97 124 126 53
Number of features 99 110 143 160 74
Total number of modules 30 35 28 55 26
Number of modules with at least three features 16 15 15 21 10
Number of edges with host immune features 22 13 95 12 0
Number of edges with host transcriptome 51 84 27 109 53

The different multiomic networks might reflect the different host‒microbe associations in subjects with psoriasis and healthy subjects. One of the modules in psoriasis-associated multiomic network positively links microbial pathways involved in microbial ppGpp biosynthesis and pyrimidine ribonucleosides salvage pathways with circulating IL-17 production in both CD4+ effector T cells and regulatory T cells (Figure 6b). The associations between these microbial pathways and IL-17 production were only observed in psoriasis samples despite that these microbial pathways are also being abundant in healthy samples (Supplementary Figure S4a).

Supplementary Figure S4.

Supplementary Figure S4

PSO-specific correlations between microbial functions and proinflammatory host response. (a) Scatter plots show positive correlations between microbial MetaCyc pathways and IL-17 production in psoriatic PBMCs but not in healthy PBMCs. (b) Scatter plots show a positive correlation between microbial UniRef90 gene families with the colonic expression of CXCL1 and CXCL3 in patients with PSO but not in healthy control. The correlations were calculated by Spearman’s rank-order correlation. PSO, psoriasis; Teff, effector T cell; Treg, regulatory T cell.

Besides psoriasis-specific host‒microbe associations, our analysis also identified host‒microbe associations that are specific in psoriasis subgroups. A multiomic network identified in PSO1 consists of a module that links microbial gene families with activated nonmemory CD4+ T cells and IL-17 production capacity in CD4+ effector T cells (Figure 6c), suggesting potential microbial controls in T-cell activation and effector function in this psoriasis subgroup. Indeed, 95 of 124 host‒microbe associations in PSO1 link the features of host circulating immunity to microbial features, which is higher than the networks in PSO2 and PSO3 patients (Table 3). In addition, we also identified some subgroup-specific associations in psoriasis-dominant PSO2 and PSO3. In PSO2, the expression of proinflammatory chemokine CXCL8 is positively correlated with microbial putrescine biosynthesis pathway in PSO2 patients (Figure 6d). We identified a module in PSO3 network that negatively correlates microbial tetrapyrrole biosynthesis pathway with several genes in B-cell biology, including CD79B, PAX5, TCL1A, and MYBL1 (Figure 6e). Tetrapyrroles are metal-binding compounds that serve as different cofactors, such as heme, cobalamin (vitamin B12), and coenzyme F430, and have crucial roles in regulating diverse cell functions.

Discussion

The pathogenesis of psoriasis is highly heterogeneous, which poses challenges in diagnosis and disease control. Accumulating evidence from recent studies suggests that the heterogeneity of psoriasis pathogenesis may be the result of the interplay between microbiome and host immune response. Our study focused on studying the heterogeneity of psoriasis pathogenesis through the lens of multiomic datasets assessing both host and microbial features. In this study, we show that psoriasis gut microbiome has increased B. vulgatus and P. excrementihominis and reduced P. succinatutens compared with the ones in healthy controls. These microbial features have also been associated with intestinal inflammation. Increased intestinal colonization of B. vulgatus and elevated B. vulgatus reactive serum antibodies have been reported in patients with ulcerative colitis (Matsuda et al., 2000). It is intriguing that the gut microbiome of patients with psoriasis in our cohort shares some microbial features associated with IBD or irritable bowel syndrome despite no intestinal symptoms reported in our patients.

Enterotypes defined by different microbiome compositions have been previously described in healthy subjects (Arumugam et al., 2011). Our study revealed three psoriasis subgroups in our cohort as defined by the differential abundance of microbial gene families. Each psoriasis subgroup has distinct microbial and host features (Table 4). The reduced Turicibacter species in PSO2 and PSO3 are reminiscent of the gut microbiome associated with pediatric Crohn’s disease (El Mouzan et al., 2018; Wang et al., 2016). PSO2 has the most distinct microbial functional profile (Figure 3c) from that of other psoriasis subgroups. The gut microbiome in PSO2 shows a higher biosynthetic capacity of several important immune regulators, including pyridoxal 5-phosphate (vitamin B6), L-ornithine, and flavin. We also found lower capacity in arginine and polyamine biosynthesis in PSO2. Some of these molecules have been linked to immune-mediated intestinal inflammation. For example, vitamin B6 plays a crucial role in lymphocyte trafficking into the intestines (Yoshii et al., 2019). Vitamin B6 deficiency has been linked to several immune-mediated diseases, including rheumatoid arthritis and IBD (Selhub et al., 2013; Yoshii et al., 2019). Having increased capacity of vitamin B6 biosynthesis may have a protective role for subjects in PSO2 from autoimmune diseases. On the contrary, polyamines play a crucial role in intestinal mucosa maintenance and resident immune cell development (Yoshii et al., 2019). Interestingly, it has been shown that spermine, a class of polyamine, reduces the secretion of proinflammatory IL-18 cytokine by inhibiting NLRP6 inflammasome activation (Levy et al., 2015). Our data suggest that gut microbial communities in PSO2 have increased vitamin B6 biosynthesis and reduced polyamine production, but the clinical implication requires further study.

Table 4.

Summary of Host and Microbial Features of Psoriasis Subgroups

Characteristic PSO2 and PSO3 Common Features PSO2-Specific Features PSO3-Specific Features
Metagenomics (stool) Turicibacter spp. ↓
Phascolarctobacterium succinatutens ↓
Bacteroides xylanisolvans
Prevetella copri ↓
Streptococcus thermophilus ↓
Coprococcus sp ART55 1 arginine and polyamine biosynthesis
pyridoxal 5-phosphate biosynthesis ↑
Megamonas spp.
Ruminococcaceae bacterium D16 ↓
Lachnospiraceae bacterium 1 1 57FAA ↓
Transcriptomics (sigmoid colon) NA SULT1C2 and SULT1C3
FOHL1
PDE9A ↑
FOHL1 ↑
UCP2 ↑
HTR3A ↑
Gut immune cell population (sigmoid colon digital cytometry) NA NA CD8 T cells ↑
Activated CD4 T cells
NK cells
Immune population (PBMC flow cytometry) NA Reduced activated memory CD4 and CD8 NA
Cytokine production (PBMC flow cytometry) NA Higher IL-22 production in CD8 and higher TNF-α production in γδ T cells NA

Abbreviation: NA, not applicable.

Although both PSO2 and PSO3 are psoriasis-enriched subgroups, we have observed some differences in host circulatory and intestinal immune profiles between the two subgroups. PSO2 has increased memory CD8+ T-cell population in peripheral blood and higher IL-22 production capacity from CD8+ T cells (Figure 4 and Supplementary Table S9). Although PSO3 has no obvious immune signatures in peripheral blood, PSO3 has higher CD8+ T cells and lower active CD4+ T cells in sigmoidal gut as identified by in silico immunoprofiling. Conventionally, CD8+ T cells are known for their cytotoxic activities and are important for clearing infected cells or cancerous cells. Recent studies have been focusing on cytokine-producing CD8+ T cells because they were found in psoriatic skin to be an important source of proinflammatory IL-17 and IL-22 (Hijnen et al., 2013; Liu et al., 2021). Interestingly, expanded proinflammatory cytokine-producing CD8+ T cells have been found in PBMCs of patients with psoriatic arthritis (Diani et al., 2019). Our findings confirm the presence of cytokine-producing CD8+ T cells in patients with psoriasis and implicate differential roles of CD8+ T cells in different psoriasis subgroups.

Many of the host transcriptomic signatures found in PSO3 are similar to the ones reported in patients with IBD or IBD animal models, including elevated expression of folate hydrolase (FOLH1) (Noble et al., 2010; Rais et al., 2016), one of the serotonin receptors (HTR3A) (Shajib et al., 2019), and mitochondrial UCP2 (Jin et al., 2017; Yu et al., 2009). Elevated FOLH1 expression has been reported in intestinal biopsies of patients with IBD, and inhibiting FOLH1 activity ameliorates IBD-associated abnormalities in mouse models (Noble et al., 2010; Rais et al., 2016). UCP2 encodes for mitochondrial UCP2 and has been implicated in several autoimmune diseases (Yu et al., 2009). Expression of UCP2 is elevated in a dextran sodium sulfate‒induced mouse model of IBD, and the severity of IBD can be ameliorated by knocking down UCP2 expression through an expression by small interfering RNA (Jin et al., 2017). In addition to the IBD-related signatures, patients in PSO3 also have a reduced abundance of Ruminococcaceae and Lachnospiraceae, which is also observed in the gut microbiome of psoriatic arthritis (Scher et al., 2015). It is worth noting that all patients in PSO3 reported having joint pain or swelling, whereas only a fraction of patients in PSO1 or PSO2 did (Supplementary Figure S3h). Both IBD and psoriatic arthritis are common psoriatic comorbidities, and our study suggests an intriguing possibility that these psoriasis subgroups might represent psoriasis populations with differential risk for developing comorbidities such as IBD and psoriatic arthritis.

Our multiomic analysis revealed interesting associations between microbial pathways with circulating IL-17A production and expression of proinflammatory chemokines, CXCL1 and CXCL3, in the colon (Figure 6b). These host‒microbe associations are only observed in patients with psoriasis but not in healthy subjects (Supplementary Figure S4a and b), suggesting that psoriasis-specific host‒microbe associations might be crucial drivers of the proinflammatory changes in patients with psoriasis. In addition to psoriasis-specific host‒microbe associations, our analysis also revealed specific host‒microbe associations in each psoriasis subgroup in our cohort. Together, our data suggest that the host‒microbe interaction can be context dependent, whereby the same microbial species or function may have a different effect on the basis of the disease state or disease subgroup.

We are aware of several limitations of our study that could be improved by future work. Although multidimensional, our analysis is limited by a modest cohort size, so it would be important to validate our findings in a larger independent psoriasis cohort. In addition to validation, a larger cohort will allow for the application of machine learning approaches to better characterize psoriasis subgroups. Owing to the scope of the study, we focused our efforts on cross-sectional observations of our cohort. Host‒microbe interaction can be relatively dynamic over time, and some of the features or associations identified may be relatively transient. Collecting longitudinal data may assist in the identification of stable associations compared with transient associations. In addition, a longitudinal dataset would allow for better tracking of host‒microbe associations associated with the development of comorbidities. In this study, we have identified several microbial species that are differentially abundant in psoriasis gut compared with those in healthy controls. However, their roles in psoriasis and inflammation are not previously well understood. Testing the microbial species identified in this study for their roles in eliciting local and system inflammation can shed some insights into their roles in modulating psoriasis. Even with these limitations, the findings of this study provide valuable insights into the complexity of psoriasis biology that can help us to better identify patients with psoriasis with a higher risk of comorbidities and unique biological pathways. To date, most of the disease-related microbiome studies are still focusing on signatures identification aimed to identify specific microbes or microbial functions that contribute to the pathogenesis of the disease. Our results suggest that pathogenesis of psoriasis and its comorbidities is highly individualized and that both host and microbiome should be considered when assessing disease risks and strategizing disease management in both clinical and scientific settings. The findings of this study are relevant because they may have diagnostic potential for psoriasis comorbidities and may provide insights in developing personalized disease management strategies for patients with psoriasis.

Materials and Methods

Cohort recruitment and sample collection

Adult patients with psoriasis and healthy volunteers recruited from the San Francisco Bay area were enrolled in the study after providing written informed consent. All procedures performed on human subjects were reviewed and approved by Institutional Review Board at the University of California San Francisco (Institutional Review Board protocols 10-02830 and 10-01218). Individuals with abnormal coagulation parameters, positive HIV screening test, and history of bleeding disorders, abdominal surgery, gastrointestinal cancer, IBD, AIDS, immunodeficiency or immunosuppressive medications, or concurrent inflammatory skin condition were excluded. All patients with psoriasis had a diagnosis of psoriasis from a physician for at least 6 months before study enrollment, which was verified by study staff. To assess the psoriatic microbiome in an untreated state, subjects were excluded if they had received systemic biologic therapy in the last 6 months, nonbiologic systemic medications (methotrexate, cyclosporine, corticosteroids, cyclophosphamide, retinoids, and photochemotherapy) or antibiotics in the last month, or phototherapy or topical therapy in the last 2 weeks before the clinical visit. Healthy volunteers had no personal or family history of psoriasis. Gut biopsies, stool samples, and blood samples were collected on the same day. Gut biopsies were taken from the sigmoid colon of each subject. All patients with psoriasis in our cohort had a normal-appearing lower gastrointestinal endoscopic examination. Two biopsies were preserved in RNAlater solution (Ambion, Austin, TX) and kept at 4 °C overnight before storage at ‒80 °C. Stool samples were collected before gut biopsies and stored at ‒80 °C. Blood samples were taken on the same day as and preceded the gut biopsies. PBMCs were isolated from whole blood and stored in liquid nitrogen. We collected a total of 48 stool samples (15 from healthy subjects and 33 from patients with psoriasis) for shotgun metagenomics, and a total of 42 gut biopsies and blood samples (16 from healthy subjects and 26 from patients with psoriasis) were collected for host transcriptomics and immune profiling.

Diet habit analysis

We assessed dietary habits of healthy controls and patients with psoriasis by an in-person distribution of the 2009‒2010 National Health and Nutrition Examination Survey dietary screener questionnaire. The questionnaire consists of 25 questions about nutrient intake frequencies of different food groups. The differences in intake frequencies of each food group were compared between disease status and psoriasis subgroups with Fisher’s exact test, and results with P < 0.05 were considered significant. To further assess the quality of dietary habits, we summarized diet survey by calculating a Mediterranean diet score that would quantify the overall quality of diet. The traditional Mediterranean diet has been shown to reduce the risk of developing heart disease, metabolic syndrome, diabetes, and depression. The foundation for the Mediterranean diet consists of an abundance of fresh fruits, vegetables, whole grains, legumes, and omega-3 fatty acids and limited food such as milk, cheese, red meats, and sweets with added sugars. We adopted Mediterranean score calculation from a previous study (Panagiotakos et al., 2006). Briefly, we used six components of the Mediterranean diet (nonrefined cereals, fruit, vegetables, potatoes, whole grains, and legumes). For the consumption of food groups that aligned with the Mediterranean diet, we assigned scores 0, 1, 2, 3, 4, and 5 when a participant reported no consumption, rare (one time per month), frequent (2‒3 times per /month), very frequent (one time per week), weekly (2‒6 times per week), and daily (1‒2 times per day), respectively. For non-Mediterranean diet food components, we assigned scores on a reverse scale with scores 0, 1, 2, 3, 4, and 5 when a participant reported daily (1‒2 times per day), weekly (2‒6 times per week), very frequent (one time per week), rare (one time per month), and no consumption, respectively. All scores were then combined to produce a total score that ranged from 0 to 50, where the higher the score, the better the diet. To test for statistical significance between both participant groups, we utilized the Wilcox test to compare the means and P-values of our data.

Shotgun metagenomics

DNA was extracted from fecal samples with the modified cetyl trimethylammonium bromide method. The quality of the extracted DNA was assessed by gel electrophoresis and Quant-iT PicoGreen dsDNA Assay (Life Technologies, Carlsbad, CA). The extracted DNA was submitted to Vincent J. Coates Genomic Sequencing Laboratory at the California Institute of Technology for Quantitative Biosciences (San Francisco, CA) (www.qb3.berkeley.edu/gsl) for metagenomic library construction and sequencing. The metagenomic libraries were constructed using PrepX DNA library kit (Takara Bio, Kusatsu, Japan) and sequenced on HiSeq4000 (Illumina, San Diego, CA) for pair-end 150 base pair sequencing. An average of 22 million pair-end total reads were generated per sample. Reads were first mapped to human genome (GRCh38, GENCODE release 25) by bowtie2 (version 2.2.8) to identify human DNA. The nonhuman reads were extracted and sorted by using samtool (version 0.1.19). This data processing workflow results in an average of 16 million nonhuman reads per sample. The resulting nonhuman reads were used for the subsequent taxonomic and functional profiling.

Taxonomic profiles were generated using MetaPhlAn2 (version 2.6.0), which uses a library of ∼1 million unique clade-specific marker genes for taxonomic profiling (Franzosa et al., 2018). The abundance table of bacterial species was extracted for the subsequent analysis. Functional profiles were generated by HUMAnN2 (version 0.11.1), which is an assemble-free method to infer the functional capacities for each microbiome (Franzosa et al., 2018). For each sample, HUMAnN2 maps read the pangenomes of species identified in the sample and performed additional translated searches on unclassified reads. The resulting dataset is the abundance table of microbial gene families (UniRef90), which is summarized into the higher-level MetaCyc gene pathways. Our analysis identified a total of 339 bacterial species, 409 MetaCyc pathways, and 650,048 UniRef90 gene families in our dataset.

Subsequent metagenomic analyses were performed using the R package phyloseq (McMurdie and Holmes, 2013). The abundance of microbial species, UniRef90 gene families, and MetaCyc pathway was normalized to relative abundance to account for different library sizes and input as phyloseq objects and used for calculating microbial community diversities. Owing to the high dimensionality and high dropout rate of the microbial gene families table, we focus our analysis on the microbial gene families with at least three counts for at least 80% of the total samples, which results in 3,356 UniRef90 gene families. Alpha diversity was calculated as four indices: observed (number of observed microbial features), chao1, Shannon, and Simpson index, which summarize both richness and evenness of the community. The alpha diversities were compared by disease status and psoriasis subgroups with two-sided Wilcox test. Dissimilarities between each microbiome isolated are represented by Bray‒Curtis dissimilarity matrix and visualized on a principal coordinate analysis plot.

Unfiltered counts per million tables were used for differential abundance analysis. Differential abundant microbial features between psoriasis samples and healthy samples were identified using DESeq2 (version 1.18.1) (Love et al., 2014) with the design ⋍sex+age.bin+batch+Status. Microbial features with adjusted P < 0.05 and absolute log2 fold change > 0.6 are considered statistically significant. Differential abundant microbial features associated with each subgroup identified in this cohort were identified by pairwise comparison of each subgroup using DESeq2 (version 1.18.1) (Love et al., 2014). Microbial features with adjusted P < 0.1 and absolute log2 fold change > 0.6 are considered statistically significant. Differential abundance microbial features with none zero counts for at least 10 samples were plotted on the heatmap to exclude features with high dropout rates.

Clustering analysis to identify subgroups in the cohort

The subgroups in this cohort were identified by complete linkage hierarchical clustering on the differentially abundant microbial UniRef90 gene families identified as described earlier. The clustering was done using the R package pheatmap (version 1.0.12), which implements hclust for hierarchical clustering, or the gapstat_ord function in phyloseq (version 1.22.3). Gap statistics on differentially abundant UniRef90 gene families estimated three clusters present in our dataset. Gap statistics were done using the clusGap function from the cluster package (version 2.0.7-1) with bootstrapping 1,000 times (B = 1,000). The optimal number of clusters was determined as the smallest k that is within one standard error from the local maximal as suggested in Tibshirani et. al. (2001). The hierarchical dendrogram was cut by cutree function for three clusters (k = 3) in stats package (version 3.4.2) to assign membership of clusters for each sample.

Host RNA-seq

RNA was extracted from sigmoidal colon biopsies by RNeasy Plus Universal Kits (Qiagen, Hilden, Germany). Total RNA integrity was assessed using Agilent 2100 Nano and Pico RNA kit, and all samples have RNA integrity number score >7. cDNA libraries construction and sequencing were done by Beijing Genomics Institute (Shenzhen, China). In brief, ribosomal RNA was removed from the total RNA by Ribo-Zero Gold Kit (Illumina), and library was constructed by TruSeq Stranded Total RNA Library Prep kit (Illumina). cDNA libraries were sequenced on HiSeq4000 (Illumina) for paired-end 100 base pair sequencing. The average sequencing depth is 66 million per sample. Sequencing reads were mapped to the human genome (GRCh38, GENCODE release 25) using STAR (version 2.4.2a). The number of counts mapped to each transcript is summarized by HTSeq-count (version 0.6.0) and used as input for differential analysis in DESeq2 (version 1.18.1) (Love et al., 2014). Raw read counts were normalized by median of ratios method using DESeq2. Differentially expressed genes between psoriasis samples and healthy samples were determined by DESeq2 with the model design ∼sex+age+batch+status. Genes with adjusted P < 0.05 and absolute log2 fold change > 0.6 are considered significantly different between psoriasis samples and healthy samples. A similar analysis was performed to identify differentially expressed genes between different subgroups (genecluster.group) in this cohort with the DESeq2 model design ∼sex+age+batch+genecluster.group. Results of pairwise comparisons were extracted, and significant differentially expressed genes were determined with the same significant criteria. The expression of the top 5,000 most variable genes was transformed by variance stabilizing transformation for principal component analysis.

In silico cytometry methods

The colon immune cell composition was imputed from the sigmoid colon bulk RNA-seq data by CIBERSORTx (https://cibersortx.stanford.edu/) (Newman et al., 2019). In brief, single-cell RNA-seq dataset associated with sigmoid colon was extracted from colon immune atlas of Gut Cell Atlas project by Seurat (James et al., 2020). The first part of the CIBERSORTx workflow computed the signature matrix of immune cell populations in the sigmoid colon using the sigmoid colon single-cell RNA-seq dataset. The second part of the CIBERSORTx workflow imputed cell fractions in the sigmoid colon by comparing bulk sigmoid colon RNA-seq with sigmoid colon immune cell signature matrix with the following parameters: relative run mode, 100 permutations, disabled quantile normalization, and S-mode batch correction, which was implemented to account for cross-platform deconvolution. The cell compositions were compared by disease status and psoriasis subgroups with two-sided Wilcoxon test or Kruskal‒Wallis test, and results with P < 0.05 were considered significant. Statistical analyses were performed with R.

Multiomic analysis

We integrated six different datasets collected from three different measurement types: shotgun metagenomic sequencing, flow cytometry, and host RNA-seq from 14 healthy subjects and 26 patients with psoriasis. We included the following datasets for data integration: (i) bacterial species from shotgun metagenomics with nonzero count in at least 10 samples, (ii) microbial MetaCyc pathway from shotgun metagenomics with nonzero count in at least 10 samples, (iii) microbial UniRef90 gene families from shotgun metagenomics with at least three counts for 80% of the total samples with metagenomic data, (iv) cell population from flow cytometry, (v) cytokine production capacity from flow cytometry, and (vi) top 500 most variable genes plus all differentially expressed genes identified by DEseq2 from host RNA-seq. More information about datasets included in the multiomic analysis is summarized in Supplementary Table S10. Pairwise datasets from different measurement types were integrated by Spearman’s rank-order correlation. We constructed interaction networks with only strong and robust correlations that fit the following criteria: correlations need to have a false discovery rate‒adjusted P < 0.1 and absolute correlation coefficient > 0.6. We required microbial or host features with a nonzero count for >70% of samples within the group of interest. Multiomic analyses were done within all subjects (both healthy subjects and subjects with psoriasis), within healthy subjects alone, within subjects with psoriasis alone, and within each psoriasis subgroups. The significant host‒microbe associations identified in our analyses are listed in Supplementary Table S11. The multiomic networks were visualized using the R package, igraph (version 1.2.4.1) The community modules within each network were identified using fast greedy modularity optimization algorithm (Clauset et al., 20041).

Host flow cytometry

Flow cytometry for cell population

Frozen PBMCs were thawed and counted, and one million cells were plated in a 96-well V-bottom plate. The cells were centrifuged for 5 minutes at 400g and surfaced stained with a predetermined antibody staining cocktail for 15 minutes at room temperature. The cells were washed with sort buffer, centrifuged, resuspended in BD Perm/Wash Buffer (BD Biosciences, Franklin Lakes, NJ), and centrifuged for 5 minutes at 700g. The supernatant was aspirated, and the cells were stained for FoxP3 by adding diluted fluorophore-labeled anti-human FoxP3 antibody in BD Perm/Wash Buffer for 30 minutes at room temperature. Cells were centrifuged for 5 minutes at 700g, the supernatant was aspirated, and the wash was repeated with BD Perm/Wash buffer. The cells were resuspended a final time in 50 ul of PBS and held at 4 ºC until analyzed on a BD LSR-II instrument (BD Biosciences). Brilliant Violet Buffer was added to each staining step. Fluorescent-minus-one controls were used for setting gates. A minimum of 300,000 events were analyzed for each sample. The gating strategy is provided in Supplementary Figure S5, and the antibodies used are listed in Table 5.

Supplementary Figure S5.

Supplementary Figure S5

Supplementary Figure S5

Representative flow cytometry gating for identifying various cell populations. (a) Cell populations (CD4+ Teff, CD4+ Treg, CD8+ T cell, γδ T cell, and innate lymphoid cells) and (b) their memory and activation state. APC, allophycocyanin; FSC-A, forward scatter area; FSC-H, forward scatter height; K, thousand; PE, phycoerythrin; Q, quadrant; SSC-A, side scatter area; Teff, effector T cell; Treg, regulatory T cell.

Table 5.

List of the Antibodies Used for the Flow Cytometry

Marker Antibody Cell Type
Phenotype panel
AARD AARD Viability
CD45 CD45-APCH7 Lymphocytes
CD3 CD3-PerCpCy55 T cells
CD4 CD4-APC CD4+ T cells
CD8 CD8-BV650 CD8+ T cells
FoxP3 FoxP3-A488 Treg
CD25 CD25-PECF594 Treg
TCRgd TCRgd-BV711 γδ T cells
HLADR HLADR-BV421 Activation
CD38 CD38-PE Activation
CD117 CD117-BV786 ILCs
NKp44 NKp44-BV605 ILC3
CD45RO CD45RO-PECy7 Memory
Functional panel
AARD AARD Viability
CD45 CD45-APC-Cy7 Lymphocytes
CD3 CD3-PerCpCy55 T cells
CD4 CD4-APC CD4+ T cells
CD8 CD8-BV650 CD8+ T cells
FoxP3 FoxP3-A488 Treg
CD127 CD127-PE-Dazzle594 Low in Tregs
TCRVS2 TCRVS2-BV711 Vd2 T cells
NKp44 NKp44-BV605 ILC3
CD117 CD117-BV786 ILC3
TNF-α TNFa-A700 Cytokine
IFNγ IFNg-PE-Cy7 Cytokine
IL-17 IL17-BV421 Cytokine
IL-22 IL22-PE Cytokine

Abbreviations: APC, allophycocyanin; PE, phycoerythrin; Treg, regulatory T cell.

Flow cytometry for cytokine production

Frozen PBMCs were thawed and counted and one million cells were plated in a 96-well U-bottom plate. PBMCs were left unstimulated in complete culture media (RPMI + 10% heat-inactivated fetal bovine serum supplemented with penicillin/streptomycin/L-glutamine) or stimulated with 5 ng/ml of phorbol myristate acetate and 0.5 ug/ml of ionomycin for 4 hours in the presence of brefeldin A (10 ug/ml). After 4 hours of stimulation, the cells were centrifuged and stained with an antibody staining cocktail listed for 15 minutes at room temperature. The cells were washed with sort buffer, centrifuged, and resuspended in BD Perm/Wash Buffer and centrifuged for 5 minutes at 700g. The supernatant was aspirated, and the cells were stained for intracellular antigens by adding diluted fluorophore-labeled antibodies in BD Perm/Wash Buffer for 30 minutes at room temperature. Cells were centrifuged for 5 minutes at 700g, the supernatant was aspirated, and the wash was repeated with BD Perm/Wash buffer. The cells were resuspended a final time in 50 ul of PBS and held at 4 ºC until analyzed on a BD LSR-II instrument. Brilliant Violet Buffer was added to each staining step. Fluorescent-minus-one controls were used for setting gates. A minimum of 300,000 events were analyzed for each sample. Reported data represent the stimulated cytokine expression minus the unstimulated cytokine expression referred to as background corrected cytokine expression. fcs files were analyzed by FlowJo (version 10; Tree Star, Ashland, OR), and the immune profiles were exported as txt files, which are imported into R for statistical analysis. Wilcox test was used to assess the significance of the differences between disease status or disease subgroups. The gating strategy is provided in Supplementary Figure S6, and the antibodies used are listed in Table 5.

Supplementary Figure S6.

Supplementary Figure S6

Supplementary Figure S6

Representative flow cytometry gating for measuring cytokine production in various cell populations after 4 hours of PMA and ionomycin stimulation. (a) Gating strategies for identification of Teff, Treg, CD8+ T cells, and γδ T cells. (b) Gating strategies for measuring the production of IL-17A, IL-22, TNF-α, and IFNγ. APC, allophycocyanin; FSC-A, forward scatter area; FSC-H, forward scatter height; K, thousand; PMA, phorbol myristate acetate; Teff, effector T cell; Treg, regulatory T cell; vs. versus.

Data availability statement

Expression data have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus and is accessible through Gene Expression Omnibus Series accession number GSE150851. Metagenomics sequence data are accessible through Sequence Read Archive BioProject PRJNA634145. All clinical and experimental data available are shown in Supplementary Table S12.

ORCIDs

Hsin-Wen Chang: http://orcid.org/0000-0003-2881-245X

Di Yan: http://orcid.org/0000-0003-1338-9847

Rasnik Singh: http://orcid.org/0000-0002-7372-6692

Audrey Bui: http://orcid.org/0000-0003-3055-463X

Kristina Lee: http://orcid.org/0000-0002-5879-817X

Alexa Truong: http://orcid.org/0000-0002-4748-5581

Jeffrey M. Milush: http://orcid.org/0000-0002-0773-6411

Ma Somsouk: http://orcid.org/0000-0001-6116-3645

Wilson Liao: http://orcid.org/0000-0001-7883-6439

Author Contributions

Conceptualization: WL, HWC; Methodology (Colonic Biopsies Collection): MS; Formal Analysis (Diet): AT; Flow Cytometry Data Analysis: JMM, HWC; Methodology (Flow Cytometry): JMM; Methodology (In Silico Cytometry): AB; Methodology: WL, HWC, JMM, MS; Methodology (Patient Recruitment): DY, RS, KL; Supervision: WL; Formal Analysis (Bioinformatics): HWC; Writing - Original Draft Preparation: WL, HWC

Acknowledgments

Metagenomic sequencing was performed by the Vincent J. Coates Genomics Sequencing Laboratory at the University of California Berkeley (Berkeley, CA), supported by the National Institutes of Health S10 OD018174 Instrumentation Grant. Flow cytometry was performed by the San Francisco General Hospital Flow Core Facility, supported by a grant from the National Institutes of Health, University of California San Francisco-Gladstone Institute of Virology and Immunology Center for AIDS Research (P30 AI027763). This work was performed at the University of California San Francisco (San Francisco, CA). This study was supported in part by a National Psoriasis Foundation Translational Research Award and National Institutes of Health grants to WL (R01AR065174 and U01AI119125) and a National Psoriasis Foundation Medical Research Fellowship to DY.

Conflict of Interest

WL has received research grant funding from AbbVie, Amgen, Janssen, Leo, Novartis, Pfizer, Regeneron, and TRex Bio. The remaining authors state no conflict of interest.

accepted manuscript published online 10 March 2022; corrected proof published online 12 May 2022

Footnotes

Cite this article as: JID Innovations 2022;X:100115

1

Clauset A, Newman MEJ, Moore C. Finding community structure in very large networks. arXiv 2004.

Supplementary material is linked to the online version of the paper at www.jidonline.org, and at https://doi.org/10.1016/j.xjidi.2022.100115.

Supplementary Materials

Supplementary Table S1.

List of Microbial Species that Are Differentially Abundant between Psoriasis and Healthy Samples

Microbial Species Base Mean Log2 Fold Change P-Value Padj
Prevotella_disiens 12.48 26.94 5.07382E–16 2.08027E–14
Clostridium_sp_KLE_1755 95.27 26.20 3.98903E–15 9.08612E–14
Bacteroides_coprocola 1,787.37 22.57 1.30691E–11 1.33958E–10
Erysipelotrichaceae_bacterium_5_2_54FAA 4.22 21.86 5.56657E–11 5.18704E–10
Dorea_unclassified 2.09 20.75 4.90663E–10 3.59236E–09
Prevotella_buccae 2.70 18.20 4.78393E–08 3.26902E–07
Brachyspira_unclassified 0.73 16.74 5.16488E–07 3.30875E–06
Haemophilus_pittmaniae 4.37 16.29 9.98499E–07 6.02036E–06
Gemella_sanguinis 3.32 15.09 5.99289E–06 3.41262E–05
Roseburia_unclassified 1,416.55 14.32 1.53925E–05 8.09092E–05
Bacteroides_oleiciplenus 12.19 13.46 5.31623E–05 0.000272457
Burkholderiales_bacterium_1_1_47 579.13 12.17 3.16928E–08 2.24035E–07
Weissella_unclassified 1.08 11.51 0.000557326 0.002393849
Lachnospiraceae_bacterium_9_1_43BFAA 56.00 10.31 0.001862232 0.007485442
Clostridium_hylemonae 3.72 10.17 0.002285859 0.008841529
Anaerotruncus_unclassified 7.27 9.86 0.003047001 0.011567319
Megasphaera_unclassified 3,017.86 9.60 0.001021066 0.004186369
Collinsella_stercoris 11.98 9.54 0.004187753 0.015061219
Enterococcus_faecium 291.35 9.24 0.003988014 0.014598981
Dialister_invisus 11,846.88 9.16 0.00038163 0.001738535
Eubacterium_dolichum 126.53 8.43 0.000771769 0.003228827
Parabacteroides_goldsteinii 179.97 6.59 0.010261261 0.035346823
Parasutterella_excrementihominis 232.32 4.89 0.006925784 0.024479064
Sutterella_wadsworthensis 2,783.26 4.82 0.011048633 0.037130653
Bacteroides_vulgatus 16,890.23 2.08 0.003156479 0.01176506
Clostridiaceae_bacterium_JC118 68.59 ‒5.10 0.010345412 0.035346823
Phascolarctobacterium_succinatutens 16,877.87 ‒7.49 0.012182033 0.040279303
Bifidobacterium_pseudocatenulatum 843.21 ‒8.15 0.000560511 0.002393849
Peptostreptococcaceae_noname_unclassified 407.41 ‒8.64 6.40326E–05 0.00031254
Streptococcus_mitis_oralis_pneumoniae 31.93 ‒9.77 1.50858E–05 8.09092E–05
Atopobium_parvulum 1.79 ‒10.15 0.002104515 0.008296648
Coprococcus_sp_ART55_1 17,348.22 ‒11.50 0.000206357 0.000961438
Lactobacillus_delbrueckii 5.36 ‒11.56 0.000500643 0.002231124
Mitsuokella_multacida 247.94 ‒12.11 9.84218E–05 0.00046922
Fusobacterium_ulcerans 188.80 ‒13.32 6.19636E–05 0.000309818
Mitsuokella_unclassified 1,185.04 ‒14.76 1.95683E–06 1.14614E–05
Leuconostoc_lactis 12.28 ‒14.80 8.64274E–06 4.78854E–05
Gemella_unclassified 1.35 ‒16.59 6.05617E–07 3.76217E–06
Desulfovibrio_desulfuricans 43.84 ‒17.53 1.0359E–07 6.85032E–07
Proteus_penneri 0.22 ‒20.93 3.3238E–10 2.52363E–09
Citrobacter_freundii 1.54 ‒21.36 1.38522E–10 1.09219E–09
Leuconostoc_gelidum 0.28 ‒21.40 1.32504E–10 1.08653E–09
Proteus_unclassified 0.29 ‒21.44 1.22431E–10 1.04577E–09
Streptococcus_anginosus 6.60 ‒21.57 9.26092E–11 8.2543E–10
Porphyromonas_somerae 4.71 ‒21.89 5.18674E–11 5.06325E–10
Lactobacillus_curvatus 13.67 ‒22.92 5.84E–12 6.30105E–11
Alloscardovia_omnicolens 2.16 ‒23.22 3.21802E–12 3.66497E–11
Lactobacillus_sakei 1.23 ‒23.34 2.45901E–12 2.96528E–11
Streptococcus_sp_BS35b 2.20 ‒23.89 7.47425E–13 9.57638E–12
Enterococcus_avium 2.11 ‒24.12 4.44676E–13 6.07724E–12
Bacteroides_gallinarum 16.02 ‒24.34 2.59664E–13 3.80222E–12
Carnobacterium_maltaromaticum 12.49 ‒24.74 1.15388E–13 1.81958E–12
candidate_division_TM7_single_cell_isolate_TM7c 0.18 ‒25.12 4.69133E–14 8.01436E–13
Weissella_cibaria 6.65 ‒25.57 1.65562E–14 3.08547E–13
Subdoligranulum_variabile 28.34 ‒25.57 1.27799E–14 2.61988E–13
Bacteroides_sp_1_1_6 133.01 ‒26.24 3.4666E–15 8.88316E–14
Citrobacter_unclassified 4.40 ‒26.39 1.9069E–15 5.58448E–14
Butyricicoccus_pullicaecorum 12.00 ‒26.47 1.86029E–15 5.58448E–14
Bacteroides_sp_2_1_22 425.75 ‒29.27 1.42946E–18 7.326E–17
Corynebacterium_glutamicum 1.95 ‒29.98 2.24401E–19 1.54839E–17
Ruminococcus_champanellensis 1,472.34 ‒30.00 2.26594E–19 1.54839E–17
Campylobacter_hominis 1.51 ‒30.00 2.09163E–19 1.54839E–17

Abbreviation: adj, adjusted.

Supplementary Table S2.

List of Microbial Gene Families that Are Differentially Abundant between Psoriasis and Healthy Samples

UniRef90_Genefamilies Base Mean Log2 Fold Change P-Value Padj
UniRef90_D4JKB5: NO_NAME 20.44971008 8.184774568 3.76E–07 3.33E–05
UniRef90_D4JKB4: NO_NAME 9.737750875 7.318193992 2.7E–06 0.000218
UniRef90_D4JKB3: NO_NAME 5.604615949 5.972567491 2.98E–06 0.000239
UniRef90_C6JHT6: NO_NAME 11.59795247 3.503976143 3.74E–06 0.000297
UniRef90_V8CCN6: NO_NAME 7.911723223 ‒8.466033916 2.02E–05 0.001506
UniRef90_R5HHX1: NO_NAME 3.291298903 5.617246745 2.23E–05 0.001652
UniRef90_B0MUZ9: NO_NAME 17.2243469 7.779663835 2.63E–05 0.001914
UniRef90_B0NKZ9: NO_NAME 4.866770601 4.68406674 0.000104 0.007173
UniRef90_E1YTP4: NO_NAME 9.980238914 4.364093512 0.000124 0.008475
UniRef90_C0EYV3: NO_NAME 5.843250501 ‒1.156773775 0.00013 0.008856
UniRef90_K9DSD1: NO_NAME 20.70420605 7.4409631 0.000134 0.009178
UniRef90_D4V9S6: Toxin-antitoxin system, antitoxin component family protein 3.433982731 3.624603291 0.000137 0.009363
UniRef90_T4Z7D4: NO_NAME 12.26511689 ‒1.374370748 0.00014 0.009561
UniRef90_A7A642: NO_NAME 9.61390528 ‒3.999917984 0.00018 0.012157
UniRef90_C4Z8L3: NO_NAME 8.953334146 4.14025387 0.000198 0.013365
UniRef90_D4L5Q7: Dockerin type I repeat 3.709257355 4.458714365 0.000214 0.014395
UniRef90_C4ZDT0: NO_NAME 11.02944175 2.61803876 0.000221 0.014875
UniRef90_Q89YV2: NO_NAME 3.960842604 2.459102717 0.000231 0.015496
UniRef90_R5ENA9: NO_NAME 7.31968454 2.809734777 0.000273 0.018301
UniRef90_R5VN00: NO_NAME 6.66944193 2.726824008 0.000283 0.018902
UniRef90_C0FYT5: NO_NAME 5.608127022 5.494622505 0.000289 0.019302
UniRef90_U2DKA0: Toxin-antitoxin system, antitoxin component, Xre family 2.41752419 ‒2.07552754 0.000315 0.021031
UniRef90_U2NXE6: NO_NAME 0.823846701 4.956194737 0.000317 0.0212
UniRef90_R5F076: NO_NAME 6.570912784 2.6,53219048 0.000329 0.021951
UniRef90_I7ASX7: Aminoglycoside 6-adenylyltransferase 5.981679914 ‒1.229156672 0.000339 0.022617
UniRef90_D6E2K8: NO_NAME 6.220313047 7.882367351 0.00035 0.023302
UniRef90_C4ZCN8: NO_NAME 4.739971235 4.531804983 0.000367 0.02438
UniRef90_A8SDV6: NO_NAME 3.111716292 ‒6.463421902 0.00037 0.024535
UniRef90_A7B7Y7: NO_NAME 3.835647149 ‒4.216283036 0.000381 0.02528
UniRef90_R5DHM8: 50S ribosomal protein L14 8.05231442 ‒1.091553729 0.000387 0.025643
UniRef90_F7JRF6: NO_NAME 2.10652993 ‒2.258144862 0.000397 0.026326
UniRef90_R5ENA5: NO_NAME 6.145444386 2.76594925 0.000404 0.026733
UniRef90_C4ZB30: NO_NAME 13.98634953 6.26094269 0.000416 0.027527
UniRef90_C4ZHW5: NO_NAME 7.601796234 2.21832247 0.000424 0.028048
UniRef90_A6NXS5: NO_NAME 3.085119571 ‒1.949411921 0.000431 0.028513
UniRef90_R5ES70: NO_NAME 6.142418982 2.70370766 0.000447 0.029252
UniRef90_R5ENC3: NO_NAME 6.187279035 2.701041839 0.00045 0.029276
UniRef90_R5VTV0: NO_NAME 11.84385896 ‒8.859754987 0.000484 0.031431
UniRef90_R5ES82: NO_NAME 6.218044543 2.631291787 0.000495 0.03205
UniRef90_A8SDV1: PTS system, lactose/cellobiose family IIC component 1.577472175 ‒5.394020156 0.000498 0.032135
UniRef90_R5F071: NO_NAME 5.795869956 2.671859836 0.0005 0.032226
UniRef90_R5EN96: NO_NAME 6.316661771 2.597760365 0.000522 0.033606
UniRef90_D4LVD7: Bacterial mobilisation protein (MobC) 2.997706936 ‒3.203584633 0.000527 0.033927
UniRef90_R5EP09: NO_NAME 6.497763827 2.664311777 0.000545 0.035039
UniRef90_A8SDV2: PTS system, Lactose/Cellobiose specific IIB subunit 1.67041892 ‒5.420005822 0.000571 0.036645
UniRef90_R5ENY1: NO_NAME 6.055002091 2.605312597 0.000597 0.038285
UniRef90_R5EXU5: NO_NAME 6.640770231 2.590729061 0.000608 0.039021
UniRef90_R5B346: Peptidyl-prolyl cis-trans isomerase 1.333716976 ‒2.172687536 0.000617 0.039563
UniRef90_R5F1R6: NO_NAME 6.195974482 2.670392787 0.00062 0.039746
UniRef90_R5ENZ3: NO_NAME 5.847885346 2.536486311 0.000661 0.042259
UniRef90_R5F1S4: NO_NAME 6.301820127 2.594059152 0.000673 0.043049
UniRef90_F9Z9M1: Putative transposase 3.863722566 2.56148742 0.000689 0.044002
UniRef90_R5F5V1: NO_NAME 6.214726081 2.736286892 0.000691 0.044147
UniRef90_C0B519: NO_NAME 2.42564728 ‒2.370035967 0.000713 0.045513
UniRef90_J9GFK7: Transposase 10.34428213 3.257865985 0.000716 0.045681
UniRef90_R5F1T5: NO_NAME 7.6286111 2.573141893 0.000747 0.047598
UniRef90_R5ES58: NO_NAME 6.279915456 2.537809692 0.000753 0.047953
UniRef90_R5END3: NO_NAME 6.459389346 2.620179025 0.000759 0.048362
UniRef90_C4ZAI7: Putative regulatory components of sensory transduction system 8.358269459 1.883595829 0.000763 0.04859
UniRef90_R5ENB7: NO_NAME 6.194816868 2.563624339 0.000778 0.049511

Abbreviation: adj, adjusted.

Supplementary Table S3.

List of Microbial Metacyc Pathways that Are Differentially Abundant between Psoriasis and Healthy Samples

MetaCyc_Pathway Base Mean Log2 Fold Change P-Value Padj
P163-PWY: L-lysine fermentation to acetate and butanoate 0.174625879 ‒17.84752508 8.47E–08 1.63E–05
PWY-5304: superpathway of sulfur oxidation (Acidianus ambivalens) 0.135640038 ‒18.83320162 1.60E–08 6.16E–06
PWY-7200: superpathway of pyrimidine deoxyribonucleoside salvage 0.627023649 12.74062955 6.12E–05 0.007856

Abbreviation: adj, adjusted.

Supplementary Table S4.

Summary of Statistics of Diet Survey

Diet P-Value (Disease Status) P-Value (Enterotype)
Milk 0.76 0.88
Fruit 0.66 0.16
Cereal 0.12 0.93
Soda 0.51 0.52
Pure fruit juices 0.87 0.86
Sweetened tea or coffee 0.25 0.49
Sweetened fruit drinks 0.6 0.87
Salad 0.44 0.48
Fried potatoes 0.1 0.07
Other kinds of potatoes 0.0564 0.09
Beans 0.29 0.18
Brown rice 0.64 0.67
Other veggies 0.48 0.28
Salsa 0.11 0.0958
Pizza 0.38 0.69
Tomato sauces 0.68 0.12
Cheese 0.47 0.46
Red meat 0.65 0.65
Processed meat 0.13 0.28
Whole grain bread 0.74 0.1
Chocolate/candy 0.84 0.62
Doughnuts 0.46 0.33
Pastries 0.5 0.07
Frozen desserts 0.79 0.23
Popcorn 0.52 0.75

Supplementary Table S5.

List of Microbial Species that Are Differentially Abundant between Different Subgroup in the Cohort

Microbial Species Base Mean Log2 Fold Change P-Value Padj Comparison Pair
Turicibacter unclassified 148.1650263 ‒25.09944303 1.48E–25 2.77E–23 c2c1
Coprococcus sp ART55 1 17,348.22439 ‒30 4.07E–22 3.80E–20 c2c1
Phascolarctobacterium succinatutens 16,877.86806 ‒30 5.75E–20 3.58E–18 c2c1
Turicibacter sanguinis 30.89947547 ‒23.71828011 1.25E–19 5.84E–18 c2c1
Coprococcus sp ART55 1 17,348.22439 ‒32.10303282 4.81E–18 9.82E–16 c2c3
P. succinatutens 16,877.86806 ‒27.87316104 1.19E–12 8.08E–11 c2c3
Turicibacter unclassified 148.1650263 ‒20.02159204 2.65E–12 1.35E–10 c2c3
Megamonas unclassified 13,738.87404 ‒10.70556321 1.26E–06 1.03E–05 c2c1
Megamonas unclassified 13,738.87404 ‒12.64875264 1.93E–06 2.81E–05 c2c3
T. sanguinis 30.89947547 ‒15.01564462 2.39E–06 3.25E–05 c2c3
Ruminococcaceae bacterium D16 318.9654536 ‒8.1375798 0.000312498 0.003363951 c3c1
T. sanguinis 30.89947547 ‒8.702635493 0.000532915 0.005417965 c3c1
Lachnospiraceae bacterium 1 1 57FAA 2,253.140867 ‒6.756348377 0.000705118 0.006791396 c3c1
Streptococcus thermophilus 2,905.078536 ‒4.197322843 0.002619562 0.020410751 c2c1
Prevotella copri 69,851.06368 ‒4.796452699 0.003805748 0.028466996 c2c1
S. thermophilus 2,905.078536 ‒5.017689601 0.002810256 0.035830762 c2c3
Bacteroides xylanisolvens 1,773.658642 3.277724558 0.005561142 0.039997442 c2c1

Abbreviation: adj, adjusted.

Supplementary Table S6.

List of Microbial Gene Families that Are Differentially Abundant between Different Subgroups in the Cohort

UniRef90_Genefamilies Base Mean Log2 Fold Change P-Value Padj Comparison Pair
UniRef90_R5F5T8: NO_NAME 6.13241864 3.855214514 8.38E–09 9.43E–06 c2c3
UniRef90_R5F5U3: NO_NAME 7.146133182 3.742380578 6.57E–09 7.87E–06 c2c3
UniRef90_R5F5U7: NO_NAME 5.872476462 3.736858351 3.22E–09 5.40E–06 c2c3
UniRef90_R5F5V1: NO_NAME 6.214726081 3.729014981 1.70E–07 8.76E–05 c2c3
UniRef90_R5F5V6: NO_NAME 6.843317692 3.810172235 7.23E–08 4.24E–05 c2c3
UniRef90_R5F6I7: NO_NAME 5.66902435 3.508892329 1.28E–08 1.30E–05 c2c3
UniRef90_R5F7E6: NO_NAME 7.654869731 3.781212969 8.36E–08 4.80E–05 c2c3
UniRef90_R5F7E9: NO_NAME 6.014946714 3.700272305 1.07E–07 5.95E–05 c2c3
UniRef90_R5F7F4: NO_NAME 6.252090198 3.70429003 2.70E–09 4.97E–06 c2c3
UniRef90_R5F7F6: NO_NAME 7.03037544 3.73575232 6.86E–08 4.14E–05 c2c3
UniRef90_R5F826: NO_NAME 6.25853359 3.553654489 5.32E–06 0.002125 c2c3
UniRef90_R5F830: NO_NAME 4.020772359 3.303446928 0.000126 0.041501 c2c3
UniRef90_R6BXA1: Pyridoxal biosynthesis lyase PdxS 18.20827345 –4.738434723 0.000139 0.025007 c2c1
UniRef90_R6EYN1: Phosphoenolpyruvate carboxykinase [ATP] 12.14532041 ‒5.300791062 0.000284 0.045255 c2c1
UniRef90_R5UHP6: NO_NAME 7.404456209 –2.502048554 0.000151 0.048523 c2c3
UniRef90_R5V1S5: NO_NAME 6.469822898 3.674332424 3.90E–08 2.69E–05 c2c3
UniRef90_R5VEH7: NO_NAME 5.638179874 3.783296451 3.44E–08 2.46E–05 c2c3
UniRef90_R5VN00: NO_NAME 6.66944193 3.804730154 9.58E–10 2.11E–06 c2c3
UniRef90_R6FSV7: dTDP-4-dehydrorhamnose 3 5-epimerase 3.836852934 ‒4.712602303 0.00015 0.026547 c2c1
UniRef90_UPI00046F6872: NO_NAME 3.841073774 ‒3.288653597 7.37E–05 0.025081 c2c3
UniRef90_R6YLJ5: dTDP-glucose 4 6-dehydratase 25.36463194 ‒5.501203549 4.07E–05 0.008208 c2c1
UniRef90_R6YTB4: Virulence protein 6.475252167 3.598839048 1.42E–11 5.13E–08 c3c1
UniRef90_R7ERI0: Protein phosphatase 2C 3.917056058 2.488315558 0.000229 0.03792 c2c1
UniRef90_W1I1K8: Uncultured bacterium extrachromosomal DNA RGI00018 44.18955643 ‒8.421153783 7.98E–05 0.027004 c2c3
UniRef90_W1I4A6: Uncultured bacterium extrachromosomal DNA RGI00018 19.44468222 ‒5.622500646 0.000293 0.046458 c2c1

Abbreviation: adj, adjusted.

Supplementary Table S7.

List of Microbial Metacyc Pathways that Are Differentially Abundant between Different Subgroups in the Cohort

MetaCyc_Pathway Base Mean Log2 Fold Change P-Value Padj Comparison Pair
NAD-BIOSYNTHESIS-II: NAD salvage pathway II 6.885975523 ‒2.032470385 0.004596 0.081003 c2c1
POLYAMINSYN3-PWY: super pathway of polyamine biosynthesis II 16.58305195 ‒2.61298583 0.002204 0.069068 c2c1
PWY-5121: superpathway of geranylgeranyl diphosphate biosynthesis II (via MEP) 39.92534701 ‒1.094952419 0.003983 0.081003 c2c1
PWY-5384: sucrose degradation IV (sucrose phosphorylase) 6.824519225 ‒3.287493629 0.0009 0.061079 c2c1
PWY-6147: 6-hydroxymethyl-dihydropterin diphosphate biosynthesis I 54.32244565 ‒0.893067591 0.002654 0.074673 c2c1
PWY-6167: flavin biosynthesis II (archaea) 5.972757342 ‒5.596005317 0.005971 0.098575 c2c1
PWY-6270: isoprene biosynthesis I 88.15679759 ‒1.066870021 0.006642 0.098575 c2c1
PWY-6859: all-trans-farnesol biosynthesis 4.012058217 ‒3.717715695 0.001083 0.061079 c2c1
PWY-7392: taxadiene biosynthesis (engineered) 9.386270018 ‒3.60306079 0.000283 0.042061 c2c1
PWY-7560: methylerythritol phosphate pathway II 79.44418324 ‒1.069297573 0.007032 0.099156 c2c1
ANAEROFRUCAT-PWY: homolactic fermentation 143.8794075 0.779635572 0.0046 0.097618 c2c3
COLANSYN-PWY: colanic acid building blocks biosynthesis 35.8384929 1.550221291 0.0044 0.097618 c2c3
HEXITOLDEGSUPER-PWY: superpathway of hexitol degradation (bacteria) 54.54908312 0.909153433 0.002063 0.066483 c2c3
OANTIGEN-PWY: O-antigen building blocks biosynthesis (E. coli) 99.70191306 ‒1.121948089 0.006771 0.097618 c2c3
PHOSLIPSYN-PWY: superpathway of phospholipid biosynthesis I (bacteria) 70.46008483 1.331355977 0.001855 0.066483 c2c3
PWY4FS-7: phosphatidylglycerol biosynthesis I (plastidic) 51.5490991 1.391404667 0.006811 0.097618 c2c3
PWY-5101: L-isoleucine biosynthesis II 66.49625492 1.214578922 0.005292 0.097618 c2c3
PWY-6168: flavin biosynthesis III (fungi) 172.2879499 0.689038903 0.005629 0.097618 c2c3
PWY-7323: superpathway of GDP-mannose-derived O-antigen building blocks biosynthesis 25.89416668 1.771497516 0.002522 0.067784 c2c3
UDPNAGSYN-PWY: UDP-N-acetyl-D-glucosamine biosynthesis I 67.18613175 ‒1.506925835 0.001515 0.066483 c2c3
ARG+POLYAMINE-SYN: super pathway of arginine and polyamine biosynthesis 69.58601257 ‒0.960789029 0.000298 0.042061 c2c1
ARG+POLYAMINE-SYN: super pathway of arginine and polyamine biosynthesis 69.58601257 ‒1.05230553 0.000918 0.066483 c2c3
ARGININE-SYN4-PWY: L-ornithine de novo biosynthesis 91.08538996 1.466876718 0.001004 0.066483 c2c3
ARGININE-SYN4-PWY: L-ornithine de novo biosynthesis 91.08538996 1.144773909 0.001917 0.069068 c2c1
PWY-6471: peptidoglycan biosynthesis IV (Enterococcus faecium) 10.68397596 ‒2.509515632 0.004524 0.081003 c2c1
PWY-6471: peptidoglycan biosynthesis IV (Enterococcus faecium) 10.68397596 ‒3.293509459 0.001507 0.066483 c2c3
PYRIDOXSYN-PWY: pyridoxal 5-phosphate biosynthesis I 83.95260245 1.49487838 0.002165 0.066483 c2c3
PYRIDOXSYN-PWY: pyridoxal 5-phosphate biosynthesis I 83.95260245 1.201030452 0.002913 0.074673 c2c1

Abbreviation: adj, adjusted.

Supplementary Table S8.

List of Differentially Expressed Genes that Are Differentially Abundant between Different Subgroups in the Cohort

Ensembl_Gene_ID Base Mean Log2 Fold Change Padj Gene Name Comparison Pair
ENSG00000155966 70.519 3.74964 0.000392968 AFF2 c3c1
ENSG00000233680 35.444 3.02891 0.001437268 HNRNPA1P27 c2c1
ENSG00000187527 19.131 2.03977 0.049354713 ATP13A5 c2c3
ENSG00000214671 84.817 0.83427 0.022303138 RPL6P12 c2c1
ENSG00000214595 713.756 1.01421 0.018629526 EML6 c3c1
ENSG00000173040 124.774 ‒0.84891 0.038601379 EVC2 c2c3
ENSG00000238025 58.599 ‒1.37982 0.039362413 ZDHHC4P1 c2c1
ENSG00000162897 190.900 1.35991 0.02346219 FCAMR c3c1
ENSG00000086205 10.244 ‒2.40497 0.000829552 FOLH1 c2c3
ENSG00000181867 0.673 ‒20.54710 0.000897703 FTMT c2c1
ENSG00000158089 21.123 2.26847 0.013980259 GALNT14 c3c1
ENSG00000274618 372.111 1.52264 0.008495718 HIST1H4F c3c1
ENSG00000166736 16.410 ‒3.99495 0.032119336 HTR3A c2c3
ENSG00000166736 16.410 3.54715 0.013195954 HTR3A c3c1
ENSG00000241244 319.624 ‒1.36024 0.020133239 IGKV1D-16 c2c3
ENSG00000129451 40.860 ‒4.35734 0.035837072 KLK10 c2c3
ENSG00000129451 40.860 3.74074 0.025102134 KLK10 c3c1
ENSG00000225128 0.677 23.60138 0.005620613 LINC00972 c2c3
ENSG00000225128 0.677 ‒17.31295 0.038958367 LINC00972 c3c1
ENSG00000182333 0.399 12.17337 0.003881171 LIPF c2c3
ENSG00000264251 0.669 10.28858 0.024886441 RN7SL819P c2c3
ENSG00000182333 0.399 ‒12.36307 5.74E–05 LIPF c3c1
ENSG00000249333 0.470 ‒14.41122 1.34E–06 LL22NC03-23C6.12 c2c1
ENSG00000234031 27.687 1.34310 0.039573133 RPS3AP44 c2c3
ENSG00000249333 0.470 ‒16.86785 1.46E–05 LL22NC03-23C6.12 c2c3
ENSG00000271765 63.342 0.91097 0.038601379 RN7SKP299 c2c3
ENSG00000118308 1,060.651 1.25761 0.049743284 LRMP c3c1
ENSG00000266276 0.285 ‒17.22075 0.004989858 MIR4743 c3c1
ENSG00000240666 3.840 ‒5.06250 0.015579404 MME-AS1 c2c3
ENSG00000228232 975.118 ‒1.27782 0.014609163 GAPDHP1 c2c3
ENSG00000215182 291.333 ‒4.57590 0.005872574 MUC5AC c2c3
ENSG00000215182 291.333 4.07199 0.002093636 MUC5AC c3c1
ENSG00000185697 288.454 1.51200 0.012506363 MYBL1 c3c1
ENSG00000160505 11.579 ‒5.48847 0.008074731 NLRP4 c2c3
ENSG00000265018 49.486 ‒2.47429 0.009651198 AGAP12P c2c3
ENSG00000160505 11.579 4.54221 0.006739737 NLRP4 c3c1
ENSG00000163545 1,334.424 ‒0.72970 0.030687143 NUAK2 c2c3
ENSG00000278561 12.597 ‒3.56800 0.024920457 PTPN20CP c2c3
ENSG00000189233 376.826 ‒1.37169 0.015794615 NUGGC c2c3
ENSG00000160191 3,535.762 1.00276 0.026512132 PDE9A c2c1
ENSG00000236717 0.463 ‒23.22885 6.47E–16 RP11-100G15.10 c2c1
ENSG00000236717 0.463 ‒23.12038 1.84E–08 RP11-100G15.10 c2c3
ENSG00000231090 0.555 ‒13.99578 7.79E–05 RP11-101C11.1 c2c1
ENSG00000231090 0.555 ‒17.07458 0.000194189 RP11-101C11.1 c2c3
ENSG00000257513 3.422 ‒4.72468 0.008074731 NPIPB1P c2c3
ENSG00000272988 0.267 ‒15.44717 0.023939688 RP11-150D20.5 c3c1
ENSG00000259772 165.237 ‒0.83726 0.011698659 RP11-16E12.2 c2c3
ENSG00000282950 0.907 ‒26.60203 6.09E–05 RP11-174O3.6 c2c3
ENSG00000260303 23.670 ‒4.86844 0.01227057 RP11-203B7.2 c2c3
ENSG00000260303 23.670 4.12079 0.010443355 RP11-203B7.2 c3c1
ENSG00000258675 0.457 ‒12.87191 0.006683019 RP11-299L17.3 c3c1
ENSG00000275314 0.741 10.03950 0.024849813 RP11-32A1.2 c2c3
ENSG00000275314 0.741 ‒10.90604 0.000262234 RP11-32A1.2 c3c1
ENSG00000259476 1.221 13.39380 0.001571351 RP11-50C13.2 c2c3
ENSG00000259476 1.221 ‒13.63577 2.56E–05 RP11-50C13.2 c3c1
ENSG00000206532 8.420 ‒3.15479 0.04914993 RP11-553A10.1 c2c3
ENSG00000249877 3.764 ‒13.38306 0.010980955 RP11-706F1.2 c3c1
ENSG00000280022 44.512 ‒5.02298 0.001165293 RP11-707O23.1 c2c1
ENSG00000258962 113.659 2.06291 0.004479337 RP11-747H7.1 c2c1
ENSG00000260711 1,768.668 1.06849 0.039573133 RP11-747H7.3 c2c3
ENSG00000203620 0.321 13.96751 0.032593228 RP11-84A19.2 c2c3
ENSG00000203620 0.321 ‒13.23645 0.008565686 RP11-84A19.2 c3c1
ENSG00000231654 8.334 ‒2.42875 0.024773164 RPS6KA2-AS1 c2c1
ENSG00000170054 54.004 6.27792 0.001444378 SERPINA9 c3c1
ENSG00000142583 152.205 1.50167 0.008107758 SLC2A5 c3c1
ENSG00000243770 14.943 ‒1.76051 0.02152624 RN7SL65P c3c1
ENSG00000198203 1,020.018 ‒1.26205 0.000897703 SULT1C2 c2c1
ENSG00000198203 1,020.018 ‒1.24560 0.049945372 SULT1C2 c2c3
ENSG00000196228 130.871 ‒1.37044 0.006397675 SULT1C3 c2c1
ENSG00000157303 105.695 1.10998 0.013195954 SUSD3 c3c1
ENSG00000187621 53.221 2.55063 0.013195954 TCL6 c3c1
ENSG00000088992 54.739 1.29575 0.013195954 TESC c3c1
ENSG00000160182 2,794.990 1.52164 0.038058354 TFF1 c3c1
ENSG00000160181 62.719 1.80337 0.021301346 TFF2 c3c1
ENSG00000169903 40.103 3.38885 0.012506363 TM4SF4 c3c1
ENSG00000275154 16.556 2.73116 0.024773164 U3 c2c1
ENSG00000175567 592.509 ‒0.78874 0.006712427 UCP2 c2c3
ENSG00000112303 146.742 ‒1.84313 0.027895978 VNN2 c2c3
ENSG00000112303 146.742 1.50589 0.040547819 VNN2 c3c1
ENSG00000101842 47.686 ‒2.79232 0.049611364 VSIG1 c2c3
ENSG00000101842 47.686 2.42312 0.034021561 VSIG1 c3c1
ENSG00000158023 52.914 1.39125 0.012506363 WDR66 c3c1

Abbreviations: adj, adjusted; ID, identification.

Supplementary Table S9.

Comparison of Immune Profiles from Flow Cytometry and Cibersort among Different Psoriasis Subgroups

Data Source Immune Population AVG ± SD
Significance (Wilcox Test)
Healthy PSO1 PSO2 PSO3 Healthy Versus PSO1 Healthy Versus PSO2 Healthy Versus PSO3 PSO1 Versus PSO2 PSO1 Versus PSO3 PSO2 Versus PSO3
Immunephenotype Teff (CD3+ CD4+ CD25‒ FoxP3‒) 80.49 ± 5.98% 83.33 ± 3.15% 83.49 ± 3.71% 80.8 ± 3.29% n.s. n.s. n.s. n.s. n.s. n.s.
Active Teff (CD3+ CD4+ CD25+ FoxP3‒ CD38+ HLADR+) 0.13 ± 0.09% 0.27 ± 0.26% 0.09 ± 0.03% 0.36 ± 0.25% n.s. n.s. 0.035 n.s. n.s. 0.024
Memory Teff (CD3+ CD4+ CD25‒ FoxP3‒ CD45RO+) 33.72 ± 10.14% 34.55 ± 13.64% 35.81 ± 12.12% 36.3 ± 10.2% n.s. n.s. n.s. n.s. n.s. n.s.
Active memory Teff (CD3+ CD4+ CD25‒ FoxP3‒ CD45RO+ CD38+ HLADR+) 0.31 ± 0.34% 0.55 ± 0.55% 0.18 ± 0.08% 0.67 ± 0.56% n.s. n.s. n.s. 0.018 n.s. 0.02
Tregs (CD3+ CD4+ CD25+ FoxP3+) 6.46 ± 2.66% 5.81 ± 1.03% 6.13 ± 1.22% 6.53 ± 1.26% n.s. n.s. n.s. n.s. n.s. n.s.
Activated Tregs (CD3+ CD4+ CD25+ FoxP3+, CD45RO+) 70.65 ± 11.6% 67.99 ± 9.84% 66.94 ± 10.33% 69.95 ± 6.3% n.s. n.s. n.s. n.s. n.s. n.s.
Cytokine production Teff (CD3+ CD4+ CD127+) 58.51 ± 15.43% 52.79 ± 18.71% 58.85 ± 10.83% 39.48 ± 18.33% n.s. n.s. 0.02 n.s. n.s. 0.04
Teff IL17+ only 0.93 ± 0.87% 1.1 ± 1.56% 0.78 ± 0.53% 1.11 ± 0.5% n.s. n.s. n.s. n.s. n.s. n.s.
Teff IL22+ only 0.66 ± 0.51% 0.68 ± 0.39% 1.75 ± 2% 0.89 ± 0.58% n.s. n.s. n.s. n.s. n.s. n.s.
Teff IL17+ IL22+ 0.13 ± 0.15% 0.15 ± 0.16% 0.32 ± 0.41% 0.15 ± 0.06% n.s. n.s. n.s. n.s. n.s. n.s.
Teff TNF-α+ only 0.69 ± 0.77% 0.84 ± 0.87% 1.28 ± 1.37% 0.6 ± 0.21% n.s. n.s. n.s. n.s. n.s. n.s.
Teff IFNγ+ only 10.73 ± 3.92% 11 ± 4.07% 12.02 ± 7.43% 11.81 ± 4.17% n.s. n.s. n.s. n.s. n.s. n.s.
Teff TNF-α+ IFNγ+ 0.63 ± 0.71% 0.44 ± 0.3% 1 ± 1.64% 0.44 ± 0.23% n.s. n.s. n.s. n.s. n.s. n.s.
CIBERSORTx Th1 0.02 ± 0.02% 0.01 ± 0.01% 0.03 ± 0.02% 0.02 ± 0.02% n.s. n.s. n.s. 0.012 n.s. n.s.
Th17 0.08 ± 0.02% 0.08 ± 0.02% 0.09 ± 0.02% 0.09 ± 0.03% n.s. n.s. n.s. n.s. n.s. n.s.
Treg 0.05 ± 0% 0.05 ± 0% 0.05 ± 0.01% 0.06 ± 0.01% n.s. n.s. n.s. n.s. n.s. 0.02
Tcm 0.02 ± 0% 0.02 ± 0% 0.02 ± 0% 0.02 ± 0% n.s. n.s. n.s. n.s. n.s. n.s.
Tfh 0.01 ± 0% 0.01 ± 0% 0.01 ± 0% 0.01 ± 0% n.s. n.s. n.s. n.s. n.s. n.s.
Activated CD4 T 0.14 ± 0.03% 0.14 ± 0.02% 0.14 ± 0.02% 0.11 ± 0.01% n.s. n.s. 0.014 n.s. 0.024 0.02
Immunephenotype CD8(CD3+ CD8+) 30.19 ± 10.33% 32.09 ± 12.33% 24.66 ± 9.14% 31 ± 12.52% n.s. n.s. n.s. n.s. n.s. n.s.
Active CD8 (CD3+ CD8+ CD38+ HLADR+) 0.52 ± 0.28% 1.3 ± 1.65% 0.44 ± 0.25% 1.09 ± 0.81% n.s. n.s. n.s. n.s. n.s. n.s.
Memory CD8 (CD3+ CD8+ CD45RO+) 15.36 ± 6.22% 15.57 ± 8.13% 25.34 ± 6.52% 16.45 ± 6.41% n.s. 0.002 n.s. 0.02 n.s. 0.029
Activated memory CD8 (CD3+ CD8+ CD45RO+ CD38+ HLADR+) 2.04 ± 1.61% 3.5 ± 4.8% 1.18 ± 1.01% 3.09 ± 1.81% n.s. n.s. n.s. n.s. n.s. 0.04
Cytokine production CD8 (CD3+ CD8+) 27.21 ± 10.1% 30.32 ± 12.55% 23.69 ± 9.09% 27.08 ± 13.52% n.s. n.s. n.s. n.s. n.s. n.s.
CD8+ IL17+ only 0.12 ± 0.08% 0.15 ± 0.08% 0.28 ± 0.36% 0.17 ± 0.08% n.s. n.s. n.s. n.s. n.s. n.s.
CD8+ IL22+ only 0.14 ± 0.19% 0.07 ± 0.06% 0.47 ± 0.64% 0.08 ± 0.04% n.s. n.s. n.s. 0.023 n.s. n.s.
CD8+ IL17+ IL22+ 0.02 ± 0.01% 0.02 ± 0.01% 0.15 ± 0.2% 0.02 ± 0.03% n.s. n.s. n.s. n.s. n.s. n.s.
CD8+ TNF-α+ only 0.04 ± 0.07% 0.09 ± 0.22% 0.14 ± 0.18% 0.06 ± 0.06% n.s. n.s. n.s. n.s. n.s. n.s.
CD8+ IFNγ+ only 16.47 ± 7.85% 16.09 ± 9.52% 20.17 ± 12.71% 20.45 ± 8.55% n.s. n.s. n.s. n.s. n.s. n.s.
CD8+ TNF-α+ IFNγ+ 0.4 ± 0.29% 0.35 ± 0.35% 0.46 ± 0.65% 0.66 ± 0.58% n.s. n.s. n.s. n.s. n.s. n.s.
CIBERSORTx CD8 T 0.00375 ± 0.0065% 0.00326 ± 0.00408% 0.00053 ± 0.00133% 0.01483 ± 0.01089% n.s. n.s. 0.026 0.043 0.042 0.011

Abbreviations: AVG, average; n.s., not significant; Tcm, T central memory; Teff, effector T cell; tfh, T follicular helper; Th, T helper; Treg, regulatory T cell.

Supplementary Table S10.

Summary of Datasets Used in Multiomic Analysis

Dataset Measurement Type Inclusion Criteria No. of Features Included Normalization Method
Microbial spp. (n = 40) Shotgun metagenomics from stool samples Nonzero counts in at least 10 samples 142 Counts per million
Microbial UniRef90 gene families (n = 40) Shotgun metagenomics from stool samples At least three counts for 80% of all metagenomics samples (total is 52 samples) 2,488 Counts per million
Microbial MetaCyc pathways (n = 40) Shotgun metagenomics from stool samples All metacyc pathways identified by humann2 334 Counts per million
Flow cell population (n = 40) Flow cytometry from PBMCs Include all data collected 25 Percentage of parental gate
Flow cytokine production (n = 40) Flow cytometry from PBMCs Include all data collected 29 Percentage of parental gate
Host RNA-seq (n = 40) RNA-seq from sigmoidal colon biopsies Top 500 most variable genes + all DE genes identified by either disease comparison or PSO subgroup comparisons + inflammatory genes 601 vst transformation to ensure variance is constant across the data

Abbreviations: DE, differentially expressed; No., number; PSO, psoriasis; RNA-seq, RNA sequencing; vst, variance stabilizing transformation.

Supplementary Table S11.

List of Significant Associations in Host Microbial Multiomics Network

From To Cor P-Value Padj Greedy Membership
Significant associations in health-associated network
s__Catenibacterium_mitsuokai CD8_TNFa_only 0.80837 0.000118 0.047311 1
PWY4FS-7: phosphatidylglycerol biosynthesis I (plastidic) Teff_IFNg_Only 0.643956 0.00019 0.095866
PWY4FS-8: phosphatidylglycerol biosynthesis II (non-plastidic) Teff_IFNg_Only 0.643956 0.00019 0.095866
PWY4FS-7: phosphatidylglycerol biosynthesis I (plastidic) Teff_TNFa_INFg 0.621978 1.49E–06 0.003106
PWY4FS-8: phosphatidylglycerol biosynthesis II (non-plastidic) Teff_TNFa_INFg 0.621978 1.49E–06 0.003106
CD8_TNFa_only IL1R2 ‒0.68647 2.35E–06 0.006511
s__Catenibacterium_mitsuokai NOS2 0.805281 0.000223 0.096093
Teff_TNFa_INFg NOS2 0.775824 0.000257 0.091006
CD8_TNFa_only ST6GAL2 ‒0.64466 3.23E–05 0.030873
s__Catenibacterium_mitsuokai ST6GAL2 ‒0.66447 4.78E–06 0.005155
PHOSLIPSYN-PWY: superpathway of phospholipid biosynthesis I (bacteria) CD8_IFNg_only 0.767033 0.000162 0.092121 2
Treg_INFg_only CCDC144A 0.749451 0.000105 0.062263
Treg_INFg_only RP11-219A15.1 0.796066 2.5E–05 0.02777
CD8_IFNg_only RP11-336A10.5 0.818482 2.1E–05 0.02777
CD8_IL22_only RP11-336A10.5 0.752476 0.000181 0.080494
Treg_INFg_only RP11-336A10.5 0.673268 0.000104 0.062263
Treg_TNFa_IFNg RP11-336A10.5 0.790289 0.000219 0.088193
Treg_INFg_only USP32P1 0.783672 0.000112 0.06403
PWY-6125: superpathway of guanosine nucleotides de novo biosynthesis II activated.memory.Teff 0.686469 0.000357 0.021066 3
PWY-7184: pyrimidine deoxyribonucleotides de novo biosynthesis I activated.memory.Teff 0.638767 0.000445 0.025005
PWY-7197: pyrimidine deoxyribonucleotide phosphorylation activated.memory.Teff 0.613862 0.001624 0.073648
s__Escherichia_coli activated.memory.Teff 0.60066 1.89E–05 0.003131
FUC-RHAMCAT-PWY: super pathway of fucose and rhamnose degradation Active.Teff 0.786207 0.001056 0.051489
PWY-6125: super pathway of guanosine nucleotides de novo biosynthesis II Active.Teff 0.63631 0.001602 0.073212
PWY-6628: superpathway of L-phenylalanine biosynthesis Active.Teff 0.728019 7.38E–06 0.0016
s__Escherichia_coli Active.Teff 0.660784 8.59E–06 0.001669
UniRef90_K6B179: NO_NAME Active.Teff 0.604715 1.58E–06 0.041485
s__Eubacterium_ventriosum IGLV8-61 ‒0.73511 0.000141 0.073261 4
s__Ruminococcus_obeum IGLV8-61 ‒0.6 0.000147 0.0755
UniRef90_A5ZMH0: NO_NAME IGLV8-61 ‒0.81588 2.75E–06 0.065021
UniRef90_D4LLH7: ATPases involved in chromosome partitioning IGLV8-61 ‒0.76686 3.19E–06 0.070247
UniRef90_R6S2S2: Ketol-acid reductoisomerase IGLV8-61 ‒0.71356 5.13E–06 0.091049
GLYCOLYSIS: glycolysis I (from glucose 6-phosphate) memory.CD8 0.78022 0.000181 0.011891 5
PWY-5484: glycolysis II (from fructose 6-phosphate) memory.CD8 0.789011 0.000203 0.013021
UniRef90_D4M0P1: ADP-ribose pyrophosphatase memory.CD8 0.899252 1.03E–05 0.087323
NONOXIPENT-PWY: pentose phosphate pathway (non-oxidative branch) RP11-716A19.3 0.61012 7.69E–06 0.031569 6
UniRef90_R7JV77: NO_NAME RP11-716A19.3 0.610793 4.62E–06 0.086393
NONOXIPENT-PWY: pentose phosphate pathway (non-oxidative branch) RP4-672N11.1 0.61012 7.69E–06 0.031569
UniRef90_R7JV77: NO_NAME RP4-672N11.1 0.610793 4.62E–06 0.086393
PPGPPMET-PWY: ppGpp biosynthesis activated.memory.CD8 0.601323 0.001952 0.08521 7
PWY-821: superpathway of sulfur amino acid biosynthesis (Saccharomyces cerevisiae) activated.memory.CD8 0.686806 7.64E–05 0.006382
NONMEVIPP-PWY: methylerythritol phosphate pathway I ADGRG7 0.868132 2.01E–05 0.062622 8
s__Prevotella_copri ADGRG7 0.871288 3.52E–05 0.0262
GLUCUROCAT-PWY: superpathway of β-D-glucuronide and D-glucuronate degradation SULT1C2 ‒0.92527 2.97E–05 0.078414 9
UniRef90_D4K8E3: Predicted phosphatase homologous to the C-terminal domain of histone macroH2A1 SULT1C2 ‒0.9083 7.16E–07 0.027384
CD8_Immune.function ZG16 0.732674 0.000189 0.080761 10
CD8_Immune.Pop ZG16 0.767033 3.68E–05 0.065578
UniRef90_C7H1R7: NO_NAME MST1L ‒0.90309 1.41E–06 0.040723 11
UniRef90_C7H1R7: NO_NAME MST1P2 ‒0.89428 3.41E–07 0.020076
s__Eubacterium_hallii AP000350.5 0.91339 9.22E–05 0.05574 12
s__Eubacterium_hallii AP000350.6 0.792502 0.000112 0.062974
UniRef90_D4K1Q8: Predicted integral membrane protein HELLPAR 0.719472 1.51E–06 0.04237 13
UniRef90_E2ZN70: NO_NAME HELLPAR 0.803967 4.87E–06 0.089006
s__Bacteroides_dorei LL22NC03-23C6.12 0.61662 7.52E–05 0.049355 14
s__Bacteroides_dorei RP11-174O3.6 0.604902 9.1E–05 0.055665
UniRef90_G2SYP7: NO_NAME RP11-745A24.3 0.704199 2.9E–06 0.065432 15
UniRef90_G2T527: Filamentation induced by cAMP protein Fic RP11-745A24.3 0.72507 3.86E–06 0.07883
Teff_TNFa_only IGHV6-1 ‒0.80968 2.48E–05 0.02777 16
Teff_TNFa_only REG1A 0.610445 7.99E–05 0.062263
PWY-7219: adenosine ribonucleotides de novo biosynthesis ITGA2 ‒0.91969 2.74E–05 0.076724 17
s__Dorea_formicigenerans Teff_Immune.Pop ‒0.91209 0.000234 0.020021 18
Treg_IL17_IL22 IGLV2-18 ‒0.76005 0.000208 0.086495 19
CD8_TNFa_IFNg TM4SF1 ‒0.65714 0.000312 0.098059 20
UniRef90_R6LGS5: NO_NAME AC131056.3 ‒0.89989 2.79E–06 0.065021 21
ILC3 IGHD 0.748352 7.96E–06 0.018936 22
UniRef90_R5TP61: 50S ribosomal protein L30 IGHV3-33 ‒0.70751 1.16E–06 0.036628 23
UniRef90_R9HTU6: NO_NAME FTMT 0.612826 1.82E–07 0.012321 24
PWY-5177: glutaryl-CoA degradation CDA 0.816282 1.43E–05 0.049172 25
s__Ruminococcus_lactaris EML6 0.76199 4.06E–05 0.029981 26
PWY-7328: superpathway of UDP-glucose-derived O-antigen building blocks biosynthesis IL17F 0.780793 2.95E–05 0.078414 27
UniRef90_D4KCX5: Rhamnulose-1-phosphate aldolase CD177P1 0.963551 2.32E–06 0.056574 28
PWY0-1061: superpathway of L-alanine biosynthesis CD8_IL17_only 0.760754 4.74E–06 0.006165 29
s__Ruminococcus_sp_5_1_39BFAA CD19 0.781079 0.000124 0.066737 30
Significant associations in psoriasis-associated network
1CMET2-PWY: N10-formyl-tetrahydrofolate biosynthesis GAPDHP1 ‒0.6841 9.75E–05 0.064441 1
ARGININE-SYN4-PWY: L-ornithine de novo biosynthesis GAPDHP1 ‒0.7126 2.17E–05 0.023794
PWY-3841: folate transformations II GAPDHP1 ‒0.65402 0.00017 0.092741
PWY-5101: L-isoleucine biosynthesis II GAPDHP1 ‒0.68194 0.000136 0.080946
PWY-6353: purine nucleotides degradation II (aerobic) GAPDHP1 0.663131 0.000167 0.091415
PWY-7282: 4-amino-2-methyl-5-phosphomethylpyrimidine biosynthesis (yeast) GAPDHP1 ‒0.69345 2.78E–05 0.027634
PWY0-845: super pathway of pyridoxal 5-phosphate biosynthesis and salvage GAPDHP1 ‒0.71761 9.73E–06 0.013577
PYRIDOXSYN-PWY: pyridoxal 5-phosphate biosynthesis I GAPDHP1 ‒0.71319 1.28E–05 0.01645
UniRef90_A7V4G2: NO_NAME GAPDHP1 ‒0.75316 5.57E–06 0.058996
UniRef90_Q5LC85: Elongation factor 4 GAPDHP1 ‒0.75949 1.37E–06 0.02837
UniRef90_Q5LES4: Sulfate adenylyltransferase subunit 2 GAPDHP1 ‒0.78468 1.2E–06 0.027047
ARGININE-SYN4-PWY: L-ornithine de novo biosynthesis MIR222HG 0.677723 0.000185 0.098183
PWY-6703: preQ0 biosynthesis MIR222HG 0.635198 0.000171 0.092877
PWY0-845: super pathway of pyridoxal 5-phosphate biosynthesis and salvage MIR222HG 0.697094 0.000186 0.098528
s__Bacteroides_uniformis MIR222HG 0.718291 8.08E–05 0.030159
PWY-6353: purine nucleotides degradation II (aerobic) MTND1P23 ‒0.73465 9.63E–05 0.06407
s__Bacteroides_uniformis NPIPB15 ‒0.60205 0.000127 0.043035
PWY-6703: preQ0 biosynthesis NUAK2 ‒0.69266 0.000187 0.098672
ARGININE-SYN4-PWY: L-ornithine de novo biosynthesis RN7SKP299 0.705078 0.000141 0.082044
PWY-5101: L-isoleucine biosynthesis II RN7SKP299 0.773598 1.22E–05 0.01603
ARGININE-SYN4-PWY: L-ornithine de novo biosynthesis RP11-16E12.2 ‒0.71602 0.000189 0.098755
UniRef90_R5EN96: NO_NAME AP001627.1 0.718549 7.47E–06 0.070498 2
UniRef90_R5ENA5: NO_NAME AP001627.1 0.705786 5.84E–06 0.060871
UniRef90_R5ENA9: NO_NAME AP001627.1 0.651182 7.75E–06 0.072187
UniRef90_R5ENB7: NO_NAME AP001627.1 0.676065 4.98E–06 0.05694
UniRef90_R5ENC3: NO_NAME AP001627.1 0.695925 6.58E–06 0.065657
UniRef90_R5END3: NO_NAME AP001627.1 0.660024 8.62E–06 0.075666
UniRef90_R5ENY1: NO_NAME AP001627.1 0.678813 5.08E–06 0.057196
UniRef90_R5ENZ3: NO_NAME AP001627.1 0.660032 9.77E–06 0.080475
UniRef90_R5EP09: NO_NAME AP001627.1 0.607497 9.06E–06 0.077542
UniRef90_R5ES58: NO_NAME AP001627.1 0.712702 7.09E–06 0.068494
UniRef90_R5ES70: NO_NAME AP001627.1 0.698935 5.74E–06 0.06032
UniRef90_R5ES82: NO_NAME AP001627.1 0.726586 7.68E–06 0.072187
UniRef90_R5EXU5: NO_NAME AP001627.1 0.665197 7.19E–06 0.069054
UniRef90_R5F071: NO_NAME AP001627.1 0.690192 6.4E–06 0.064816
UniRef90_R5F076: NO_NAME AP001627.1 0.759732 4.82E–06 0.056148
UniRef90_R5F1R6: NO_NAME AP001627.1 0.669014 9.65E–06 0.080053
UniRef90_R5F1S4: NO_NAME AP001627.1 0.698402 5.46E–06 0.058301
UniRef90_R5F1T5: NO_NAME AP001627.1 0.677638 3.89E–06 0.051875
UniRef90_R5VN00: NO_NAME AP001627.1 0.707391 5.26E–06 0.057636
UniRef90_C7H6T5: NO_NAME CXCL1 0.607639 1.68E–07 0.006928 3
UniRef90_C7H6T5: NO_NAME CXCL3 0.62271 3.3E–08 0.002159
UniRef90_G2T376: NO_NAME CXCL3 0.61847 9.73E–10 0.000185
UniRef90_G2T5J9: NO_NAME CXCL3 0.607058 2.91E–08 0.001971
PWY-7196: superpathway of pyrimidine ribonucleosides salvage Teff_IL17_IL22 0.703267 0.00045 0.078835 4
PWY-7196: superpathway of pyrimidine ribonucleosides salvage Teff_IL17_only 0.62053 0.000127 0.064567
PPGPPMET-PWY: ppGpp biosynthesis Treg_IL17_only 0.607528 0.000525 0.085532
PWY-7196: superpathway of pyrimidine ribonucleosides salvage Treg_IL17_only 0.630174 0.000704 0.095585
COLANSYN-PWY: colanic acid building blocks biosynthesis IGHV3-30 ‒0.6278 0.000129 0.077505 5
PWY-7323: super pathway of GDP-mannose-derived O-antigen building blocks biosynthesis IGHV3-30 ‒0.62012 5.2E–05 0.043212
COLANSYN-PWY: colanic acid building blocks biosynthesis RP11-16E12.2 ‒0.61959 0.00017 0.092741
ANAGLYCOLYSIS-PWY: glycolysis III (from glucose) IGHV2-70 ‒0.69299 2.94E–05 0.028746 6
CALVIN-PWY: Calvin-Benson-Bassham cycle IGHV2-70 ‒0.75205 9.6E–06 0.01349
NONOXIPENT-PWY: pentose phosphate pathway (non-oxidative branch) IGHV2-70 ‒0.75603 1.87E–05 0.021434
PWY-1042: glycolysis IV (plant cytosol) IGHV2-70 ‒0.73312 2.18E–06 0.00494
PWY-7242: D-fructuronate degradation IGHV2-70 ‒0.69802 8.1E–05 0.056949
COA-PWY: coenzyme A biosynthesis I MTND2P28 ‒0.61331 1.79E–05 0.020776 7
FAO-PWY: fatty acid β-oxidation I MTND2P28 ‒0.64143 2.21E–05 0.023794
PWY-5100: pyruvate fermentation to acetate and lactate II MTND2P28 ‒0.66359 5.47E–05 0.044043
PWY-5136: fatty acid β-oxidation II (peroxisome) MTND2P28 ‒0.61916 3.21E–05 0.030777
PWY-1269: CMP-3-deoxy-D-manno-octulosonate biosynthesis I Vd2 0.679672 0.000179 0.071033 8
PWY-1269: CMP-3-deoxy-D-manno-octulosonate biosynthesis I Vd2T 0.661191 0.000251 0.022565
PWY-1269: CMP-3-deoxy-D-manno-octulosonate biosynthesis I IGHV2-26 ‒0.63313 0.000189 0.098755
HEME-BIOSYNTHESIS-II: heme biosynthesis I (aerobic) IGKV1-17 ‒0.74011 4.13E–05 0.036607 9
PWY-5918: super pathway of heme biosynthesis from glutamate IGKV1-17 ‒0.71627 0.000159 0.087773
s__Streptococcus_parasanguinis KRTAP13-2 ‒0.6127 1.05E–05 0.006086 10
s__Streptococcus_parasanguinis RN7SL65P ‒0.62108 5.04E–06 0.003209
s__Dorea_formicigenerans CD8_TNFa_IFNg 0.651556 0.000876 0.036379 11
UniRef90_G1WU72: NO_NAME CD8_TNFa_IFNg 0.641326 6.26E–05 0.083128
PWY-5384: sucrose degradation IV (sucrose phosphorylase) AGAP12P 0.660269 2.54E–05 0.026031 12
PWY-5384: sucrose degradation IV (sucrose phosphorylase) NPIPB1P 0.648085 0.000185 0.098183
UniRef90_C9YMF0: Sigma-24 (Feci) IL1RN 0.810404 8.15E–06 0.074295 13
UniRef90_D4L1Z0: DNA-directed RNA polymerase specialized sigma subunit, sigma24 homolog IL1RN 0.807528 7.07E–06 0.068494
PWY-3001: super pathway of L-isoleucine biosynthesis I Treg_TNFa_IFNg ‒0.63367 0.000231 0.071033 14
THRESYN-PWY: super pathway of L-threonine biosynthesis Treg_TNFa_IFNg ‒0.72483 7.24E–05 0.050646
P461-PWY: hexitol fermentation to lactate, formate, ethanol and acetate SIGLEC12 0.722109 7.4E–05 0.053292 15
UniRef90_R6SAP9: Orotate phosphoribosyltransferase SIGLEC12 0.743519 2.73E–06 0.040374
FUC-RHAMCAT-PWY: super pathway of fucose and rhamnose degradation RP11-693J15.5 0.622961 0.000178 0.095618 16
UniRef90_R5DXN6: NO_NAME TMEM72 ‒0.6137 9.9E–06 0.081082 17
PWY-5695: urate biosynthesis/inosine 5-phosphate degradation IGHV3-53 ‒0.7541 0.000161 0.088313 18
UniRef90_R5J0A8: Transposase IS605 OrfB family central region SLC6A14 0.727059 9.13E–06 0.077542 19
UniRef90_R9HTU6: NO_NAME RPL6P12 0.644934 1.12E–05 0.087267 20
UniRef90_D4K3E4: Desulfoferrodoxin ferrous iron-binding domain IGLV5-45 0.677288 6.46E–06 0.065139 21
s__Odoribacter_splanchnicus PCK1 ‒0.67833 4.84E–05 0.020322 22
UniRef90_C7H7L8: NO_NAME LL22NC03-23C6.12 0.603595 6.12E–09 0.000802 23
Vd2T_TNFa_IFNg KLK10 0.719637 7.31E–09 2.1E–05 24
UniRef90_C4ZEM9: NO_NAME REG1A 0.653404 3.24E–07 0.010793 25
UniRef90_R5J281: NO_NAME Treg_TNFa 0.66736 2.24E–07 0.004274 26
UniRef90_A8SCB2: NO_NAME GSTA1 ‒0.64042 4E–06 0.052695 27
Treg_INFg_only FOS 0.640109 1.57E–05 0.015843 28
PWY-5345: super pathway of L-methionine biosynthesis (by sulfhydrylation) Teff_IFNg_Only ‒0.67739 0.000424 0.078253 29
P185-PWY: formaldehyde assimilation III (dihydroxyacetone cycle) IGLV4-60 ‒0.68048 9.42E–05 0.063081 30
UniRef90_V8CHB5: NO_NAME DSG3 0.644924 2.65E–06 0.039778 31
PWY0-1296: purine ribonucleosides degradation ALDH1L1 0.623461 0.000178 0.095618 32
HEMESYN2-PWY: heme biosynthesis II (anaerobic) Teff_Immune.Pop ‒0.6012 0.001917 0.065199 33
s__Alistipes_shahii ADAMTS4 0.607468 0.000263 0.071968 34
s__Bacteroidales_bacterium_ph8 IL22 0.621457 1.92E–07 0.000212 35
Significant associations in PSO1-associated network
activated.memory.CD8 RP11-299L17.3 0.650444 1.13E–06 0.00596 1
activated.nonMemory.CD8 RP11-299L17.3 0.650444 1.79E–07 0.001895
activeCD8 RP11-299L17.3 0.650444 2.14E–06 0.007538
UniRef90_D4JPG6: NO_NAME RP11-299L17.3 0.65273 1.51E–06 0.090737
UniRef90_U4D938: NO_NAME RP11-299L17.3 0.651584 1.37E–07 0.030625
UniRef90_D4LM01: Site-specific recombinase XerD activated.memory.CD8 0.804932 0.000156 0.095101
UniRef90_D4LWR3: RNA polymerase sigma factor, sigma-70 family activated.memory.CD8 0.712285 1.54E–05 0.033677
UniRef90_D4MWC2: Replication initiator protein A (RepA) N-terminus activated.memory.CD8 0.642109 3.2E–05 0.047927
UniRef90_U4D938: NO_NAME activated.memory.CD8 0.612961 3.6E–05 0.047962
UniRef90_D4LWR3: RNA polymerase sigma factor, sigma-70 family activated.memory.Teff 0.648507 2.11E–05 0.038011
UniRef90_C4Z761: Citrate synthase activeCD8 0.617548 0.000109 0.08439
UniRef90_G2T5D6: Glutathione synthase activeCD8 0.624565 3.18E–05 0.047927
s__Flavonifractor_plautii activated.nonMemory.Teff 0.666676 6.31E–10 1.56E–07 2
s__Ruminococcus_sp_5_1_39BFAA activated.nonMemory.Teff 0.616463 0.000705 0.069518
UniRef90_C6JCI4: NO_NAME activated.nonMemory.Teff 0.645614 0.000177 0.098154
UniRef90_D4LLE0: Aldehyde dehydrogenase, molybdenum-binding subunit apoprotein activated.nonMemory.Teff 0.660215 9.81E–05 0.082059
UniRef90_R5HR90: NO_NAME activated.nonMemory.Teff 0.635567 6.87E–05 0.069169
UniRef90_R5HVG1: NO_NAME activated.nonMemory.Teff 0.690686 1.74E–05 0.034534
UniRef90_R5I7T7: ATP-dependent 6-phosphofructokinase activated.nonMemory.Teff 0.695959 0.000192 0.098154
UniRef90_R5XT20: Rhamnulose-1-phosphate aldolase activated.nonMemory.Teff 0.706504 0.000132 0.092377
UniRef90_R6GH13: NO_NAME activated.nonMemory.Teff 0.731108 3.57E–05 0.047962
UniRef90_R6PH30: NO_NAME activated.nonMemory.Teff 0.682456 1.65E–05 0.034534
UniRef90_R6Q745: NO_NAME activated.nonMemory.Teff 0.742962 2.36E–05 0.040223
UniRef90_R7CD60: NO_NAME activated.nonMemory.Teff 0.695959 8.4E–05 0.076379
UniRef90_R7CD72: NO_NAME activated.nonMemory.Teff 0.695959 0.000145 0.092467
UniRef90_R7CDD0: NO_NAME activated.nonMemory.Teff 0.700709 0.000163 0.096324
UniRef90_R7CGR6: NO_NAME activated.nonMemory.Teff 0.685965 0.00013 0.092377
UniRef90_R7CGS0: NO_NAME activated.nonMemory.Teff 0.73592 0.000135 0.092377
UniRef90_R7CGX0: Nicotinate phosphoribosyltransferase activated.nonMemory.Teff 0.64437 0.000179 0.098154
UniRef90_R7CGX6: NO_NAME activated.nonMemory.Teff 0.607018 0.00012 0.09182
UniRef90_R7CH88: NO_NAME activated.nonMemory.Teff 0.697716 0.000174 0.098154
UniRef90_R7CIC0: NO_NAME activated.nonMemory.Teff 0.666676 0.000155 0.095101
UniRef90_R7CIF8: NO_NAME activated.nonMemory.Teff 0.64386 0.000128 0.092377
UniRef90_R7CK52: NO_NAME activated.nonMemory.Teff 0.741653 0.000141 0.092467
UniRef90_R7CLQ2: Cyclic pyranopterin monophosphate synthase accessory protein activated.nonMemory.Teff 0.674929 9.2E–05 0.080527
UniRef90_R7CLX2: NO_NAME activated.nonMemory.Teff 0.655537 0.000181 0.098154
UniRef90_R7CPD9: NO_NAME activated.nonMemory.Teff 0.652022 3.76E–05 0.048
UniRef90_R7CPY8: NO_NAME activated.nonMemory.Teff 0.721193 0.000124 0.092377
UniRef90_R7CR35: ATP synthase activated.nonMemory.Teff 0.698246 0.000102 0.082059
UniRef90_R7CRE2: Acetylglutamate kinase activated.nonMemory.Teff 0.700709 0.00019 0.098154
UniRef90_R7CSG7: NO_NAME activated.nonMemory.Teff 0.675439 8.46E–05 0.076379
UniRef90_R7CSS1: NO_NAME activated.nonMemory.Teff 0.734623 0.000134 0.092377
UniRef90_R7DCE8: NO_NAME activated.nonMemory.Teff 0.601055 5.27E–05 0.058851
UniRef90_R7NEY7: Acetyltransferases activated.nonMemory.Teff 0.625457 1.66E–06 0.008485
UniRef90_T5LV87: IS605 OrfB family transposase activated.nonMemory.Teff 0.624561 0.000135 0.092377
UniRef90_T5M0M6: NO_NAME activated.nonMemory.Teff 0.711034 6.03E–05 0.062774
UniRef90_E1YTP4: NO_NAME activated.nonMemory.Teff 0.790135 5.17E–07 0.003527 3
UniRef90_R7CII3: NO_NAME activated.nonMemory.Teff 0.670823 0.000184 0.098154
UniRef90_R7CQN6: TrkA-N domain-containing protein activated.nonMemory.Teff 0.699664 0.000171 0.098154
UniRef90_D4LQI2: Site-specific recombinases, DNA invertase Pin homologs Teff_IL17_IL22 0.652583 2.91E–06 0.015436
UniRef90_C6J953: NO_NAME Teff_IL17_only 0.671967 1.62E–06 0.011888
UniRef90_C6JBL1: NO_NAME Teff_IL17_only 0.610238 4.71E–05 0.065888
UniRef90_C6JEF2: NO_NAME Teff_IL17_only 0.62527 2.19E–05 0.047594
UniRef90_D4LQI1: Site-specific recombinases, DNA invertase Pin homologs Teff_IL17_only 0.698422 4.94E–05 0.066521
UniRef90_D4LQI2: Site-specific recombinases, DNA invertase Pin homologs Teff_IL17_only 0.627224 3.5E–06 0.016735
UniRef90_E1YTP4: NO_NAME Teff_IL17_only 0.62082 6.42E–06 0.022067
UniRef90_R5XAV3: Heavy-metal-associated domain Teff_IL17_only 0.62566 9.01E–07 0.008609
UniRef90_R7CFY1: NO_NAME Teff_IL17_only 0.669696 2.91E–05 0.054164
UniRef90_R7CHB7: Na+/H+ antiporter NhaC Teff_IL17_only 0.811625 1.33E–05 0.035418
UniRef90_R7CII3: NO_NAME Teff_IL17_only 0.610643 0.000114 0.091508
UniRef90_R7CMK6: NO_NAME Teff_IL17_only 0.637328 1.29E–07 0.002051
UniRef90_R7CN26: NO_NAME Teff_IL17_only 0.666082 3.98E–06 0.017267
UniRef90_R7CQN6: TrkA-N domain-containing protein Teff_IL17_only 0.61839 0.00011 0.090629
UniRef90_A5ZNM4: NO_NAME Teff_IL17_IL22 0.734266 5.1E–06 0.019503 4
UniRef90_A5ZYV3: NO_NAME Teff_IL17_IL22 0.706294 3.54E–05 0.06035
UniRef90_A5ZYV6: NO_NAME Teff_IL17_IL22 0.686516 2.57E–06 0.015322
UniRef90_C6JC21: NO_NAME Teff_IL17_IL22 0.607808 6.47E–06 0.022067
UniRef90_D4JAB3: Glyoxalase/Bleomycin resistance protein/Dioxygenase superfamily Teff_IL17_IL22 0.794387 0.000109 0.090629
UniRef90_D4LKT7: 50S ribosomal protein L7/L12 Teff_IL17_IL22 0.684624 0.000117 0.091508
UniRef90_D4LRE4: Transcriptional regulator, BadM/Rrf2 family Teff_IL17_IL22 0.666693 9.86E–05 0.086726
UniRef90_D4LVZ1: Predicted transcriptional regulators Teff_IL17_IL22 0.766035 8.23E–07 0.008609
UniRef90_D4MTV2: Transcriptional regulator, MerR family Teff_IL17_IL22 0.713542 5.75E–05 0.073251
UniRef90_D4MUD3: Transcriptional regulator, MarR family Teff_IL17_IL22 0.729829 1.16E–06 0.009737
UniRef90_F7JEX2: NO_NAME Teff_IL17_IL22 0.711285 3.92E–06 0.017267
UniRef90_R5Z6G3: NO_NAME Teff_IL17_IL22 0.678483 2.85E–06 0.015436
UniRef90_R7CCT5: NO_NAME Teff_IL17_IL22 0.678393 0.000148 0.099236
UniRef90_D4MUD3: Transcriptional regulator, MarR family Teff_IL17_only 0.75747 8.66E–08 0.002044
s__Bacteroides_thetaiotaomicron CD300E 0.640982 2.63E–05 0.02332 5
UniRef90_G2T376: NO_NAME CXCL2 0.735024 1.52E–06 0.090737
s__Bacteroides_thetaiotaomicron CXCL3 0.704029 3.68E–06 0.007238
UniRef90_G2T376: NO_NAME CXCL3 0.699686 6.32E–07 0.045297
UniRef90_G2T5J9: NO_NAME CXCL3 0.727956 7.33E–07 0.0507
UniRef90_G2T376: NO_NAME LRFN5 0.657281 4.08E–07 0.040956
s__Bacteroides_thetaiotaomicron SOCS3 0.704029 8.85E–05 0.050575
UniRef90_D4LVH8: KaiC activated.memory.CD8 0.78459 0.000144 0.092467 6
UniRef90_R9JXY1: 50S ribosomal protein L33 activated.nonMemory.Teff 0.648507 0.000165 0.096324
UniRef90_A7B829: NO_NAME Active.Teff 0.620145 0.000188 0.098154
UniRef90_D4LVH8: KaiC Active.Teff 0.697716 0.000175 0.098154
UniRef90_G2T5D0: NO_NAME Active.Teff 0.651408 0.000155 0.095101
UniRef90_K1S8B5: Transcriptional regulator Active.Teff 0.739437 0.000158 0.09531
UniRef90_R9JXY1: 50S ribosomal protein L33 Active.Teff 0.669014 0.00014 0.092467
UniRef90_U2PAU4: NO_NAME Active.Teff 0.870181 5.14E–07 0.003527
s__Barnesiella_intestinihominis LL22NC03-23C6.12 0.651584 9.5E–05 0.052803 7
UniRef90_C7H7L9: ABC transporter, ATP-binding protein LL22NC03-23C6.12 0.65273 2.34E–07 0.036053
s__Barnesiella_intestinihominis SERPINA9 0.747806 3.18E–05 0.026686
s__Eubacterium_hallii GSTA1 0.72028 0.000187 0.078443 8
UniRef90_E2ZGA1: NO_NAME GSTA1 ‒0.82312 1.06E–06 0.071004
s__Eubacterium_ventriosum Vd2T_TNFa_only 0.797189 5.33E–05 0.006695 9
UniRef90_B0NZF6: NO_NAME Vd2T_TNFa_only 0.64675 0.000105 0.090542
UniRef90_D4M5F3: dTDP-glucose pyrophosphorylase memory.Teff ‒0.88422 0.000101 0.082059 10
UniRef90_D4M5F3: dTDP-glucose pyrophosphorylase nonMemory.Teff 0.880707 5.17E–05 0.058792
DZ_duration CD8_IL17_IL22 0.654946 0.000478 0.040147 11
PWY-5104: L-isoleucine biosynthesis IV DZ_duration 0.736492 0.000281 0.068952
UniRef90_B0G2Q7: NO_NAME Treg_IL17_IL22 0.776803 7.01E–05 0.077728 12
UniRef90_D4J5V6: Site-specific recombinase XerD Treg_IL17_IL22 0.693832 9.86E–05 0.086726
UniRef90_A5ZSX2: NO_NAME CD8_TNFa_IFNg 0.907182 7.34E–05 0.077728 13
UniRef90_D4JAA1: Phage integrase family CD8_TNFa_IFNg 0.795496 6.81E–05 0.077728
s__Parabacteroides_distasonis IL17A 0.649429 0.000219 0.085842 14
s__Parabacteroides_distasonis LINC00939 0.751742 2.07E–05 0.020128
s__Bacteroides_dorei FOSB 0.605649 4.51E–05 0.033931 15
s__Bacteroides_dorei Treg_IL22_only 0.643114 3.18E–05 0.004358
Treg_INFg_only RP11-301M17.1 0.650444 2.22E–06 0.009133 16
UniRef90_G2SYP7: NO_NAME Teff_TNFa_only 0.704243 5.65E–06 0.020765 17
UniRef90_C4Z763: GTP pyrophosphokinase Teff_TNFa_INFg 0.804196 0.000117 0.091508 18
PWY0-1297: superpathway of purine deoxyribonucleosides degradation PASI 0.713137 1.85E–05 0.018036 19
UniRef90_C0FPA9: BAAT/acyl-CoA thioester hydrolase C-terminal domain protein nonMemory.CD8 ‒0.87834 0.000123 0.092377 20
s__Alistipes_putredinis CD55 0.881119 0.000117 0.059771 21
s__Dorea_formicigenerans TRHDE 0.895105 4.16E–05 0.032046 22
s__Coprococcus_comes CCDC85A ‒0.63398 0.000222 0.086043 23
s__Lachnospiraceae_bacterium_3_1_46FAA UCA1 0.609458 0.000162 0.072164 24
THRESYN-PWY: super pathway of L-threonine biosynthesis Treg_TNFa_IFNg ‒0.85365 7.05E–06 0.016049 25
UniRef90_C7H2P0: NO_NAME CD8_TNFa_only 0.704029 8.19E–05 0.079819 26
s__Roseburia_inulinivorans CD177 ‒0.83916 1.39E–06 0.003509 27
PYRIDNUCSAL-PWY: NAD salvage pathway I URAD 0.882718 1.02E–07 0.005333 28
Significant associations in PSO2-associated network
BIOTIN-BIOSYNTHESIS-PWY: biotin biosynthesis I CYP2B7P 0.833333 2.98E–05 0.009035 1
FASYN-ELONG-PWY: fatty acid elongation -- saturated CYP2B7P 0.833333 1.4E–05 0.005053
FASYN-INITIAL-PWY: super pathway of fatty acid biosynthesis initiation (E. coli) CYP2B7P 0.826362 2.64E–05 0.008035
GLUDEG-I-PWY: GABA shunt CYP2B7P 0.714286 0.000278 0.074586
PWY-5022: 4-aminobutanoate degradation V CYP2B7P 0.714286 0.000334 0.087365
PWY-5971: palmitate biosynthesis II (bacteria and plants) CYP2B7P 0.833333 2.8E–05 0.008503
PWY-6113: superpathway of mycolate biosynthesis CYP2B7P 0.833333 4.57E–05 0.013734
PWY-6282: palmitoleate biosynthesis I (from (5Z)-dodec-5-enoate) CYP2B7P 0.833333 7.21E–05 0.021414
PWY-6284: super pathway of unsaturated fatty acids biosynthesis (E. coli) CYP2B7P 0.833333 4.07E–05 0.012269
PWY-6519: 8-amino-7-oxononanoate biosynthesis I CYP2B7P 0.833333 3.09E–05 0.009339
PWY-7388: octanoyl-[acyl-carrier protein] biosynthesis (mitochondria, yeast) CYP2B7P 0.833333 1.83E–05 0.006357
PWY-7664: oleate biosynthesis IV (anaerobic) CYP2B7P 0.833333 1.18E–05 0.004438
PWY0-862: (5Z)-dodec-5-enoate biosynthesis CYP2B7P 0.833333 2.54E–05 0.007747
PWYG-321: mycolate biosynthesis CYP2B7P 0.833333 1.31E–05 0.004806
BIOTIN-BIOSYNTHESIS-PWY: biotin biosynthesis I STC1 –0.7619 0.00013 0.03689
FASYN-ELONG-PWY: fatty acid elongation -- saturated STC1 –0.7619 0.000173 0.047907
FASYN-INITIAL-PWY: super pathway of fatty acid biosynthesis initiation (E. coli) STC1 –0.7545 0.000271 0.073002
PWY-5971: palmitate biosynthesis II (bacteria and plants) STC1 ‒0.7619 0.000209 0.05735
PWY-6113: super pathway of mycolate biosynthesis STC1 ‒0.7619 0.000287 0.076805
PWY-6282: palmitoleate biosynthesis I (from (5Z)-dodec-5-enoate) STC1 ‒0.7619 0.00028 0.075069
PWY-6284: super pathway of unsaturated fatty acids biosynthesis (E. coli) STC1 ‒0.7619 0.000347 0.090509
PWY-6519: 8-amino-7-oxononanoate biosynthesis I STC1 ‒0.7619 0.0002 0.055236
PWY-7388: octanoyl-[acyl-carrier protein] biosynthesis (mitochondria, yeast) STC1 ‒0.7619 0.00016 0.044499
PWY-7664: oleate biosynthesis IV (anaerobic) STC1 ‒0.7619 0.000235 0.063921
PWY0-862: (5Z)-dodec-5-enoate biosynthesis STC1 ‒0.7619 0.000229 0.062421
PWYG-321: mycolate biosynthesis STC1 ‒0.7619 0.00012 0.0343
s__Bacteroides_ovatus Teff_TNFa_only 0.666667 0.000994 0.091836 2
s__Bacteroides_ovatus Vd2T_IL17_only 0.761905 0.00021 0.043201
UniRef90_C0FN95: NO_NAME Vd2T_IL17_only 0.880952 5.48E–07 0.026184
UniRef90_D6E3G6: Site-specific recombinase XerD Vd2T_IL17_only 0.90368 1.25E–07 0.011927
ANAEROFRUCAT-PWY: homolactic fermentation FOS ‒0.97619 0.000119 0.034027 3
GLYCOLYSIS: glycolysis I (from glucose 6-phosphate) FOS ‒0.95238 8.01E–05 0.023227
PWY-5484: glycolysis II (from fructose 6-phosphate) FOS ‒0.95238 5.05E–05 0.015132
ANAEROFRUCAT-PWY: homolactic fermentation PDE4C ‒0.90476 9.72E–05 0.028114
ARGSYN-PWY: L-arginine biosynthesis I (via L-ornithine) PASI 0.754505 0.001298 0.055236 4
PENTOSE-P-PWY: pentose phosphate pathway PASI 0.833333 0.002607 0.055236
PWY-5154: L-arginine biosynthesis III (via N-acetyl-L-citrulline) PASI 0.785714 0.001279 0.055236
PWY-7400: L-arginine biosynthesis IV (archaebacteria) PASI 0.754505 0.001234 0.055236
ARGSYN-PWY: L-arginine biosynthesis I (via L-ornithine) XIST ‒0.64672 7.14E–06 0.00271
ARGSYNBSUB-PWY: L-arginine biosynthesis II (acetyl cycle) XIST ‒0.92857 4.15E–06 0.001618
GLUTORN-PWY: L-ornithine biosynthesis XIST ‒0.6627 2.01E–07 9.8E–05
PWY-5154: L-arginine biosynthesis III (via N-acetyl-L-citrulline) XIST ‒0.63474 1.07E–05 0.004031
PWY-7400: L-arginine biosynthesis IV (archaebacteria) XIST ‒0.62655 1.07E–05 0.004046
PWY-6121: 5-aminoimidazole ribonucleotide biosynthesis I BMX ‒0.95238 0.000205 0.056554 5
PWY-6122: 5-aminoimidazole ribonucleotide biosynthesis II BMX ‒0.97619 0.000225 0.061454
PWY-6277: super pathway of 5-aminoimidazole ribonucleotide biosynthesis BMX ‒0.97619 0.000225 0.061454
PWY-6121: 5-aminoimidazole ribonucleotide biosynthesis I CCDC129 ‒0.88095 0.000105 0.030152
PWY-6122: 5-aminoimidazole ribonucleotide biosynthesis II CCDC129 ‒0.90476 0.000107 0.030556
PWY-6277: superpathway of 5-aminoimidazole ribonucleotide biosynthesis CCDC129 ‒0.90476 0.000107 0.030556
PWY-6121: 5-aminoimidazole ribonucleotide biosynthesis I SH2D6 ‒0.97619 2.49E–05 0.007595
PWY-6122: 5-aminoimidazole ribonucleotide biosynthesis II SH2D6 ‒0.95238 7.94E–05 0.023078
PWY-6277: superpathway of 5-aminoimidazole ribonucleotide biosynthesis SH2D6 ‒0.95238 7.94E–05 0.023078
PWY-6121: 5-aminoimidazole ribonucleotide biosynthesis I SH2D7 ‒0.95238 0.000325 0.085378
PWY-6122: 5-aminoimidazole ribonucleotide biosynthesis II SH2D7 ‒0.97619 0.000326 0.08554
PWY-6277: super pathway of 5-aminoimidazole ribonucleotide biosynthesis SH2D7 ‒0.97619 0.000326 0.08554
P461-PWY: hexitol fermentation to lactate, formate, ethanol and acetate MT1X ‒0.85714 7.8E–05 0.022737 6
P461-PWY: hexitol fermentation to lactate, formate, ethanol, and acetate MYBL1 ‒0.90476 0.000309 0.081754
P461-PWY: hexitol fermentation to lactate, formate, ethanol, and acetate NTRK2 ‒0.95238 0.000321 0.084551
s__Clostridium_bartlettii IL17F 0.76835 0.000202 0.059482 7
s__Veillonella_unclassified IL17F 0.76835 2.91E–05 0.011329
s__Clostridium_bartlettii RN7SL301P 0.76835 0.00012 0.038644
s__Veillonella_unclassified RN7SL301P 0.76835 7.23E–05 0.025955
s__Eggerthella_lenta Vd2 0.89822 0.000353 0.054394 8
s__Flavonifractor_plautii Vd2 0.785714 0.000613 0.072663
s__Eggerthella_lenta RP11-707O23.5 0.76835 1.96E–06 0.001075
s__Flavonifractor_plautii RP11-707O23.5 0.763763 0.000134 0.04259
s__Eubacterium_siraeum Vd2T_IL22_only 0.814386 0.000264 0.048712 9
s__Eubacterium_siraeum IL17C 0.76835 7.73E–06 0.003364
s__Subdoligranulum_unclassified IRX2 0.754505 0.000291 0.083959 10
s__Subdoligranulum_unclassified PAX8-AS1 ‒0.83333 0.000316 0.087851
s__Roseburia_hominis C5orf17 0.904762 0.000139 0.043995 11
s__Roseburia_hominis RP11-726G1.1 0.904762 0.000122 0.038958
GALACTUROCAT-PWY: D-galacturonate degradation I GHR 0.928571 0.000275 0.073927 12
PWY-6507: 4-deoxy-L-threo-hex-4-enopyranuronate degradation GHR 0.97619 5.83E–05 0.017377
CITRULBIO-PWY: L-citrulline biosynthesis PCK1 0.904762 0.00033 0.086443 13
PWY-4984: urea cycle PCK1 0.904762 0.000355 0.092057
PWY-6305: putrescine biosynthesis IV CXCL8 0.97619 0.000123 0.034844 14
PWY-6305: putrescine biosynthesis IV IGHV4-31 0.928571 0.000159 0.044272
FAO-PWY: fatty acid β-oxidation I IGKV1D-16 0.946125 0.000305 0.080971 15
PWY-5136: fatty acid β-oxidation II (peroxisome) IGKV1D-16 0.952381 7.44E–05 0.022051
P42-PWY: incomplete reductive TCA cycle IGLV7-46 ‒0.97619 0.000353 0.091828 16
PWY-5104: L-isoleucine biosynthesis IV IGLV7-46 ‒0.95238 0.000101 0.029049
s__Lachnospiraceae_bacterium_1_4_56FAA CD8_IL22_only 0.809524 0.000137 0.036152 17
s__Lachnospiraceae_bacterium_1_4_56FAA Teff_TNFa_INFg 0.718576 6.25E–05 0.020997
PWY-5189: tetrapyrrole biosynthesis II (from glycine) CA1 ‒0.78571 0.000311 0.082075 18
PWY-5189: tetrapyrrole biosynthesis II (from glycine) CD274 0.928571 0.000299 0.079439
FOLSYN-PWY: super pathway of tetrahydrofolate biosynthesis and salvage TMEM37 0.934148 3.07E–06 0.001317 19
PWY-6612: superpathway of tetrahydrofolate biosynthesis TMEM37 0.904762 4.47E–06 0.001739
Treg_IL17_only FCRL1 ‒0.97619 6.99E–05 0.041248 20
Treg_IL17_only FCRLA ‒1 0.000158 0.087172
PWY-6969: TCA cycle V (2-oxoglutarate:ferredoxin oxidoreductase) REG1A ‒0.76376 0.000365 0.094544 21
s__Bacteroides_xylanisolvens REG1A 0.736989 0.000162 0.048778
GLCMANNANAUT-PWY: super pathway of N-acetylglucosamine, N-acetylmannosamine, and N-acetylneuraminate degradation C11orf86 ‒0.83333 5.42E–06 0.002104 22
GLUCONEO-PWY: gluconeogenesis I IGLV3-10 ‒0.7619 0.000271 0.073002 23
s__Ruminococcus_bromii IGHM 0.904762 1.28E–05 0.005526 24
PWY-621: sucrose degradation III (sucrose invertase) CLCA4 ‒0.97619 1.29E–05 0.00476 25
PWY-2941: L-lysine biosynthesis II NLGN4Y 0.912328 0.000273 0.073524 26
PHOSLIPSYN-PWY: super pathway of phospholipid biosynthesis I (bacteria) IGLV5-37 ‒0.90476 0.000222 0.060934 27
UniRef90_B0GA46: NO_NAME CD8_TNFa_IFNg 0.8485 2.77E–06 0.088269 28
TRNA-CHARGING-PWY: tRNA charging CHIT1 ‒0.92857 1.07E–05 0.004046 29
RHAMCAT-PWY: L-rhamnose degradation I DZ_duration 0.886243 0.00443 0.078996 30
s__Clostridium_bolteae Active.Teff ‒0.87427 0.000195 0.092483 31
PWY0-1297: superpathway of purine deoxyribonucleosides degradation MYEOV 0.904762 3.78E–05 0.011407 32
NONOXIPENT-PWY: pentose phosphate pathway (non-oxidative branch) CLIC6 0.904762 0.000182 0.050256 33
PWY-3001: super pathway of L-isoleucine biosynthesis I CIDEC ‒0.88095 0.000207 0.056999 34
s__Bilophila_wadsworthia NLRP2 ‒0.95238 4.72E–05 0.018207 35
PWY-5005: biotin biosynthesis II KRT8P36 0.922172 0.000142 0.040129 36
PWY0-1061: superpathway of L-alanine biosynthesis RP11-575F12.2 0.970077 0.000254 0.068804 37
PWY0-1296: purine ribonucleosides degradation SULT1C2 ‒0.97619 0.000106 0.030556 38
MET-SAM-PWY: super pathway of S-adenosyl-L-methionine biosynthesis FRMD1 ‒0.80952 0.000175 0.048401 39
LACTOSECAT-PWY: lactose and galactose degradation I IGHV1-3 0.952381 0.000237 0.064542 40
s__Bilophila_unclassified RPS6KA2-AS1 ‒1 1.82E–05 0.007734 41
PWY-7383: anaerobic energy metabolism (invertebrates, cytosol) IGLV4-60 ‒0.92857 0.000302 0.080129 42
HEME-BIOSYNTHESIS-II: heme biosynthesis I (aerobic) RP11-958N24.2 0.788009 0.000292 0.077896 43
PWY-7234: inosine-5-phosphate biosynthesis III HERC2P4 ‒0.92857 0.000167 0.0464 44
PWY0-162: super pathway of pyrimidine ribonucleotides de novo biosynthesis ARL14 ‒0.97008 6.97E–05 0.020733 45
PWY-7229: superpathway of adenosine nucleotides de novo biosynthesis I CTAGE15 0.857143 0.000298 0.079314 46
Vd2T_IFNg_only IGLC7 0.880952 0.000167 0.08878 47
PWY-6897: thiamin salvage II MME-AS1 0.76835 0.000346 0.09024 48
PWY-5030: L-histidine degradation III AC131056.3 0.952381 0.000379 0.097971 49
s__Eubacterium_eligens RP11-259G18.3 ‒0.95181 6.08E–05 0.022759 50
Treg_IL17_IL22 IGKV1D-43 0.730552 6.75E–05 0.041248 51
s__Lachnospiraceae_bacterium_7_1_58FAA ZBTB16 0.928571 0.000347 0.095663 52
s__Coprococcus_catus THNSL2 1 5.97E–05 0.02245 53
s__Erysipelotrichaceae_bacterium_6_1_45 PTGS2 ‒0.83333 0.000359 0.098475 54
s__Bifidobacterium_longum Vd2T_TNFa_only ‒0.91573 0.000601 0.072663 55
List of significant associations in PSO3 associated network
P122-PWY: heterolactic fermentation EDDM3DP 0.664211 1.49E–05 0.022213 1
P122-PWY: heterolactic fermentation RP11-100G15.10 0.664211 1.49E–05 0.022213
PWY-5083: NAD/NADH phosphorylation and dephosphorylation EDDM3DP 0.695608 4.42E–07 0.001001
PWY-5083: NAD/NADH phosphorylation and dephosphorylation RP11-100G15.10 0.695608 4.42E–07 0.001001
PWY-821: super pathway of sulfur amino acid biosynthesis (Saccharomyces cerevisiae) RP11-100G15.10 0.695608 7.39E–05 0.070103
PWY-821: super pathway of sulfur amino acid biosynthesis (Saccharomyces cerevisiae) EDDM3DP 0.695608 7.39E–05 0.070103
PWY0-1061: superpathway of L-alanine biosynthesis RP11-100G15.10 0.654654 9.11E–05 0.081919
PWY0-1061: superpathway of L-alanine biosynthesis EDDM3DP 0.654654 9.11E–05 0.081919
s__Bacteroides_xylanisolvens EDDM3DP 0.654654 0.000129 0.063831
s__Bacteroides_xylanisolvens RP11-100G15.10 0.654654 0.000129 0.063831
s__Haemophilus_parainfluenzae EDDM3DP 0.654654 3.59E–07 0.000407
s__Haemophilus_parainfluenzae RP11-100G15.10 0.654654 3.59E–07 0.000407
PWY-5188: tetrapyrrole biosynthesis I (from glutamate) MYBL1 ‒1 1.56E–05 0.022439 2
PWY-5188: tetrapyrrole biosynthesis I (from glutamate) PAX5 ‒1 4.29E–05 0.046051
PWY-5188: tetrapyrrole biosynthesis I (from glutamate) AFF2 ‒1 1.32E–05 0.020625
PWY-5188: tetrapyrrole biosynthesis I (from glutamate) TCL1A ‒0.88571 1.28E–05 0.020213
PWY-5188: tetrapyrrole biosynthesis I (from glutamate) CD79B ‒0.88571 3.53E–05 0.040084
PWY-5345: superpathway of L-methionine biosynthesis (by sulfhydrylation) PWP2 ‒0.94286 1.25E–05 0.019878 3
PWY-5345: super pathway of L-methionine biosynthesis (by sulfhydrylation) IGKV1-9 ‒0.94286 8.57E–05 0.078976
SULFATE-CYS-PWY: super pathway of sulfate assimilation and cysteine biosynthesis IGKV1-9 ‒0.89865 8.22E–05 0.076628
COBALSYN-PWY: adenosylcobalamin salvage from cobinamide I ATP13A5 ‒1 8.76E–05 0.079978 4
DTDPRHAMSYN-PWY: dTDP-L-rhamnose biosynthesis I ATP13A5 ‒1 3.25E–05 0.037635
DTDPRHAMSYN-PWY: dTDP-L-rhamnose biosynthesis I RGS13 ‒0.94286 6.46E–05 0.063204
s__Roseburia_intestinalis IL36RN 0.845154 7.21E–05 0.042737 5
s__Roseburia_intestinalis SLC44A5 0.771429 0.000123 0.061427
s__Roseburia_intestinalis RP11-313J2.1 ‒0.82857 6.98E–05 0.041714
s__Lachnospiraceae_bacterium_7_1_58FAA RP11-462B18.1 0.654654 0.000174 0.083495 6
s__Lachnospiraceae_bacterium_7_1_58FAA TM4SF4 0.885714 3.85E–05 0.024936
s__Lachnospiraceae_bacterium_7_1_58FAA IGHV3-66 ‒1 6.57E–06 0.005182
GOLPDLCAT-PWY: superpathway of glycerol degradation to 1,3-propanediol NARF ‒1 0.000113 0.097624 7
GOLPDLCAT-PWY: superpathway of glycerol degradation to 1,3-propanediol PNLIPRP2 ‒1 4.67E–05 0.04818
MET-SAM-PWY: superpathway of S-adenosyl-L-methionine biosynthesis TMEM150B (DRAM-3 regulate autophagy) 1 3.53E–05 0.040084 8
PWY-5347: superpathway of L-methionine biosynthesis (transsulfuration) TMEM150B 1 8.06E–05 0.075717
FAO-PWY: fatty acid β-oxidation I TEKT4P2 ‒1 2.74E–05 0.035602 9
PWY-5136: fatty acid β-oxidation II (peroxisome) TEKT4P2 ‒1 3.71E–05 0.040923
UniRef90_A7B4W0: NO_NAME AQP5 0.970588 4.31E–07 0.015343 10
UniRef90_D6DY27: ABC-type antimicrobial peptide transport system, ATPase component AQP5 0.955882 1.67E–06 0.057497
UniRef90_C6JCY5: NO_NAME IGLV4-60 ‒0.98561 2.08E–06 0.070312 11
NAD-BIOSYNTHESIS-II: NAD salvage pathway II NLGN4Y 0.941124 7.1E–05 0.068116 12
CALVIN-PWY: Calvin-Benson-Bassham cycle HBEGF 1 7.11E–05 0.068116 13
GLUCONEO-PWY: gluconeogenesis I RP11-575F12.3 1 7.47E–06 0.013124 14
PWY-5097: L-lysine biosynthesis VI RP11-30P6.6 ‒1 4.54E–05 0.047844 15
PYRIDNUCSYN-PWY: NAD biosynthesis I (from aspartate) RP11-747H7.3 1 5.69E–05 0.055963 16
UniRef90_G1WWG9: NO_NAME CD55 1 2.77E–06 0.090568 17
PWY-5005: biotin biosynthesis II TDRD1 0.941124 8.24E–05 0.076628 18
OANTIGEN-PWY: O-antigen building blocks biosynthesis (E. coli) ISG15 1 4.61E–05 0.048107 19
UniRef90_D4L1Y1: Site-specific recombinases, DNA invertase Pin homologs SERPINA9 ‒1 1.8E–08 0.000665 20
PWY-2941: L-lysine biosynthesis II PI15 0.828571 4.83E–05 0.049266 21
PWY4LZ-257: superpathway of fermentation (Chlamydomonas reinhardtii) IGLC7 ‒0.94286 8.84E–05 0.080307 22
UniRef90_D4KBZ1: Ribonuclease H HRCT1 ‒1 2.95E–06 0.094916 23
UniRef90_C7H3L3: NO_NAME FOSB ‒0.95618 1.65E–06 0.057497 24
CRNFORCAT-PWY: creatinine degradation I VWA5B1 1 5.56E–06 0.010772 25
s__Anaerostipes_hadrus RP11-424G14.1 0.845154 2.53E–07 0.000295 26

Abbreviations: adj, adjusted; Cor, correlation value.

Supplementary Table S12.

Available Clinical and Experimental Data

PID Status Gender Age PASI PSO onset Disease
Duration
Metagenomics
Data (Stool)
Host Transcriptomic
Data (Sigmoid)
Flow Cytometry
Data (PBMCs)
PSO Subgroups
Identified
6532 Healthy M 61 NA NA NA Yes Yes Yes Healthy
7300 PSO F 58 5.9 18 37 Yes Yes Yes PSO2
7301 PSO M 37 27.8 24 10 Yes Yes Yes PSO2
7303 PSO F 35 NA 18 14 Yes Yes Yes PSO2
7304 PSO F 41 1.2 19 19 Yes Yes Yes PSO2
7305 PSO M 58 19.4 33 22 Yes Yes Yes PSO1
7306 PSO M 70 2.1 20 47 Yes Yes Yes PSO1
7307 PSO M 58 15.6 50 5 Yes Yes Yes PSO3
7308 PSO F 60 5.2 48 10 Yes Yes Yes PSO1
7309 PSO F 29 0.2 10 16 Yes Yes Yes PSO3
7310 PSO M 54 7.7 44 10 Yes Yes Yes PSO1
7311 PSO M 58 5.6 20 47 Yes Yes Yes PSO2
7312 PSO M 39 4.3 10 26 Yes Yes Yes PSO1
7313 PSO F 76 7.5 60 14 Yes Yes Yes PSO2
7314 PSO M 29 4 7 19 Yes Yes Yes PSO3
7315 PSO M 55 3.9 42 10 Yes Yes Yes PSO1
7317 PSO F 28 2.6 10 15 Yes Yes Yes PSO1
7320 PSO F 60 13.2 29 32 Yes Yes Yes PSO3
7322 PSO F 42 3.4 10 32 Yes Yes Yes PSO1
7323 PSO F 53 31.1 22 31 Yes Yes Yes PSO3
7324 PSO M 57 7.8 55 2 Yes Yes Yes PSO1
7325 PSO F 22 27.8 15 7 Yes Yes Yes PSO1
7327 PSO F 29 11.3 6 23 Yes Yes Yes PSO2
7328 PSO F 28 13.2 8 20 Yes Yes Yes PSO2
7330 PSO F 28 16.1 16 12 Yes Yes Yes PSO1
7331 PSO F 29 18.7 12 17 Yes Yes Yes PSO3
7332 PSO M 28 19.8 24 4 Yes Yes Yes PSO1
7352 Healthy F 60 NA NA NA Yes No No Healthy
7353 Healthy M 25 NA NA NA Yes Yes Yes Healthy
7355 Healthy F 23 NA NA NA Yes Yes Yes Healthy
7358 Healthy F 69 NA NA NA Yes Yes Yes Healthy
7360 Healthy F 38 NA NA NA Yes Yes Yes Healthy
7364 Healthy M 53 NA NA NA Yes Yes Yes Healthy
7365 Healthy F 57 NA NA NA Yes Yes Yes Healthy
7368 Healthy F 49 NA NA NA Yes Yes Yes Healthy
7369 Healthy M 38 NA NA NA Yes Yes Yes Healthy
7371 Healthy M 47 NA NA NA Yes Yes Yes Healthy
7372 Healthy M 59 NA NA NA Yes Yes Yes Healthy
7374 Healthy M 35 NA NA NA Yes Yes Yes Healthy
7375 Healthy M 36 NA NA NA Yes Yes Yes Healthy
7376 Healthy F 37 NA NA NA Yes Yes Yes Healthy
N1 PSO M 52 23.9 27 25 Yes No No PSO3
N11 PSO F 31 31.9 5 16 Yes No No PSO3
N2 PSO M 49 8.8 26 23 Yes No No PSO1
N3 PSO M 37 22.7 27 10 Yes No No PSO1
N4 PSO M 32 66.6 25 7 Yes No No PSO1
N6 PSO M 35 6.8 35 1 Yes No No PSO2
N9 PSO F 29 18.7 27 2 Yes No No PSO3

Abbreviations: F, female; M, male; NA, not available; PSO, psoriasis.

References

  1. Afifi L., Danesh M.J., Lee K.M., Beroukhim K., Farahnik B., Ahn R.S., et al. Dietary behaviors in psoriasis: patient-reported outcomes from a U.S. National survey. Dermatol Ther (Heidelb) 2017;7:227–242. doi: 10.1007/s13555-017-0183-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Arumugam M., Raes J., Pelletier E., Le Paslier D., Yamada T., Mende D.R., et al. Enterotypes of the human gut microbiome [published corrections appear in Nature 2014;506:516 and Nature 2011;474:666] Nature. 2011;473:174–180. doi: 10.1038/nature09944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Benhadou F., Mintoff D., Schnebert B., Thio H.B. Psoriasis and microbiota: a systematic review. Diseases. 2018;6:47. doi: 10.3390/diseases6020047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Brembilla N.C., Senra L., Boehncke W.-H. The IL-17 Family of cytokines in psoriasis: IL-17A and beyond. Front Immunol. 2018;9:1682. doi: 10.3389/fimmu.2018.01682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Chang H.W., Yan D., Singh R., Liu J., Lu X., Ucmak D., et al. Alteration of the cutaneous microbiome in psoriasis and potential role in Th17 polarization. Microbiome. 2018;6:154. doi: 10.1186/s40168-018-0533-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Codoñer F.M., Ramírez-Bosca A., Climent E., Carrión-Gutierrez M., Guerrero M., Pérez-Orquín J.M., et al. Gut microbial composition in patients with psoriasis. Sci Rep. 2018;8:3812. doi: 10.1038/s41598-018-22125-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Diani M., Casciano F., Marongiu L., Longhi M., Altomare A., Pigatto P.D., et al. Increased frequency of activated CD8+ T cell effectors in patients with psoriatic arthritis. Sci Rep. 2019;9:10870. doi: 10.1038/s41598-019-47310-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. El Mouzan M.I., Winter H.S., Assiri A.A., Korolev K.S., Al Sarkhy A.A., Dowd S.E., et al. Microbiota profile in new-onset pediatric Crohn’s disease: data from a non-Western population. Gut Pathog. 2018;10:49. doi: 10.1186/s13099-018-0276-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Elder J.T., Bruce A.T., Gudjonsson J.E., Johnston A., Stuart P.E., Tejasvi T., et al. Molecular dissection of psoriasis: integrating genetics and biology. J Invest Dermatol. 2010;130:1213–1226. doi: 10.1038/jid.2009.319. [DOI] [PubMed] [Google Scholar]
  10. Eppinga H., Sperna Weiland C.J., Thio H.B., van der Woude C.J., Nijsten T.E.C., Peppelenbosch M.P., et al. Similar depletion of protective Faecalibacterium prausnitzii in psoriasis and inflammatory bowel disease, but not in hidradenitis suppurativa. J Crohns Colitis. 2016;10:1067–1075. doi: 10.1093/ecco-jcc/jjw070. [DOI] [PubMed] [Google Scholar]
  11. Fahlén A., Engstrand L., Baker B.S., Powles A., Fry L. Comparison of bacterial microbiota in skin biopsies from normal and psoriatic skin. Arch Dermatol Res. 2012;304:15–22. doi: 10.1007/s00403-011-1189-x. [DOI] [PubMed] [Google Scholar]
  12. Franzosa E.A., McIver L.J., Rahnavard G., Thompson L.R., Schirmer M., Weingart G., et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat Methods. 2018;15:962–968. doi: 10.1038/s41592-018-0176-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Fu Y., Lee C.H., Chi C.C. Association of psoriasis with inflammatory bowel disease: a systematic review and meta-analysis. JAMA Dermatol. 2018;154:1417–1423. doi: 10.1001/jamadermatol.2018.3631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Fyhrquist N., Muirhead G., Prast-Nielsen S., Jeanmougin M., Olah P., Skoog T., et al. Microbe-host interplay in atopic dermatitis and psoriasis. Nat Commun. 2019;10:4703. doi: 10.1038/s41467-019-12253-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Gao Z., Tseng C.H., Strober B.E., Pei Z., Blaser M.J. Substantial alterations of the cutaneous bacterial biota in psoriatic lesions. PLoS One. 2008;3:e2719. doi: 10.1371/journal.pone.0002719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Gevers D., Kugathasan S., Denson L.A., Vázquez-Baeza Y., Van Treuren W., Ren B., et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15:382–392. doi: 10.1016/j.chom.2014.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Hidalgo-Cantabrana C., Gómez J., Delgado S., Requena-López S., Queiro-Silva R., Margolles A., et al. Gut microbiota dysbiosis in a cohort of patients with psoriasis. Br J Dermatol. 2019;181:1287–1295. doi: 10.1111/bjd.17931. [DOI] [PubMed] [Google Scholar]
  18. Hijnen D., Knol E.F., Gent Y.Y., Giovannone B., Beijn S.J.P., Kupper T.S., et al. CD8(+) T cells in the lesional skin of atopic dermatitis and psoriasis patients are an important source of IFN-γ, IL-13, IL-17, and IL-22. J Invest Dermatol. 2013;133:973–979. doi: 10.1038/jid.2012.456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Hsiao E.Y., McBride S.W., Hsien S., Sharon G., Hyde E.R., McCue T., et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155:1451–1463. doi: 10.1016/j.cell.2013.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. James K.R., Gomes T., Elmentaite R., Kumar N., Gulliver E.L., King H.W., et al. Distinct microbial and immune niches of the human colon. Nat Immunol. 2020;21:343–353. doi: 10.1038/s41590-020-0602-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Jin X., Chen D., Zheng R.H., Zhang H., Chen Y.P., Xiang Z. miRNA-133a-UCP2 pathway regulates inflammatory bowel disease progress by influencing inflammation, oxidative stress and energy metabolism. World J Gastroenterol. 2017;23:76–86. doi: 10.3748/wjg.v23.i1.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Knights D., Silverberg M.S., Weersma R.K., Gevers D., Dijkstra G., Huang H., et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med. 2014;6:107. doi: 10.1186/s13073-014-0107-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Levy M., Thaiss C.A., Zeevi D., Dohnalová L., Zilberman-Schapira G., Mahdi J.A., et al. Microbiota-modulated metabolites shape the intestinal microenvironment by regulating NLRP6 inflammasome signaling. Cell. 2015;163:1428–1443. doi: 10.1016/j.cell.2015.10.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Liu J., Chang H.W., Huang Z.M., Nakamura M., Sekhon S., Ahn R., et al. Single-cell RNA sequencing of psoriatic skin identifies pathogenic Tc17 cell subsets and reveals distinctions between CD8+ T cells in autoimmunity and cancer. J Allergy Clin Immunol. 2021;147:2370–2380. doi: 10.1016/j.jaci.2020.11.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Lloyd-Price J., Arze C., Ananthakrishnan A.N., Schirmer M., Avila-Pacheco J., Poon T.W., et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569:655–662. doi: 10.1038/s41586-019-1237-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Loesche M.A., Farahi K., Capone K., Fakharzadeh S., Blauvelt A., Duffin K.C., et al. Longitudinal study of the psoriasis-associated skin microbiome during therapy with ustekinumab in a randomized phase 3b clinical trial. J Invest Dermatol. 2018;138:1973–1981. doi: 10.1016/j.jid.2018.03.1501. [DOI] [PubMed] [Google Scholar]
  27. Love M.I., Huber W., Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Lozupone C., Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl Environ Microbiol. 2005;71:8228–8235. doi: 10.1128/AEM.71.12.8228-8235.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Matsuda H., Fujiyama Y., Andoh A., Ushijima T., Kajinami T., Bamba T. Characterization of antibody responses against rectal mucosa-associated bacterial flora in patients with ulcerative colitis. J Gastroenterol Hepatol. 2000;15:61–68. doi: 10.1046/j.1440-1746.2000.02045.x. [DOI] [PubMed] [Google Scholar]
  30. McMurdie P.J., Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One. 2013;8:e61217. doi: 10.1371/journal.pone.0061217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. McMurdie P.J., Holmes S. Waste not, want not: why rarefying microbiome data is inadmissible. PLoS Comput Biol. 2014;10 doi: 10.1371/journal.pcbi.1003531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Morgan X.C., Tickle T.L., Sokol H., Gevers D., Devaney K.L., Ward D.V., et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13:R79. doi: 10.1186/gb-2012-13-9-r79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Newman A.M., Steen C.B., Liu C.L., Gentles A.J., Chaudhuri A.A., Scherer F., et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat Biotechnol. 2019;37:773–782. doi: 10.1038/s41587-019-0114-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Noble C.L., Abbas A.R., Lees C.W., Cornelius J., Toy K., Modrusan Z., et al. Characterization of intestinal gene expression profiles in Crohn’s disease by genome-wide microarray analysis. Inflamm Bowel Dis. 2010;16:1717–1728. doi: 10.1002/ibd.21263. [DOI] [PubMed] [Google Scholar]
  35. Panagiotakos D.B., Pitsavos C., Stefanadis C. Dietary patterns: A Mediterranean diet score and its relation to clinical and biological markers of cardiovascular disease risk. Nutr Metab Cardiovasc Dis. 2006;16:559–568. doi: 10.1016/j.numecd.2005.08.006. [DOI] [PubMed] [Google Scholar]
  36. Prodanovich S., Kirsner R.S., Kravetz J.D., Ma F., Martinez L., Federman D.G. Association of psoriasis with coronary artery, cerebrovascular, and peripheral vascular diseases and mortality. Arch Dermatol. 2009;145:700–703. doi: 10.1001/archdermatol.2009.94. [DOI] [PubMed] [Google Scholar]
  37. Rais R., Jiang W., Zhai H., Wozniak K.M., Stathis M., Hollinger K.R., et al. FOLH1/GCPII is elevated in IBD patients, and its inhibition ameliorates murine IBD abnormalities. JCI Insight. 2016;1:e88634. doi: 10.1172/jci.insight.88634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Ritchlin C.T., Colbert R.A., Gladman D.D. Psoriatic arthritis [published correction appears in N Engl J Med 2017;376:2097] N Engl J Med. 2017;376:957–970. doi: 10.1056/NEJMra1505557. [DOI] [PubMed] [Google Scholar]
  39. Scher J.U., Ubeda C., Artacho A., Attur M., Isaac S., Reddy S.M., et al. Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis Rheumatol. 2015;67:128–139. doi: 10.1002/art.38892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Selhub J., Byun A., Liu Z., Mason J.B., Bronson R.T., Crott J.W. Dietary vitamin B6 intake modulates colonic inflammation in the IL10−/− model of inflammatory bowel disease. J Nutr Biochem. 2013;24:2138–2143. doi: 10.1016/j.jnutbio.2013.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Shajib M.S., Chauhan U., Adeeb S., Chetty Y., Armstrong D., Halder S.L.S., et al. Characterization of serotonin signaling components in patients with inflammatory bowel disease. J Can Assoc Gastroenterol. 2019;2:132–140. doi: 10.1093/jcag/gwy039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Takemoto A., Cho O., Morohoshi Y., Sugita T., Muto M. Molecular characterization of the skin fungal microbiome in patients with psoriasis. J Dermatol. 2015;42:166–170. doi: 10.1111/1346-8138.12739. [DOI] [PubMed] [Google Scholar]
  43. Tan L., Zhao S., Zhu W., Wu L., Li J., Shen M., et al. The Akkermansia muciniphila is a gut microbiota signature in psoriasis. Exp Dermatol. 2018;27:144–149. doi: 10.1111/exd.13463. [DOI] [PubMed] [Google Scholar]
  44. Tett A., Pasolli E., Farina S., Truong D.T., Asnicar F., Zolfo M., et al. Unexplored diversity and strain-level structure of the skin microbiome associated with psoriasis. NPJ Biofilms Microbiomes. 2017;3:14. doi: 10.1038/s41522-017-0022-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Tibshirani R., Hastie T. Hastie. Estimating the number of clusters in a data set via the gap statistic. Royal Stat Soc. 2002;457 doi: 10.1111/1467-9868.00293. [DOI] [Google Scholar]
  46. Turnbaugh P.J., Hamady M., Yatsunenko T., Cantarel B.L., Duncan A., Ley R.E., et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–484. doi: 10.1038/nature07540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Wan M.T., Shin D.B., Hubbard R.A., Noe M.H., Mehta N.N., Gelfand J.M. Psoriasis and the risk of diabetes: a prospective population-based cohort study. J Am Acad Dermatol. 2018;78:315–322.e1. doi: 10.1016/j.jaad.2017.10.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Wang F., Kaplan J.L., Gold B.D., Bhasin M.K., Ward N.L., Kellermayer R., et al. Detecting microbial dysbiosis associated with pediatric Crohn disease despite the high variability of the gut microbiota. Cell Rep. 2016;14:945–955. doi: 10.1016/j.celrep.2015.12.088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Yan D., Issa N., Afifi L., Jeon C., Chang H.W., Liao W. The role of the skin and gut microbiome in psoriatic disease. Curr Dermatol Rep. 2017;6:94–103. doi: 10.1007/s13671-017-0178-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Yoshii K., Hosomi K., Sawane K., Kunisawa J. Metabolism of dietary and microbial vitamin B family in the regulation of host immunity. Front Nutr. 2019;6:48. doi: 10.3389/fnut.2019.00048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Yu X., Wieczorek S., Franke A., Yin H., Pierer M., Sina C., et al. Association of UCP2 -866 G/A polymorphism with chronic inflammatory diseases. Genes Immun. 2009;10:601–605. doi: 10.1038/gene.2009.29. [DOI] [PubMed] [Google Scholar]
  52. Zhou W., Sailani M.R., Contrepois K., Zhou Y., Ahadi S., Leopold S.R., et al. Longitudinal multi-omics of host–microbe dynamics in prediabetes. Nature. 2019;569:663–671. doi: 10.1038/s41586-019-1236-x. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Data Availability Statement

Expression data have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus and is accessible through Gene Expression Omnibus Series accession number GSE150851. Metagenomics sequence data are accessible through Sequence Read Archive BioProject PRJNA634145. All clinical and experimental data available are shown in Supplementary Table S12.


Articles from JID Innovations are provided here courtesy of Elsevier

RESOURCES