You are on page 1of 12

Journal of Autoimmunity 137 (2023) 103055

Contents lists available at ScienceDirect

Journal of Autoimmunity
journal homepage: www.elsevier.com/locate/jautimm

Gut microbial signatures and their functions in Behcet’s uveitis and


Vogt-Koyanagi-Harada disease
Qingfeng Wang a, 1, Shuang Wu b, 1, Xingsheng Ye a, Shiyao Tan a, Fanfan Huang a, Guannan Su a,
Aize Kijlstra c, Peizeng Yang a, *
a
The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing Branch (Municipality
Division) of National Clinical Research Center for Ocular Diseases, Chongqing, People’s Republic of China
b
Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University, Nanning, People’s
Republic of China
c
University Eye Clinic Maastricht, Maastricht, the Netherlands

A R T I C L E I N F O A B S T R A C T

Handling Editor: M.E. Gershwin Background: A number of public metagenomic studies reveal an association between the gut microbiome and
various immune-mediated diseases including Behcet’s uveitis (BU) and Vogt-Koyanagi-Harada disease (VKH).
Keywords: Integrated-analysis and subsequent validation of these results could be a potentially powerful way to understand
Gut microbiome the microbial signatures and their functions in these two uveitis entities.
Behcet’s uveitis (BU)
Methods: We integrated the sequencing data of our previous metagenomic studies on two major uveitis entities,
Vogt-Koyanagi-Harada disease (VKH)
BU and VKH as well as four other publicly available immune-mediated diseases datasets, including Ankylosing
Metagenomic sequencing
Immune-mediated disease Spondylitis (AS), Rheumatoid Arthritis (RA), Crohn’s disease (CD) and Ulcerative Colitis (UC). Alpha-diversity
and beta-diversity analysis were used to compare the gut microbiome signatures between both uveitis entities
and other immune-mediated diseases and healthy controls. Amino acid homology between microbial proteins
and a uveitogenic peptide of the interphotoreceptor retinoid-binding protein (IRBP)161-180 was investigated using
a similarity search in the NCBI protein BLAST program (BLASTP). Enzyme-linked Immunosorbent Assay (ELISA)
was performed to evaluate the cross-reactive responses of experimental autoimmune uveitis (EAU)-derived
lymphocytes and BU patients-derived peripheral blood mononuclear cells (PBMCs) against homologous peptides.
The area under the curve (AUC) analysis was used to test the sensitivity and specificity of gut microbial
biomarkers.
Results: Depleted Dorea, Blautia, Coprococcus, Erysipelotrichaceae and Lachnospiraceae as well as enriched
Bilophila and Stenotrophomonas were identified in BU patients. An enriched Alistipes along with a lower level of
Dorea were observed in VKH patients. A peptide antigen (SteTDR) encoded by BU specifically enriched Steno­
trophomonas was identified to share homology with IRBP161-180. In vitro experiments showed that lymphocytes
from EAU or PBMCs from BU patients reacted to this peptide antigen as shown by the production of IFN-γ and IL-
17. Addition of the SteTDR peptide to the classical IRBP immunization protocol exacerbated EAU severity. Gut
microbial marker profiles consisted of 24 species and 32 species respectively differentiated BU and VKH from
each other as well as from the other four immune-mediated diseases and healthy controls. Protein annotation
identified 148 and 119 specific microbial proteins associated with BU and VKH, respectively. For metabolic
function analysis, 108 and 178 metabolic pathways were shown to be associated with BU and VKH, respectively.
Conclusions: Our study revealed specific gut microbial signatures and their potentially functional roles in BU and
VKH pathogenesis that differ significantly from other immune-mediated diseases as well as healthy controls.

Abbreviations: BU, Behcet’s uveitis; VKH, Vogt-Koyanagi-Harada disease; RA, Rheumatoid Arthritis; CD, Crohn’s disease; UC, Ulcerative Colitis; IRBP, inter­
photoreceptor retinoid-binding protein; EAU, experimental autoimmune uveitis.
* Corresponding author. The First Affiliated Hospital of Chongqing Medical University, Youyi Road 1, Chongqing, 400016, People’s Republic of China
E-mail address: peizengycmu@126.com (P. Yang).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.jaut.2023.103055
Received 27 February 2023; Received in revised form 27 April 2023; Accepted 3 May 2023
Available online 18 May 2023
0896-8411/© 2023 Elsevier Ltd. All rights reserved.
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

1. Introduction Table 1
The studies included in the intergated-analysis.
Uveitis is an intraocular inflammatory disease which can lead to Diseases Sample Populations PMID
serious visual impairment worldwide [1]. Although clinical features size
vary significantly among different uveitis entities, several risk factors BU 24 China 30077182
such as genetic, environmental, infectious and immunological factors VKH 71 China 31928124
have been shown to play an important role in their pathogenesis [2]. CD 86 American and 30531976
Recently evidence is emerging concerning the role of the gut micro­ Netherland
UC 65 American and 30531976
biome in the pathogenesis of uveitis [3]. Netherland
The gut microbiome plays an important role in modulating host AS 51 China 28750650
immune homeostasis [4,5]. Dysbiosis of the gut microbiota has been RA 95 China 26214836
found in a series of immune-mediated diseases including Ankylosing HC 303 China 26214836, 28750650,
30077182, 31928124
Spondylitis (AS), Rheumatoid Arthritis (RA), Crohn’s disease (CD) and
Ulcerative Colitis (UC) using metagenomic sequence (MGS) [6–8]. BU, Behcet’s uveitis; VKH, Vogt-Koyanagi-Harada disease; CD, Crohn’s disease,
Studies dealing with the pathogenesis of these immune-mediated dis­ UC, ulcerative colitis; AS, Ankylosing spondylitis; RA, rheumatoid arthritis, HC,
eases have suggested that the gut microbiome contributes to immune healthy controls; PMID, PubMed unique identifier.
disorders via production of homologous peptides, effector proteins and
metabolites [9–11]. In our previous metagenomic studies, we also as compared to other immune-mediated diseases. Although public
showed that gut microbiome composition was associated with Behcet’s metagenomic sequence data from different studies differed in their li­
uveitis (BU) and Vogt-Koyanagi-Harada disease (VKH), which are brary sizes (number of reads), read length, and how they were quality
common non-infectious uveitis entities [12,13]. BU is an auto­ controlled, the variations caused by these technical factors is relatively
inflammatory disease causing multisystemic inflammation in various small in contrast to biological variation as described in a previous study
organs including the eye (uveitis) and skin (lesions and ulcers) [14]. [21], suggesting that these variations will not influence our analysis.
VKH is different from BU and is commonly considered as an autoim­
mune disease directed against melanocyte associated antigens and 2.2. Taxonomical annotation and profiling
which is characterized by bilateral granulomatous panuveitis with sys­
temic manifestations, such as tinnitus, vertigo and meningism [15]. Raw sequence data (BioProject accession number PRJNA356102,
Several studies identified similarities and differences concerning the PRJNA431482, PRJNA400072, PRJNA375935, PRJNA356225) were
pathogenetic mechanisms between BU, VKH and various downloaded from SRA (https://www.ncbi.nlm.nih.gov/Traces/st
immune-mediated diseases including AS, RA, CD and UC [16–19]. udy/?) and then converted to fastq files by using the fastq-dump func­
However, few studies have focused on the differences of the gut tion from the NCBI SRA Toolkit (https://github.com/ncbi/sratoolkit).
microbiome composition between these diseases and this was therefore Human genes, sequences of poor quality (mean quality <20) and short
the subject of the study presented here. With a number of metagenomic sequences (reads <80 bp) from raw reads were filtered by using the
datasets, publicly available in the NCBI sequence read archive (SRA) and program KneadData (0.6.1), which combined Trimmomatic (0.39) and
the platform of MICrobiome HELminth INteractions database Bowtie2 (2.3.5) [22,23]. MetaPhlan was used for enhanced meta­
(MICHELINdb) [20], this allowed comparative analyses of the meta­ genomic taxonomic profiling. The composition (Bacteria, Archaea, Eu­
genomic studies on BU and VKH with other immune-mediated diseases. karyotes and Viruses) and gene abundance profile of microbial
In this study, we integrated the sequencing data of five metagenomic taxonomic units was profiled using MetaPhlan2 (v2.7.2) on the
studies and identified the gut microbial signatures in two separate post-processed reads [24]. Using MetaPhlan2, the annotation rate at
uveitis entities (BU and VKH). Moreover, we identified gut microbial species and strain level in this study were 92.56% ± 6.84% and 46.27%
markers and the functional consequences for these two uveitis entities ± 23.39% in the different groups, respectively.
based on their specific microbial signatures.
2.3. Alpha- and beta-diversity
2. Methods
Gut microbiome composition data of each sample from MetaPhlan2
2.1. Datasets was merged and converted to an spf file with predicted read counts by
the Python-based tool included merge_metaphlan_tables.py and meta­
To conduct the comparative analyses of the metagenomic studies on phlan_to_stamp.pl [24]. Shannon diversity index was calculated based
BU and VKH with other immune-mediated diseases, we obtained the on genera and species abundance profiles using the R package (https://
public metagenomic sequence data from 695 fecal metagenomic sam­ github.com/const-ae/ggsignif). To identify genera and species showing
ples across 5 studies and 3 populations from the NCBI SRA [6–8,12,13] different abundances in uveitis and other immune-mediated diseases as
(Table 1). The datasets included the fecal metagenomic samples of VKH, well as healthy controls, the spf file was analyzed by the Statistical
BU, CD, UC, RA and AS patients as well as healthy controls. The datasets Analysis of Metagenomic Profiles tool (STAMP). The
of VKH, BU, AS and RA were from four different Chinese metagenomic Benjamini-Hochberg procedure was used to control the false discovery
studies. CD and UC datasets were from one metagenomic study con­ rate due to multiple testing. False discovery rate (FDR) correction lower
taining patient samples from the Netherlands and USA. We only selected than 0.1 was identified as significant difference.
the active patients from the datasets of metagenomic studies on VKH and To compare the gut microbiome composition between diseases and
BU. For healthy controls, the controls from the dataset of CD and UC healthy controls, operational taxonomic units (OTU) at different levels
were excluded since non-CD and UC patients instead of healthy controls were separated from the spf files and used to compare the differences of
were included as controls in their study [6]. Furthermore, the healthy beta diversity (distance between samples based on differences in OTUs
controls samples from the four Chinese studies including BU, VKH, RA present in each sample) among different groups. Principal Component
and AS were combined for a larger sample size to compare the gut Analysis (PCA) was generated by using R package (https://github.com/
microbiome compositions between BU or VKH and healthy controls. YongxinLiu/MicrobiomeStatPlot/tree/master/223PCA) to identify
Besides the healthy controls, the non-uveitis patient samples including structural differences in the microbial community from different dis­
CD, UC, RA and AS patients from the datasets mentioned above were eases and healthy controls based on beta diversity distances. Con­
also used to further investigate the gut microbial features of BU or VKH strained PCoA analysis (CPCoA) was performed to visualize the

2
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

influence from the microbiome on the variants of different diseases and For cytokine secretion assays, cells (5 × 105/500 μl) were seeded into
healthy controls using the R package (https://github.com/YongxinLiu/ each well of a 48-well plate. The supernatants of PBMCs and lympho­
MicrobiomeStatPlot/tree/master/222CPCoA). cytes after stimulation with peptides were collected after 3 days. The
production of IFN-γ and IL-17 in the supernatants was quantified using
2.4. Linear discriminant analysis effect size Duoset ELISA development kits (R&D Systems, MN, USA) in accordance
with the manufacturer’s instructions. For statistical analysis, concen­
Linear discriminant analysis effect size (LEfSe), a method for trations below the detection limit were converted to a value of 50% of
biomarker discovery, was used to identify the microbial features of BU the lowest point of the calibration curve [28].
and VKH from other diseases and healthy controls. In this study, LEfSe
was performed by an online Galaxy web application (http://huttenhowe 2.9. Development of microbial markers
r.sph.harvard.edu/galaxy), in which the alpha value for Kruskal-Wallis
rank-sum test among classes was 0.5, and the threshold on the loga­ To establish the microbial markers for BU and VKH, extremely low
rithmic LDA score for discriminative features was 3.0 [25]. abundant taxa (mean relative abundance less than 0.001 in any group)
were removed as described previously [12]. The differentially abundant
2.5. IRBP161-180 peptide homology species in BU or VKH and other groups (including other five diseases and
healthy controls) by LDA were chosen to build a receiver-operating
To determine the sequence homology between microbial proteins characteristic (ROC) curve. Area under the curve (AUC) values were
and a uveitogenic peptide of the interphotoreceptor retinoid-binding obtained to assess the disease probability values and an AUC higher than
protein (IRBP)161-180, IRBP161-180-SGIPYIISYLHPGNTILHVD was input 0.7 was considered as a good microbial marker for the disease in
as a query sequence using the BLASTP program. Proteins encoded in question.
cultured and uncultured bacteria represented in the National Center for
Biotechnology Information (NCBI) microbial protein database were 2.10. Protein annotation and metabolic function analysis
searched for similarity to IRBP161-180 with BLAST p2.3.1 (https://blast.
ncbi.nlm.nih.gov/Blast.cgi). To investigate the functional contributions of gut microbiome
composition to the development of these diseases, the proteins of strains
2.6. Study participants associated with BU and VKH were downloaded from the NCBI database
(https://www.ncbi.nlm.nih.gov/genome/?term=) and annotated by
Eight active BU patients with uveitis were included in this study for aligning gene sequences against the proteins in the KEGG Automatic
in vitro experiments. Diagnosis of BU was based on the diagnostic Annotation Server (KAAS) [29]. Then, these proteins were mapped in
criteria of the international study group for BU [26]. Active BU was the functional pathways via KAAS to predict their potential functions.
defined according to the presence of active intraocular inflammation. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis
The study was approved by the Ethics Committee of Chongqing Medical based on these proteins were also performed via the KEGG and String
University. Signed informed consent was obtained from all participants database [30]. The shared and specific proteins or pathways were
at the beginning of the study. All procedures were performed in accor­ analyzed by an online Venn figures drawing tool (http://bioinformatics.
dance with the Declaration of Helsinki. psb.ugent.be/webtools/Venn). For metabolic function analysis, un­
mapped reads were translated against the UniRef90 protein reference
2.7. Experimental autoimmune uveitis (EAU) induction database using DIAMOND [31,32]. The generated gene lists are then
collapsed into protein families and enzyme classes/pathways using
B10.RIII mice were purchased from Jackson Laboratory (Bar Harbor, MetCyc databases [33].
ME, USA) and maintained under specific pathogen free (SPF) conditions.
For cytokine secretion assays, EAU was induced in 6–8 week old B10.RIII Availability of data and materials
mice (males and females) using 50 μg of IRBP161–180 in complete
Freund’s adjuvant (CFA) (Sigma-Aldrich, St. Louis, MO, USA) supple­ The datasets supporting the conclusions of this article are included
mented with 1.0 mg/ml Mycobacterium tuberculosis strain (MTB) as within the article and additional files. The raw sequence data (Bio­
previously described [27]. For immunization with a low dose of Project accession number PRJNA356102, PRJNA431482,
IRBP161–180, EAU was induced in B10.RIII mice using 25 μg of PRJNA400072, PRJNA375935, PRJNA356225) were downloaded from
IRBP161–180 in CFA. The animal study was approved by the Ethics SRA (https://www.ncbi.nlm.nih.gov/Traces/study/?) as mentioned in
Committee of the First Affiliated Hospital of Chongqing Medical the method section.
University.
3. Results
2.8. Real-time PCR and cytokine secretion assays
3.1. The comparison of gut microbiome composition between selected
Peripheral blood mononuclear cells (PBMCs) were purified from diseases and healthy controls
heparinized blood of BU patients with active intraocular inflammation
by human Ficoll-Hypaque density gradient centrifugation. Lymphocytes Two kinds of platforms (Illumina HiSeq 2000 and 4000) and five
from mouse spleen were filtered with cell strainers and purified by studies on three populations (China, Netherlands and America) were
mouse Ficoll-Hypaque density gradient centrifugation. The cells were included for the following analysis. To examine the influence from
cultured in culture medium consisting of RPMI medium 1640, 100 U/ml different platforms and populations on the microbiome composition, we
penicillin/streptomycin and 10% fetal bovine serum. Total RNA was first performed PCA of samples from these 5 studies in the context of
isolated by TRIzol reagent (Invitrogen, Carlsbad, CA, USA) from lym­ platforms and populations. We found that there was no significant
phoctyes. The PrimeScript RT kit (Takara Biotechnology, Dalian, China) segregation of the microbiome among different platforms or populations
was used to reverse the extracted RNA into complementary DNA. The indicating that these factors did not influence the following results and
ABI Prism 7500 system on the SYBR Premix (BIO-RAD, CA, USA) was analysis (Supplementary Figs. 1A and B). In addition, PCA of the gut
used to detect and analyze the expression. The relative expression of microbiome could not segregate BU, VKH from other immune-mediated
target genes was quantified by using the 2-△△Ct method with β-actin as diseases or healthy controls at genera level (Supplementary Fig. 2,
the internal reference. Supplementary Fig. 3 and Supplementary Fig. 4).

3
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

CPCoA based on the Bray–Curtis distance is another in depth method 3.2. Differences of the gut microbiome composition between uveitis and
to analyze differences of the gut microbiome composition between dis­ other immune-mediated diseases as well as healthy controls
eases. This method showed that CPCoA at genera level and at species
level could explain 8.82% (P = 1.0*10^-4) and 3.85% (P = 1.0*10^-4) of The aforementioned results showed a difference of gut microbiome
the total variance among these groups, respectively (Fig. 1A and B), composition between the selected diseases and healthy controls. A
showing that the difference in gut microbiome composition between further study was performed to investigate the different abundances of
these groups is statistically significant. genera and species between these diseases based on the alpha diversity.
The results of the Shannon index revealed differences concerning mi­
crobial diversity. VKH showed a much lower microbial diversity as

Fig. 1. The comparison of the gut microbiome composition between patients with various diseases and healthy controls. (A) CPCoA of the gut microbiome
composition between various diseases and healthy controls as based on the Bray–Curtis distance was performed by ANOSIM. (C and D) Circos plot depicting the
relative abundances of top 10 genera and species in six immune mediated diseases. The outer track represents the relative abundances of genera or species from six
diseases. The inner track with different colors represents different genera, species and diseases.

4
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

compared to the other five diseases and healthy controls at genera and four diseases (Fig. 2F–J), whereas Dorea was decreased in VKH. At the
species level (Supplementary Figs. 5A and B). BU also showed a lower species level, Eubacterium hallii, Dorea formicigenerans, Dorea long­
microbial diversity as compared with UC and CD but not with AS, RA icatena and Ruminococcus obeum were found to be decreased in both
and healthy controls (Supplementary Figs. 5A and B). The genera of VKH and BU (Supplementary Figs. 6A–J).
Bacteroides, Prevotella, Faecalibacterium, Alistipes, Eubacterium, Bifi­
dobacterium, Ruminococcus, Blautia, Escherichia and Roseburia were
the top 10 with the most significant differences at genera level among 3.3. Identification of a gut microbial antigen homologous with IRBP161-
the six investigated diseases. Prevotella copri, Faecalibacterium praus­ 180
nitzii, Alistipes putredinis, Bacteroides stercoris, Bacteroides plebeius,
Eubacterium rectale, Bacteroides uniformis, Bacteroides coprocola, Immune responses mediated by the retina specific protein IRBP is
Escherichia coli and Bacteroides ulgaus were the top 10 with the most thought to be involved in the pathogenesis of BU [34]. To investigate
significant differences at species level between these diseases. A Circos whether a gut microbial antigen shares homology with IRBP, we
plot depicts the relative abundance of the top 10 genera and species in analyzed the specifically enriched gut microbiome in BU patients by
these six diseases (Fig. 1C and D). using the NCBI microbial protein database for amino acid sequences
To further investigate the differences in microbial taxa between these homologous to the dominant pathogenic epitope of IRBP (residues
diseases and healthy controls, we performed a pairwise comparison 161–180). The result showed that the highest scoring sequence align­
between two uveitis entities, as well as between each disease and ment of peptide (SteTDR9-17, YVQPGNTIL) was found in the
healthy controls. In general, there was no difference in taxa between the TonB-dependent receptor (TDR) encoded by Stenotrophomonas species
two uveitis entities based on the alpha diversity. An increased abun­ and differed from IRBP169–177 by two conserved amino acids (Fig. 3A).
dance of Bilophila, Alistipes and Stenotrophomonas was found in BU as To investigate whether the Stenotrophomonas encoded SteTDR
compared with the other four selected diseases or healthy controls, could induce Th1-, and Th17-related cytokine production in BU patients,
whereas a lower abundance of Dorea, Blautia, Coprococcus, Erysipelo­ PBMCs were cultured with different concentrations (10, 50, 150 and
trichaceae and Lachnospiraceae was observed in BU (Fig. 2A–E). An 300 μg/ml) of SteTDR, 10 μg/ml IRBP161-180 as a positive control and 10
enriched Alistipes was also identified in VKH as compared with the other μg/ml human myelin oligodendrocyte glycoprotein (MOG) 35-55 as a
negative control. We found that SteTDR (300 μg/ml) and IRBP161-180

Fig. 2. Differences of the gut microbiome in uveitis and other immune-mediated diseases as well as healthy controls. (A–E) Genera showing significant differences in
the gut microbiome when comparing BU patients with other immune-mediated diseases and healthy controls. (F–J) Genera showing significant differences in the gut
microbiome when comparing VKH with other immune-mediated diseases and healthy controls. Blue, BU; Red, VKH; Green, Healthy controls; Yellow, AS; Purple, RA;
Black, CD; Brown, UC. Differentially abundant genera were identified by Wilcoxon rank sum test (FDR<0.1, corrected by the Benjamini Hochberg for multi­
ple comparison).

5
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

Fig. 3. Immune responses to IRBP161-180, MOG35-55 and SteTDR in PBMCs from BU patients and splenic lymphocytes from mice undergoing EAU. (A) Schematic
diagram of the peptide-SteTDR homology with IRBP161-180. (B) Cytokine secretion assay by ELISA, the concentrations of IFN-γ and IL-17 in the supernatants of PBMCs
and lymphocytes cultured with different peptides. n = 8 for BU patients. n = 6 for the EAU model. ***P < 0.001,**P < 0.01, *P < 0.05, ns, not significant. Wilcoxon
matched pairs test and paired samples t-test. (C) Clinical and histological uveitis in 25 μg IRBP161-180 group and two peptides mixture group. ***P < 0.001, **P <
0.01, *P < 0.05, Data was analyzed by the Mann–Whitney U test. n = 6 for each group. (D) Comparison of IFN-γ, IL-17, IL-10, MCP-1, IL-1β and TNF-α mRNA
expression between 25 μg IRBP161-180 group and two peptides mixture group, ***P < 0.001, **P < 0.01, *P < 0.05, Data was analyzed by the Mann–Whitney U test.
n = 6 for each group. (E) IRBP161-180-induced production of IFN-γ and IL-17 at protein levels in the 25 μg IRBP161-180 group and two peptides mixture group. **P <
0.01, *P < 0.05, Data was analyzed by the Mann-Whitney U test. n = 6 for each group.

were able to stimulate PBMCs from BU patients to produce more IFN-γ IFN-γ and IL-17 at the protein level was increased in the two peptides
and IL-17 as compared to the control MOG35-55 peptide group (Fig. 3B). mixture group (Fig. 3E) as compared to the 25 μg IRBP161-180 group.
B10.RIII mice are highly susceptible to the dominant pathogenic These results indicate that SteTDR derived from the gut microbiota,
murine epitope of IRBP (residues 161− 180) [35]. We stimulated lym­ shares homology and a stimulatory function with IRBP161-180 thereby
phocytes from B10.RIII mice after EAU induction using SteTDR and amplifying the antigenicity of IRBP161-180 and exacerbating EAU.
found that this peptide could induce EAU-derived lymphocytes to pro­
duce more IFN-γ and IL-17 as compared to the MOG35-55 group (Fig. 3B),
and there was no sex difference in these responses as shown by sex 3.4. Identification of gut microbial markers for BU and VKH
stratification analysis (Supplementary Fig. 7). Whether the homologous
SteTDR could also induce EAU, was tested by immunizing B10.RIII mice As mentioned above, several genera showed significant differences
with SteTDR combined with CFA. However, SteTDR could not induce concerning their abundances when comparing uveitis with the other
EAU in male or female B10.RIII mice even at a dose of 300 μg per mouse immune mediated diseases. A gut microbial marker was developed to
(data not shown). Based on the results of the immune responses test whether these genera could differentiate both uveitis entities from
mentioned above, we next questioned whether SteTDR could exacerbate the other diseases or healthy controls. After filtering the low abundant
EAU in vivo. To test this, we immunized mice with a combination of the taxa as described in the methods section, each genera including Bilo­
SteTDR peptide with a low dose of IRBP161-180. SteTDR (100 μg) was phila, Alistipes, Dorea, Blautia, Coprococcus and Lachnospiraceae was
combined with 25 μg IRBP161-180 and CFA for EAU induction. The two selected to develop microbial markers for BU to differentiate this disease
peptides mixture induced a more severe clinical and histological uveitis from the other five diseases and healthy controls (Fig. 4A). These results
in B10.RIII mice as compared to the 25ug IRBP161-180 group (Fig. 3C). showed that AUCs of the classifiers of Bilophila, Dorea and Blautia were
We then isolated splenic lymphocytes from these two groups and higher than 70%, except for VKH (Fig. 4B and Table 2). Each genera
showed that the mRNA expression of IFN-γ and IL-17 was increased in including Alistipes and Dorea was also used to develop microbial
the two peptides mixture group as compared to the 25 μg IRBP161-180 markers for VKH. The results showed that AUC of the classifier of Dorea
group (Fig. 3D). However, there was no significant difference of IL-10 were higher than 70% in discriminating VKH from the other four im­
mRNA expression between these two groups. mRNA expression of the mune mediated diseases. However, the marker could not differentiate
proinflammatory cytokines MCP-1, IL1-β and TNF-α was also increased VKH from BU. (Fig. 4C and Table 3). These results show that several gut
in the two peptides mixture group (Fig. 3D). Moreover, the result of microbial markers can be used to differentiate both uveitis entities from
ELISA measurements showed that IRBP161-180-induced production of the other four selected diseases and healthy controls, whereas other gut
microbial markers should be investigated to differentiate BU from VKH.

6
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

Fig. 4. Identification of gut microbial markers for BU and VKH. (A) The relative abundances of Bilophila, Alistipes, Dorea, Blautia, Coprococcus and Lachnospiraceae
when comparing uveitis patients with other immune-mediated diseases and healthy controls. (B) The ROC curve based on the relative abundances of Bilophila,
Alistipes, Dorea, Blautia, Coprococcus and Lachnospiraceae between BU and other groups. (C) The ROC curve based on the relative abundances of Alistipes and Dorea
between VKH and other groups.

Table 2
The AUCs of the classifiers between BU and other diseases as well as healthy controls.
AUC (Confidence Interval)

Classfiers VKH AS RA UC CD HC
g. Bilophila 69.13% (56.91%– 81.99% (71.30%– 77.52% (66.29%– 88.24% (80.36%– 91.38% (84.42%– 74.75% (63.90%–
81.34%) 92.67%) 88.75%) 96.12%) 98.33%) 85.61%)
g. Alistipes 57.35% (43.76%– 78.92% (67.75%– 71.97% (59.54%– 82.05% (72.49%– 83.37% (74.69%– 65.98% (53.67%–
70.94%) 90.09%) 84.40%) 91.61%) 93.04%) 78.29%)
g. Dorea 52.02% (38.96%– 91.50% (85.12%– 60.83% (50.41%– 79.42% (70.19%– 81.01% (73.20%– 75.70% (67.87%–
65.09%) 97.89%) 71.26%) 88.86%) 88.82%) 83.53%)
g. Blautia 57.99% (44.87%– 91.83% (85.77%– 81.49% (72.90%– 95.61% (91.38%– 97.53% (94.57%– 70.37% (74.87%–
71.10%) 97.89%) 90.08%) 99.87%) 100.5%) 84.41%)
g. Coprococcus 54.75% (41.47%– 70.59% (58.92%– 62.43% (51.51%– 68.04% (57.23%- 68.51% (58.91%– 63.22% (53.42%–
68.02%) 82.26%) 73.35%) 78.86) 78.11%) 73.01%)
g. Lachnospiraceae 55.53% (43.11%– 82.68% (72.51%– 69.25% (58.44%– 84.68% 89.92% (83.36%– 67.08% (58.71%
67.94%) 92.85%) 80.07%) (76.36–92.99%) 96.48%) 75.45%)
Gut microbial marker profiles 74.83% (63.23%– 98.37% (96.27%– 88.64% (81.09%– 99.55% (98.59%– 99.03% (97.78%– 82.82% (72.85%–
(24 species) 86.43%) 100.5%) 96.20%) 100.5%) 100.3%) 92.78%)

AUC, area under the curve; BU, Behcet’s uveitis; VKH, Vogt-Koyanagi-Harada disease; CD, Crohn’s disease, UC, ulcerative colitis; AS, Ankylosing spondylitis; RA,
rheumatoid arthritis, HC, healthy controls.

7
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

Table 3
The AUCs of the classifiers between VKH and other diseases as well as healthy controls.
AUC (Confidence Interval)

Classfiers BU AS RA UC CD HC
g. Alistipes 57.35% (43.76%– 74.70% (66.22%– 66.21% (57.72%– 77.80% (70.21%– 79.60% (72.77%– 59.51% (51.80%–
70.94%) 83.19%) 74.71%) 85.38%) 86.43%) 67.21%)
g. Dorea 52.02% (38.96%– 88.96% (82.93%– 78.34% (71.17%– 79.37% (70.35%– 81.02% (73.61%– 73.30% (64.34%–
65.09%) 94.99%) 85.52%) 88.20%) 88.43%) 79.26%)
Gut microbial marker profiles 76.42% (65.30%– 93.25% (88.67%– 96.35% (93.95%– 97.16% (93.79%– 98.53% (96.53%– 82.99% (78.29%–
(32 species) 87.53%) 97.82%) 98.74%) 100.5%) 100.5%) 87.69%)

AUC, area under the curve; BU, Behcet’s uveitis; Vogt-Koyanagi-Harada disease; CD, Crohn’s disease, UC, ulcerative colitis; AS, Ankylosing spondylitis; RA, rheu­
matoid arthritis, HC, healthy controls.

3.5. Development of gut microbial marker profiles for BU and VKH higher than 70% indicating that the usefulness of this gut microbial
marker profile in differentiating BU from the other five diseases and
To further develop microbial marker profiles for BU and VKH, we healthy controls (Fig. 5C and Table 2).
also identified the gut microbiome features of BU and VKH and For VKH, twenty-two species including Prevotella spp, Alistipes spp
compared data between these two uveitis entities and the combined and Paraprevotella spp were enriched as compared to the combined
other groups including the other immune mediated diseases and healthy other groups, whereas twenty three species including Ruminococcus spp
controls. Six species including Alitispes spp and Bilophila spp were were depleted (Fig. 5B). Thirteen species were removed due to their
enriched in BU, whereas thirty-three species including Ruminococcus extremely low abundance, and thirty-two species were used as a gut
spp, Bifidobacterium spp and Roseburia spp were depleted, respectively microbial marker profile to differentiate VKH from other diseases and
(Fig. 5A). After filtering the low abundant taxa as described in the healthy controls. The AUCs of classifiers based on the profile between
methods section, a gut microbial marker profile consisting of twenty- VKH and the other groups were also higher than 70% (Fig. 5D and
four species was used to classify BU from the other five diseases and Table 3). The result also indicated that the classifier was able to
healthy controls. The AUCs of classifiers based on the profile were all distinguish VKH from the other diseases (BU, AS, RA, CD and UC) and

Fig. 5. Development of gut microbial marker profiles for BU and VKH. (A) Histogram of LDA scores computed for species differentially abundant between BU and the
combined other groups. (B) Histogram of LDA scores computed for species differentially abundant between VKH and the other groups combined. The LDA scores
(log) > 3 are listed. (C) The ROC curve based on the profile consisting of 24 species in BU and other diseases as well as healthy controls. (D) The ROC curve based on
the profile consisted of 32 species for VKH and other diseases as well as healthy controls.

8
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

healthy controls. depleted strains were identified (Supplementary Fig. 8). After mapping
these proteins in the functional pathways, a number of pathways showed
an association with these proteins. The results showed that several
3.6. Functional analysis of the gut microbiome features in BU and VKH
proteins were mapped in a series of immune-related pathways including
patients
Th1 and Th2 cell differentiation, natural killer cell mediated cytotox­
icity, tumor necrosis factor (TNF) signaling pathway and hypoxia
To further investigate the functional roles of the gut microbiome in
inducible factor (HIF) signaling pathway (Supplementary Tables 1 and
the pathogenesis of BU and VKH, we compared the gut microbiome
2). Furthermore, two depleted proteins that mapped in the pathway of
features of BU and VKH with the combined other groups at strain level
melanogenesis were specific for VKH (Supplementary Table 2). One
and then functionally annotated the proteins using KAAS. We found that
enriched protein specific for BU was found to be mapped in the Toll and
five strains were enriched in BU, whereas thirty-three strains were
Imd signaling pathway (Supplementary Table 1).
depleted (Fig. 6A). After functional annotation, 834 and 470 proteins
It is known that the gut microbiome contributes to immunoregula­
were identified from the sequences of strains enriched and depleted in
tory function via the cumulative effects of microbial metabolites [36].
the BU patients, respectively. For VKH, 19 strains were enriched and 22
We further carried out a study to perform metabolic function analysis
strains were depleted (Fig. 6B). In these differentially abundant strains,
based on microbial features of BU and VKH patients. The result showed
790 and 536 proteins were identified from the enriched and depleted
that 108 and 178 metabolic pathways were identified associated with
strains, respectively. From the venn figure, 128 specific proteins for BU-
BU and VKH microbial features, respectively (Supplementary Tables 3
enriched strains, 20 specific proteins for BU-depleted strains, 72 specific
and 4). Among these pathways, there were 80 pathways that were
proteins for VKH-enriched strains and 47 specific proteins for VKH-

Fig. 6. Functional analysis of gut microbiome features in BU and VKH. (A) Histogram of LDA scores computed for strains differentially abundant between BU and the
other groups. The LDA scores (log) > 3 are listed. (B) Histogram of LDA scores computed for strains differentially abundant between VKH and other groups. The LDA
scores (log) > 3 are listed. (C) Diversity patterns of metabolic pathway in samples from different groups. The top of each set of stacked bars indicates the total
stratified abundance of the pathway within a single sample (log-scaled). Species and “unclassified” stratifications are linearly (proportionally) scaled within the total
bar height.

9
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

shown to be associated with both uveitis entities (Supplementary Several studies provided evidence that a gut microbiome-based
Fig. 11). Furthermore, we found that the gut microbiome of uveitis metagenomic signature may offer a universal utility as a non-invasive
patients showed a marked enrichment in the peptidoglycan biosynthesis diagnostic test for a number of diseases including schizophrenia,
I, whereas the mevalonate pathway I was depleted (Supplementary cirrhosis and nonalcoholic fatty liver disease [46–48]. Previous studies
Tables 3 and 4). We also identified 28 and 98 pathways specific for BU focused on the differences of the gut microbiome composition between a
and VKH microbial features, respectively (Supplementary Fig. 11). Of disease and a healthy control group. A number of datasets from meta­
the pathways specific for BU, the gut microbiome of BU patients was genomic studies are publicly available, and this allowed us to show that
enriched in L-methionine biosynthesis III and superpathway of L-threo­ the gut microbial signature of uveitis differs from several other immune
nine biosynthesis (Fig. 6C, Supplementary Fig. 9, Supplementary mediated diseases. However, it should be kept in mind that these results
Table 3). For VKH, we showed that the gut microbiome of VKH patients are based on a retrospective integrated-analysis and further prospective
was enriched in L-lysine biosynthesis VI and L-lysine biosynthesis III studies are needed to validate these findings.
(Fig. 6C, Supplementary Fig. 10, Supplementary Table 4). The functional annotations of proteins in the strains provides evi­
dence for a role of the gut microbiome in the pathogenesis of various
4. Discussion immune-mediated diseases. In the present study, we revealed that the
proteins in the enriched and depleted strains from BU and VKH were
This study shows that the gut microbial composition in two selected mapped in IL-17 signaling pathway and MAPK signaling pathway, which
uveitis entities is significantly different from a selection of four other have been reported earlier to be involved in the pathogenesis of BU and
immune-mediated diseases. A peptide antigen (SteTDR) encoded by BU- VKH [15,49–53]. The HIF signaling pathway, for instance, plays an
enriched Stenotrophomonas was identified as a homologous peptide important role in triggering both innate and adaptive immune response
with IRBP161-180. The specific pathways based on these microbial dif­ [54,55]. This finding combined with our results suggested that the mi­
ferences were also identified in BU and VKH via functional annotations. crobial proteins from BU and VKH patients, mapped in these pathways,
BU and VKH are the major uveitis entities in China which cause sight- might contribute to the activation of innate and adaptive immune re­
threatening intraocular inflammation [37,38]. In the present study, we sponses involved in the pathogenesis of both uveitis entities [15,56]. In
enlarged the sample size of healthy controls and further emphasized the addition, we found that melanogenesis was a specific pathway for VKH
aberrant abundances of Bilophila, Stenotrophomonas and Alistipes in after functional annotation of proteins encoded by depleted strains in
the BU and VKH patients, which are consistent with the results of our VKH. This finding is consistent with the fact that VKH is thought to be
previous studies on BU and VKH [12,13]. More importantly, these caused by an autoimmune reaction against melanocyte-associated an­
genera could differentiate BU or VKH from other diseases including AS, tigens and thus affects organs and tissues expressing these antigens,
RA, UC and CD. Bilophila is known as a sulfate-reducing bacterium which for instance explains the clinical feature of vitiligo in VKH. Thus,
(SRB) which inhibits butyrate β-oxidation whereby degraded butyrate the specific pathways identified for BU or VKH might offer clues that
may cause a Treg cell dysfunction [39–41]. In addition, both Bilophila explain their different clinical manifestations and pathogenesis.
and Stenotrophomonas are commonly considered as opportunistic Through metabolic pathway analysis, a number of metabolic path­
pathogens which produce lipopolysaccharide (LPS) that can trigger an ways including peptidoglycan biosynthesis I showed associations with
innate inflammatory response via TLRs [41]. Thus, these opportunistic BU and VKH. Previous studies have reported the associations of these
pathogens may contribute to the decrease of Treg cells, and the activa­ two uveitis entities with the gene polymorphisms of TLRs, which belong
tion of innate immune responses in BU pathogenesis. Alistipes is usually to the pattern recognition receptors recognizing peptidoglycan [57,58].
considered as a pro-inflammatory bacterium and positively correlated These findings provide important indications that infectious agents from
with increased levels of immune-related indicators (B cell and CD4+ T the gut may trigger aberrant immune responses and contribute to the
cells) [42], which might partly explain the infiltration of CD4+ T cells pathogenesis of these diseases. Furthermore, we observed a marked
and B cells reported in the different lesions of patients with VKH [43]. enrichment of L-lysine biosynthesis VI and L-lysine biosynthesis III in
However, it should be mentioned that some differences were apparent VKH as well as a marked enrichment of L-methionine biosynthesis III and
due to the fact that in our current study we used a larger group of healthy superpathway of L-threonine biosynthesis in BU. These observations are
controls and that we now used another method of taxonomical anno­ consistent with the results of previous studies showing that the levels of
tation [12,13]. these metabolites were increased in the synovial fluid and plasma from
The gut microbiome has been shown to be involved in a variety of BU and VKH patients, respectively [59,60]. Methionine, especially it’s
inflammatory diseases by molecular mimicry with self-antigens. The derivatives (such as N-formyl-methionine), is a potent neutrophil che­
group of Rachel Caspi has identified a number of sequences with partial moattractant that can trigger a variety of neutrophil-mediated responses
homology with IRBP161-180 derived from Turicibacter sp H121, which is leading to tissue damage [61], which are considered to play an impor­
a new species of Turicibacter sp found in R161H mice. None of these tant role in the pathogenesis of BD. Threonine is a target amino acid of
sequences were able to induce T cell proliferation from R161H mice protein kinase for phosphorylation that is able to activate the MAPK
expressing a TCR specific for peptide IRBP161-180 or induce disease upon pathway and subsequently enhances inflammatory responses during
immunization of WT mice [44,45]. Different from these latter studies, immune-mediated disease [62]. These findings suggest that higher
we investigated the microbial peptide homology with IRBP161-180 based levels of methionine and threonine might be involved in BD pathogen­
on the results of gut microbiome composition in human samples. esis by enhancing neutrophil activation and inflammatory responses.
Although PBMCs from BU patients and lymphocytes from EAU mice L-lysine is necessary to assist T cells in the specific recognition of anti­
responded to the IRBP cross-reactive peptide we identified in the Sten­ gens [63]. High lysine levels may activate or enhance autoimmune re­
otrophomonas species, we could not induce disease upon immunization sponses, which may contribute to VKH development. These observations
of B10.RIII mice. The in vitro response as reflected by the release of collectively suggest that the dysbiosis of the gut microbiome in BU and
IFN-γ and IL-17 was positive, following stimulation with SteTDR but was VKH patients may contribute to the development of these two diseases
significantly lower than that seen with the self-antigen IRBP161-180. This via release of their metabolites into the circulation.
may be due to the small differences in the amino acid sequence between The gut microbial signatures and their functions in BU and VKH
SteTDR and IRBP161–180. While our data could not provide direct evi­ patients showed in this study also provided a possibility of targeting the
dence that a gut microbiome mimic of a self-antigen can induce auto­ gut microbiome as a tool to prevent or treat uveitis. For example, we
immune uveitis by itself, SteTDR was able to play a synergistic effect noticed that Lactobacillus and Bifidobacterium species were depleted in
whereby its presence could enhance the antigenicity of IRBP in our EAU VKH patients, as compared to the other groups. A previous study showed
model. that a probiotic formulation consisting of Lactobacillus and

10
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

Bifidobacterium had a potential therapeutic effect on experimental References


autoimmune uveitis [64]. On the other hand, targeted antibiotic killing
of pathogenic bacteria may also be envisaged as a means to manage [1] T. Tsirouki, A. Dastiridou, C. Symeonidis, O. Tounakaki, I. Brazitikou,
C. Kalogeropoulos, et al., A focus on the epidemiology of uveitis, Ocul. Immunol.
intraocular inflammation in uveitis patients. Inflamm. 26 (2018) 2–16.
We would like to mention the limitations of our study. The biological [2] R.W. Read, Uveitis: advances in understanding of pathogenesis and treatment,
factors, including donor characteristics such as age, lifestyle, diet, sex, Curr. Rheumatol. Rep. 8 (2006) 260–266.
[3] A. Lu, Z. Liu, G. Su, P. Yang, Global research status regarding uveitis in the last
genetic background, and diet, potentially introduce considerable vari­ decade, Ocul. Immunol. Inflamm. (2023) 1–10.
ation among datasets in microbiome composition. With the future [4] M.G. Rooks, W.S. Garrett, Gut microbiota, metabolites and host immunity, Nat.
publication of additional microbiome datasets with detailed metadata of Rev. Immunol. 16 (2016) 341–352.
[5] K. Atarashi, T. Tanoue, T. Shima, A. Imaoka, T. Kuwahara, Y. Momose, et al.,
donors, stratification analysis on a specific factor may potentially Induction of colonic regulatory T cells by indigenous Clostridium species, Science
address this issue and help us to understand the effects of these factors 331 (2011) 337–341.
on the contribution of gut microbiome to uveitis pathogenesis. [6] E.A. Franzosa, A. Sirota-Madi, J. Avila-Pacheco, N. Fornelos, H.J. Haiser,
S. Reinker, et al., Gut microbiome structure and metabolic activity in inflammatory
Furthermore, there may be a correlation between gut microbiome and
bowel disease, Nat. Microbiol. 4 (2019) 293–305.
disease status. Thus, further longitudinal studies comparing early dis­ [7] C. Wen, Z. Zheng, T. Shao, L. Liu, Z. Xie, E. Le Chatelier, et al., Quantitative
ease and more advanced disease patient cohorts are needed to investi­ metagenomics reveals unique gut microbiome biomarkers in ankylosing
gate the association between disease development and gut microbiome. spondylitis, Genome Biol. 18 (2017) 142.
[8] X. Zhang, D. Zhang, H. Jia, Q. Feng, D. Wang, D. Liang, et al., The oral and gut
In conclusion, our study revealed a disease-specific gut microbiome microbiomes are perturbed in rheumatoid arthritis and partly normalized after
composition in uveitis via an integrating metagenomic sequencing data treatment, Nat. Med. 21 (2015) 895–905.
analysis. Our results also provide evidence that a uveitis-specific gut [9] A. Larabi, N. Barnich, H.T.T. Nguyen, New insights into the interplay between
autophagy, gut microbiota and inflammatory responses in IBD, Autophagy 16
microbiome may contribute to the pathogenesis of disease via a cumu­ (2020) 38–51.
lative effect of microbial antigens and their metabolites. The observa­ [10] G. Scalise, A. Ciancio, D. Mauro, F. Ciccia, Intestinal microbial metabolites in
tions shown in the present study are focused on the microbial signatures ankylosing spondylitis, J. Clin. Med. (2021) 10.
[11] L. Lin, J. Zhang, Role of intestinal microbiota and metabolites on gut homeostasis
in uveitis but a similar approach may also be used for other immune- and human diseases, BMC Immunol. 18 (2017) 2.
mediated diseases. [12] Z. Ye, N. Zhang, C. Wu, X. Zhang, Q. Wang, X. Huang, et al., A metagenomic study
of the gut microbiome in Behcet’s disease, Microbiome 6 (2018) 135.
[13] Z. Ye, C. Wu, N. Zhang, L. Du, Q. Cao, X. Huang, et al., Altered gut microbiome
Author contributions composition in patients with Vogt-Koyanagi-Harada disease, Gut Microb. 11
(2020) 539–555.
Q.W. and P.Y. conceived and directed the study. Q.W., S.W., X.Y., S. [14] M. Pineton de Chambrun, B. Wechsler, G. Geri, P. Cacoub, D. Saadoun, New
insights into the pathogenesis of Behcet’s disease, Autoimmun. Rev. 11 (2012)
T. and F.H analyzed the data. P.Y., G.S. and A.K. reviewed the data 687–698.
interpretation and helped revise the final versions of the manuscript. All [15] L. Du, A. Kijlstra, P. Yang Vogt-Koyanagi-Harada disease, Novel insights into
authors read and approved the final manuscript. pathophysiology, diagnosis and treatment, Prog. Retin. Eye Res. 52 (2016) 84–111.
[16] J.M. Moll, I. Haslock, I.F. Macrae, V. Wright, Associations between ankylosing
spondylitis, psoriatic arthritis, Reiter’s disease, the intestinal arthropathies, and
Funding Behcet’s syndrome, Medicine 53 (1974) 343–364.
[17] K. Aksu, A. Donmez, G. Keser, Inflammation-induced thrombosis: mechanisms,
disease associations and management, Curr. Pharmaceut. Des. 18 (2012)
This study was supported by the National Natural Science Founda­ 1478–1493.
tion Key Program (81930023), National Natural Science Foundation Key [18] M. al-Janadi, S. al-Balla, A. al-Dalaan, S. Raziuddin, Cytokine profile in systemic
Program (82230032), National Natural Science Foundation lupus erythematosus, rheumatoid arthritis, and other rheumatic diseases, J. Clin.
Immunol. 13 (1993) 58–67.
(82101106), Natural Science Key Project of Chongqing Science and [19] Q. Wang, G. Su, X. Tan, J. Deng, L. Du, X. Huang, et al., UVEOGENE: an SNP
Technology Bureau (CSTC2021jscx-gksb-N0010), Foundation of database for investigations on genetic factors associated with uveitis and their
Chongqing (cstc2021jcyj-bsh0055), Chongqing Outstanding Scientists relationship with other systemic autoimmune diseases, Hum. Mutat. 40 (2019)
258–266.
Project (2019), Chongqing Chief Medical Scientist Project (2018), [20] R. Scotti, S. Southern, C. Boinett, T.P. Jenkins, A. Cortes, C. Cantacessi,
Chongqing Key Laboratory of Ophthalmology (CSTC, 2008CA5003) and MICHELINdb: a web-based tool for mining of helminth-microbiota interaction
Chongqing Science & Technology Platform and Base Construction Pro­ datasets, and a meta-analysis of current research, Microbiome 8 (2020) 10.
[21] S. Nayfach, K.S. Pollard, Toward accurate and quantitative comparative
gram (cstc2014pt-sy10002).
metagenomics, Cell 166 (2016) 1103–1116.
[22] A.M. Bolger, M. Lohse, B. Usadel, Trimmomatic: a flexible trimmer for Illumina
Data sharing statement sequence data, Bioinformatics 30 (2014) 2114–2120.
[23] B. Langmead, S.L. Salzberg, Fast gapped-read alignment with Bowtie 2, Nat.
Methods 9 (2012) 357–359.
Raw sequence data (BioProject accession number PRJNA356102, [24] D.T. Truong, E.A. Franzosa, T.L. Tickle, M. Scholz, G. Weingart, E. Pasolli, et al.,
PRJNA431482, PRJNA400072, PRJNA375935, PRJNA356225) were MetaPhlAn2 for enhanced metagenomic taxonomic profiling, Nat. Methods 12
(2015) 902–903.
downloaded from SRA (https://www.ncbi.nlm.nih.gov/Traces/st [25] J. Goecks, A. Nekrutenko, J. Taylor Galaxy, A comprehensive approach for
udy/?). The datasets used and/or analyzed during the current study supporting accessible, reproducible, and transparent computational research in the
are available from the corresponding author on reasonable request. life sciences, Genome Biol. 11 (2010) R86.
[26] Criteria for diagnosis of behcet’s disease. International study group for behcet’s
disease, Lancet 335 (1990) 1078–1080.
[27] G. Jiang, Y. Ke, D. Sun, G. Han, H.J. Kaplan, H. Shao, Reactivation of uveitogenic T
Declaration of competing interest
cells by retinal astrocytes derived from experimental autoimmune uveitis-prone
B10RIII mice, Invest. Ophthalmol. Vis. Sci. 49 (2008) 282–289.
The authors declare no competing interests. [28] W. de Jager, B.J. Prakken, J.W. Bijlsma, W. Kuis, G.T. Rijkers, Improved multiplex
immunoassay performance in human plasma and synovial fluid following removal
of interfering heterophilic antibodies, J. Immunol. Methods 300 (2005) 124–135.
Data availability [29] Y. Moriya, M. Itoh, S. Okuda, A.C. Yoshizawa, M. Kanehisa, KAAS: an automatic
genome annotation and pathway reconstruction server, Nucleic Acids Res. 35
Data will be made available on request. (2007) W182–W185.
[30] D. Szklarczyk, A. Franceschini, M. Kuhn, M. Simonovic, A. Roth, P. Minguez, et al.,
The STRING database in 2011: functional interaction networks of proteins, globally
Appendix A. Supplementary data integrated and scored, Nucleic Acids Res. 39 (2011) D561–D568.
[31] B.E. Suzek, Y. Wang, H. Huang, P.B. McGarvey, C.H. Wu, UniRef clusters: a
comprehensive and scalable alternative for improving sequence similarity
Supplementary data to this article can be found online at https://doi. searches, Bioinformatics 31 (2015) 926–932.
org/10.1016/j.jaut.2023.103055.

11
Q. Wang et al. Journal of Autoimmunity 137 (2023) 103055

[32] B. Buchfink, C. Xie, D.H. Huson, Fast and sensitive protein alignment using [49] K. Hamzaoui, Th17 cells in Behcet’s disease: a new immunoregulatory axis, Clin.
DIAMOND, Nat. Methods 12 (2015) 59–60. Exp. Rheumatol. 29 (2011) S71–S76.
[33] R. Caspi, T. Altman, R. Billington, K. Dreher, H. Foerster, C.A. Fulcher, et al., The [50] Y. He, C. Wang, G. Su, B. Deng, Z. Ye, Y. Huang, et al., Decreased expression of A20
MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of is associated with ocular Behcet’s disease (BD) but not with Vogt-Koyanagi-Harada
Pathway/Genome Databases, Nucleic Acids Res. 42 (2014) D459–D471. (VKH) disease, Br. J. Ophthalmol. 102 (2018) 1167–1172.
[34] M. Takeuchi, Y. Usui, Y. Okunuki, L. Zhang, J. Ma, N. Yamakawa, et al., Immune [51] B. Deng, Z. Ye, L. Li, D. Zhang, Y. Zhu, Y. He, et al., Higher expression of NOD1 and
responses to interphotoreceptor retinoid-binding protein and S-antigen in Behcet’s NOD2 is associated with vogt-koyanagi-harada (VKH) syndrome but not behcet’s
patients with uveitis, Invest. Ophthalmol. Vis. Sci. 51 (2010) 3067–3075. disease (BD), Curr. Mol. Med. 16 (2016) 424–435.
[35] L. Song, J. Le, F. Ye, H. Shao, H.J. Kaplan, D. Sun, Sequence 168 to 177 of [52] A.G. Commodaro, C.R. Bombardieri, J.P. Peron, K.C. Saito, P.M. Guedes, D.
interphotoreceptor retinoid-binding protein (IRBP) is an antigenic epitope for E. Hamassaki, et al., p38{alpha} MAP kinase controls IL-17 synthesis in vogt-
autoreactive CD8 T cells in the B10RIII mouse, J. Neuroimmunol. 193 (2008) koyanagi-harada syndrome and experimental autoimmune uveitis, Invest.
68–76. Ophthalmol. Vis. Sci. 51 (2010) 3567–3574.
[36] Y. Moon, Microbiome-linked crosstalk in the gastrointestinal exposome towards [53] F. Ture-Ozdemir, A. Tulunay, M.O. Elbasi, I. Tatli, A.M. Maurer, G. Mumcu, et al.,
host health and disease, Pediatric Gastroenterol. Hepatol. Nutrition 19 (2016) Pro-inflammatory cytokine and caspase-1 responses to pattern recognition receptor
221–228. activation of neutrophils and dendritic cells in Behcet’s disease, Rheumatology 52
[37] P. Yang, Z. Zhang, H. Zhou, B. Li, X. Huang, Y. Gao, et al., Clinical patterns and (2013) 800–805.
characteristics of uveitis in a tertiary center for uveitis in China, Curr. Eye Res. 30 [54] P. Sadiku, S.R. Walmsley, Hypoxia and the Regulation of Myeloid Cell Metabolic
(2005) 943–948. Imprinting: Consequences for the Inflammatory Response, EMBO reports, 2019,
[38] P. Yang, L. Du, Z. Ye, How to deal with uveitis patients? Curr. Mol. Med. 17 (2018) p. 20.
468–470. [55] T. Takahashi, Roles of nAChR and Wnt signaling in intestinal stem cell function and
[39] G.M. Nava, F. Carbonero, J.A. Croix, E. Greenberg, H.R. Gaskins, Abundance and inflammation, Int. Immunopharm. 81 (2020), 106260.
diversity of mucosa-associated hydrogenotrophic microbes in the healthy human [56] E. Alpsoy, B.C. Bozca, A. Bilgic Behcet Disease, An update for dermatologists, Am.
colon, ISME J. 6 (2012) 57–70. J. Clin. Dermatol. 22 (2021) 477–502.
[40] Y. Furusawa, Y. Obata, S. Fukuda, T.A. Endo, G. Nakato, D. Takahashi, et al., [57] G.G. Song, S.J. Choi, J.D. Ji, Y.H. Lee, Toll-like receptor polymorphisms and
Commensal microbe-derived butyrate induces the differentiation of colonic vasculitis susceptibility: meta-analysis and systematic review, Mol. Biol. Rep. 40
regulatory T cells, Nature 504 (2013) 446–450. (2013) 1315–1323.
[41] G. Muyzer, A.J. Stams, The ecology and biotechnology of sulphate-reducing [58] Y. Jiang, H. Wang, H. Yu, L. Li, D. Xu, S. Hou, et al., Two Genetic Variations in the
bacteria, Nat. Rev. Microbiol. 6 (2008) 441–454. IRF8 region are associated with Behcet’s disease in Han Chinese, Sci. Rep. 6
[42] B.J. Parker, P.A. Wearsch, A.C.M. Veloo, A. Rodriguez-Palacios, The genus (2016), 19651.
Alistipes: gut bacteria with emerging implications to inflammation, cancer, and [59] L. Chen, R. Chang, S. Pan, J. Xu, Q. Cao, G. Su, et al., Plasma metabolomics study of
mental health, Front. Immunol. 11 (2020) 906. Vogt-Koyanagi-Harada disease identifies potential diagnostic biomarkers, Exp. Eye
[43] A.M. Abu El-Asrar, J. Van Damme, S. Struyf, G. Opdenakker, New perspectives on Res. 196 (2020), 108070.
the immunopathogenesis and treatment of uveitis associated with vogt-koyanagi- [60] J.K. Ahn, S. Kim, J. Kim, J. Hwang, K.H. Kim, H.S. Cha, A comparative
harada disease, Front. Med. 8 (2021), 705796. metabolomic evaluation of behcet’s disease with arthritis and seronegative
[44] R. Horai, P.B. Silver, J. Chen, R.K. Agarwal, W.P. Chong, Y. Jittayasothorn, et al., arthritis using synovial fluid, PLoS One 10 (2015), e0135856.
Breakdown of immune privilege and spontaneous autoimmunity in mice [61] B. Duvvuri, A.A. Baddour, K.D. Deane, M.L. Feser, J.L. Nelson, M.K. Demoruelle, et
expressing a transgenic T cell receptor specific for a retinal autoantigen, al., Mitochondrial N-formyl methionine peptides associate with disease activity as
J. Autoimmun. 44 (2013) 21–33. well as contribute to neutrophil activation in patients with rheumatoid arthritis,
[45] C.R. Zarate-Blades, R. Horai, M.J. Mattapallil, N.J. Ajami, M. Wong, J.F. Petrosino, J. Autoimmun. 119 (2021), 102630.
et al., Gut microbiota as a source of a surrogate antigen that triggers autoimmunity [62] L. Yuan, F. Zhang, M. Shen, S. Jia, J. Xie, Phytosterols suppress phagocytosis and
in an immune privileged site, Gut Microb. 8 (2017) 59–66. inhibit inflammatory mediators via ERK pathway on LPS-triggered inflammatory
[46] F. Zhu, Y. Ju, W. Wang, Q. Wang, R. Guo, Q. Ma, et al., Metagenome-wide responses in RAW264.7 macrophages and the correlation with their structure,
association of gut microbiome features for schizophrenia, Nat. Commun. 11 (2020) Foods 8 (2019).
1612. [63] E. Suleiman, D. Damm, M. Batzoni, V. Temchura, A. Wagner, K. Uberla, et al.,
[47] T.G. Oh, S.M. Kim, C. Caussy, T. Fu, J. Guo, S. Bassirian, et al., A Universal Gut- Electrostatically driven encapsulation of hydrophilic, non-conformational peptide
Microbiome-Derived Signature Predicts Cirrhosis, Cell metabolism, 2020. epitopes into liposomes, Pharmaceutics 11 (2019).
[48] R. Loomba, V. Seguritan, W. Li, T. Long, N. Klitgord, A. Bhatt, et al., Gut [64] J. Kim, S.H. Choi, Y.J. Kim, H.J. Jeong, J.S. Ryu, H.J. Lee, et al., Clinical effect of
microbiome-based metagenomic signature for non-invasive detection of advanced IRT-5 probiotics on immune modulation of autoimmunity or alloimmunity in the
fibrosis in human nonalcoholic fatty liver disease, Cell Metabol. 25 (2017) eye, Nutrients 9 (2017).
1054–10562 e5.

12

You might also like