You are on page 1of 9

Journal of Autoimmunity 133 (2022) 102920

Contents lists available at ScienceDirect

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

Transcriptomic profiling of iris tissue highlights LCK signaling and T


cell-mediated immunity in Behcet’s uveitis
Yang Deng a, Yinan Zhang b, c, Tao Cai a, Qingfeng Wang a, Wanyun Zhang a, Zhijun Chen a,
Xiang Luo a, Guannan Su a, Peizeng Yang a, *
a
The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing Branch of National
Clinical Research Center for Ocular Diseases, Chongqing, PR China
b
Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for
Ocular Immunology and Retinal Injury Repair, Zhengzhou, PR China
c
The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China

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

Keywords: Uveitis is the most common form of ocular lesions in Behcet’s disease, severely affecting visual function. Mo­
RNA sequencing lecular pathological changes of ocular lesions in patients with Behcet’s uveitis (BU) are largely unknown. In this
Behcet disease study, we performed the first comprehensive transcriptomic profiling of iris specimens from BU patients and
Uveitis
healthy donors to provide an insight into intraocular immunopathogenesis. The mRNA sequencing identified
T cells
LCK
1633 differentially expressed genes (DEGs) between the BU group and healthy controls. GO functional enrich­
Iris ment analysis on DEGs showed that T cell activation was the most significantly enriched biological process.
KEGG analysis of DEGs also revealed several prominently enriched T cell-related pathways, including the T cell
receptor signaling pathway, Th17 cell differentiation, and Th1 and Th2 cell differentiation. The lymphocyte-
specific protein tyrosine kinase (LCK) was identified as the key hub gene in the protein interaction network of
DEGs. Western blot analysis further showed increased expression of active LCK in the BU group, suggesting
activation of LCK signaling. Using publicly accessible single-cell RNA-sequencing data of the healthy iris, LCK
was found to be expressed in clusters of activated T cells but not in other iris cell clusters, suggesting an overt
association between LCK upregulation and T cell-mediated immune dysregulation. Additionally, 16 drugs were
predicted to be potential inhibitors of LCK. Overall, these findings not only highlighted the central role of T cell-
mediated immunity and previously unreported LCK signaling in intraocular immunopathogenesis but also
revealed the potential value of LCK as a new therapeutic target for BU patients.

1. Introduction from vision-threatening intraocular inflammation, greatly reducing


their quality of life [4]. Behcet’s uveitis (BU), characterized by recurrent
Behcet’s disease (BD) is a complex multi-system disorder with bilateral non-granulomatous panuveitis, is the most common form of BD
autoimmune and autoinflammatory features [1]. It is more prevalent in ocular lesions [5]. Although genetic factors and peripheral immuno­
the countries along the old Silk Road, such as Turkey, Iran, Israel, Japan, logical mechanisms have been extensively explored [6–9], intraocular
and China [2]. Recurrent oral aphthosis, genital aphthosis, ocular le­ immunopathogenesis in BU patients remains poorly understood. Its
sions, skin lesions, and other systemic lesions are the characteristic treatment also remains challenging, with approximately 25% of patients
clinical manifestations of BD [3]. Over two-thirds of BD patients suffer reported to have blindness despite treatment [2]. Therefore, it is

Abbreviations: BD, Behcet’s disease; BU, Behcet’s uveitis; scRNA-seq, single-cell RNA sequencing; DEGs, differentially expressed genes; AAU, acute anterior
uveitis; VKH, Vogt-Koyanagi-Harada; KEGG, Kyoto Encyclopedia of Genes and Genomes; GO, Gene Ontology; GSEA, Gene Set Enrichment Analysis; NES, normalized
enrichment scores; FDR, false discovery rate; PPI, protein-protein interaction; MCODE, Molecular Complex Detection; DGIdb, Drug–Gene Interaction Database; PCA,
principal component analysis; t-SNE, T-Distributed Stochastic Neighbor Embedding; RT-qPCR, Real-time quantitative polymerase chain reaction; PMSF, phenyl­
methanesulfonyl fluoride; LCK, lymphocyte-specific protein tyrosine kinase.
* Corresponding author. The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute,
Chongqing Branch of National Clinical Research Center for Ocular Diseases, 1 You Yi Road, Yu Zhong District, Chongqing, 400016, China.
E-mail address: peizengycmu@126.com (P. Yang).

https://doi.org/10.1016/j.jaut.2022.102920
Received 22 July 2022; Received in revised form 19 September 2022; Accepted 19 September 2022
Available online 30 September 2022
0896-8411/© 2022 Elsevier Ltd. All rights reserved.
Y. Deng et al. Journal of Autoimmunity 133 (2022) 102920

clinically necessary and urgent to comprehensively clarify the BU DESeq2 R package (version 1.20.0) was used to identify DEGs with
pathogenesis and develop more effective and targeted therapies. the criteria of absolute log2 (fold change) ≥1 and adjusted P-value
Due to the limited availability of samples from intraocular inflam­ (padj) ≤0.05. ClusterProfiler R package (version 3.8.1) was applied to
matory sites and the lack of ideal animal models, most previous studies examine DEGs enrichment in the Kyoto Encyclopedia of Genes and
relied on peripheral blood samples from BU patients to explore the Genomes (KEGG) pathways and Gene Ontology (GO) terms. A padj
pathogenic mechanism. However, analysis of samples obtained from ≤0.05 indicated significant enrichment. Moreover, Gene Set Enrichment
peripheral sites alone cannot fully disclose intraocular pathophysiolog­ Analysis (GSEA) was conducted with the KEGG and GO data sets as
ical changes. In recent years, increasing studies have focused on the previously described [20]. The threshold of GSEA on significance was
analysis of intraocular aqueous humor samples, revealing the charac­ normalized enrichment scores (NES) > 1, nominal P < 0.05, or false
teristic intraocular immune microenvironment [10–12]. Differences in discovery rate (FDR) q-value<0.25 [21].
cytokine profiles between intraocular aqueous humor and peripheral
serum or plasma have also been reported [13,14]. However, research on 2.3. Analysis of protein-protein interaction (PPI) network and
the iris, one of the key intraocular inflammatory sites, remains unex­ identification of hub genes
plored, and its investigation may improve our understanding of BU
intraocular immunopathogenesis. PPI network of DEGs was obtained based on the STRING database,
The recently booming single-cell RNA sequencing (scRNA-seq) and its visualization was constructed by Cytoscape software (version
technologies can decipher pathophysiological changes at single-cell 3.9.1). Molecular Complex Detection (MCODE), a Cytoscape plugin
resolution among heterogeneous cells [15]. Additionally, growing (version 2.0.0), was also used to find clusters of highly interconnected in
research has shown the potential of combined analysis of scRNA-seq and the network with the default settings. KEGG pathways enriched by DEGs
bulk RNA-seq data to elucidate the exact cellular and molecular mech­ in clusters of interest were identified and visualized using the Cytoscape
anism in various diseases, such as osteoarthritis, BD, and cancers plugin ClueGO (version 2.5.8).
[16–18]. In this study, we performed comprehensive transcriptomic Hub genes were the critical nodes in the network, identified by the
profiling of iris specimens obtained during peripheral iridectomy in BU Cytoscape plugin cytoHubba (version 0.1). To optimize sensitivity and
patients and compared them with healthy iris specimens. Also, we in­ specificity, five topological algorithms (Degree, MNC, EPC, Closeness,
tegrated publicly available scRNA-seq data of the healthy iris with our and Radiality) were applied to identify their top 20 hub genes respec­
bulk RNA-seq data to reveal cellular expression patterns of differentially tively, and the genes located at their intersections were finally chosen as
expressed genes (DEGs) and provide an insight into DEGs. the hub genes in this study [22].

2. Materials and methods 2.4. Drug-gene interactions

2.1. Iris sample collection Drug–Gene Interaction Database (DGIdb 4.0) was used to annotate
whether the gene of interest was targeted by any known drugs or
All samples were collected at the First Affiliated Hospital of belonged to any putatively druggable gene categories [23]. The infor­
Chongqing Medical University between December 2019 and August mation provided by DGIdb is based mainly on publications and potential
2022, with written informed consent obtained from each participant. All data sources such as TTD, Chembl, DrugBank, Drug Target Commons,
study procedures were performed according to the Declaration of Hel­ and PharmGKB.
sinki principles and were authorized by the institutional review board at
the First Affiliated Hospital of Chongqing Medical University. All of the 2.5. scRNA-seq
included BU patients met the two diagnostic criteria proposed by the
International Study Group [19] and the International Revision Team The scRNA-seq data of healthy iris from a male donor aged 39 was
[3]. These patients developed nearly complete posterior synechiae and downloaded from the GEO database (GSE147979) and was analyzed by
underwent a preventive peripheral iridectomy to avoid secondary the R package Seurat (version 4.0.4). Before analysis, we filtered out
glaucoma or increased intraocular pressure. Iris samples from the pa­ cells with less than 200 genes and only considered genes detected in at
tients with other two most common uveitis entities, acute anterior least three cells. Outliers in data filtering indicators (features, counts,
uveitis (AAU) and Vogt-Koyanagi-Harada (VKH) disease, were also and mitochondrial genes) were removed. After data quality control, the
collected as disease controls. The eye bank provided the age- and data was normalized by the function NormalizeData. The function
sex-matched healthy iris without any history of eye disease or eye sur­ FindVariableFeatures was used to find the top 2000 variable genes, and
gery. These specimens as healthy controls were obtained using a similar the function ScaleData was simultaneously used to scale data. Iris data
surgical approach. The donors’ causes of death were car accidents, ce­ was then investigated by principal component analysis (PCA) and
rebral hemorrhage, and suicide by dichlorvos overdose. The time from clustering analysis. We selected the top 20 principal components and set
death to enucleation was 2–6 h. This study involved a total of 14 BU the parameter resolution at 0.2 for clustering analysis. T-Distributed
patients, 10 VKH patients, 10 AAU patients and 10 healthy donors, and Stochastic Neighbor Embedding (t-SNE) was performed after PCA and
collected one iris specimen from each patient and healthy individual for clustering. For the annotation of cell types, we used the same gene
analysis. The basic features of all enrolled patients are detailed in Sup­ markers as reported in this scRNA-seq data [24].
plemental Table 1.
2.6. Real-time quantitative polymerase chain reaction (RT-qPCR)
2.2. Bulk RNA-seq, identification of DEGs and enrichment analysis
According to the manufacturer’s protocols, total RNA of the iris was
The iris from four BU patients and four healthy controls were chosen extracted by SteadyPure Universal RNA Extraction Kit (Accurate
for the bulk RNA-seq. Bulk RNA-seq was performed by Novogene (Bei­ Biology, China) and reverse transcribed into cDNA by RT Master Mix for
jing, China) under the condition that total RNA quality met library qPCR II (MCE, USA). RT-qPCR was conducted using SYBR Green qPCR
construction and mRNA sequencing requirements. Libraries were con­ Master Mix (MCE, USA) on an ABI 7500 Real-Time PCR System (Applied
structed using the NEBNext® Ultra™ RNA Library Prep Kit for Illu­ Biosystems, USA). The primers synthesized by Sangon (Shanghai, China)
mina®. After the Libraries were qualified, sequencing was carried out by are as follows: β-actin (forward primer-
the Illumina NovaSeq 6000. High-quality clean data were generated by ACTGGAACGGTGAAGGTGACAG and reverse primer-
removing unqualified reads from raw data before further analysis. GGTGGCTTTTAGGATGGCAA G), and LCK (forward primer-

2
Y. Deng et al. Journal of Autoimmunity 133 (2022) 102920

TGGACAACGGTGGCTTCTAC and reverse primer-GCCCATCTGAA clusters, and samples from the same group exhibited a similar expression
GCATTGGTG). Relative expression was calculated using the 2− ΔΔCt pattern (Fig. 1C). Supplemental Table 2 provides information on all
method. DEGs in detail.

2.7. Western blot 3.2. Prominence of T cell-mediated biological processes in the


pathogenesis of BU
Total protein was extracted using RIPA Lysis Buffer (Beyotime,
China) supplemented with phenylmethanesulfonyl fluoride (PMSF) and We conducted GO and KEGG enrichment analyses to determine the
phosphatase inhibitor cocktail A. Proteins were separated on an 8% functions and potential biological roles of these DEGs. GO is a
precast gel (ACE Biotechnology, China) and transferred onto a PVDF comprehensive database documenting gene functions and comprises
membrane. Membranes were blocked with NcmBlot blocking buffer three categories: biological process, cellular component, and molecular
(NCM Biotech, China). The membranes were then incubated overnight function. The bar graph represented the ten most significantly enriched
at 4 ◦ C with the following primary antibodies: rabbit polyclonal anti- GO items in each category (Fig. 2A). T cell activation was found to be the
LCK (1:1000, ABclonal, A2177); rabbit polyclonal anti-Phospho-LCK- most significantly enriched biological process, followed by leukocyte
Y394 (1:500, ABclonal, AP0182); and rabbit polyclonal anti-GAPDH migration, adaptive immune response, and lymphocyte differentiation.
(1:20000, Affinity, AF7021). After incubation with corresponding sec­ T cell activation involved 111 upregulated DEGs and 7 downregulated
ondary antibodies for 1 h at room temperature, the proteins were DEGs. The GSEA using the GO dataset further demonstrated a prominent
detected on an Image Lab system (Bio-Rad, USA) using a BeyoECL Star association of T cell activation with BU phenotype (Fig. 2B). In addition,
kit (Beyotime, China). All procedures followed the manufacturers’ in­ several T cell-related KEGG pathways, including the T cell receptor
structions, and protein bands were quantized by ImageJ software. signaling pathway, Th17 cell differentiation, and Th1 and Th2 cell dif­
ferentiation, were also markedly enriched by DEGs (Fig. 2C). Similarly,
2.8. Immunohistochemistry the GSEA using the KEGG dataset confirmed that these T cell-related
KEGG pathways were largely responsible for the BU phenotype
The paraffin-embedded skin biopsy specimens were from three pa­ (Fig. 2D–F). These results suggest that T cell-mediated immune re­
tients with BD-related skin lesions (erythema nodosum) and from three sponses feature prominently in the pathogenesis of BU.
age-and sex-matched healthy donors who underwent plastic surgery (C.
T.). Immunohistochemical staining of LCK (1:200) was performed 3.3. Identification of important modules and hub genes in PPI network
following the previously described protocol [25]. Images were acquired
by a 3DHISTECH slide scanner and its SlideViewer software (version We next explored the protein-protein interactions of these dysregu­
2.5). lated genes using the STRING database. Overall, 1014 nodes (protein-
coding genes) and 6003 edges (protein-protein interactions) were found
3. Results to constitute a vast and intricate PPI network (Fig. 3A). Two important
modules were recognized in the PPI network using the MCODE plugin in
3.1. Identification of dysregulated genes in the iris of BU patients Cytoscape software (Fig. 3B). The module 1 (score = 37.68) consisted of
38 nodes and 697 edges, and its relevant DEGs were significantly
To reveal the pathogenic molecular features in the iris of BU patients, enriched in the cytokine-related pathways such as cytokine-cytokine
we performed mRNA-seq on this tissue. Principal component analysis receptor interaction, chemokine signaling pathway, IL-17 signaling
(PCA) showed a clear separation between the BU group and healthy pathway, and TNF signaling pathway (Fig. 3C). The T cell-related
controls (Fig. 1A), indicating the presence of disease-specific gene pathways, including T cell receptor signaling pathway, Th17 cell dif­
expression patterns. A total of 1633 dysregulated DEGs were identified, ferentiation, and Th1 and Th2 cell differentiation, were highly enriched
with 1142 being upregulated (Fig. 1B). Cluster analysis on DEGs showed by DEGs in the module 2 (score = 13.13) containing 40 nodes and 256
that samples from BU patients and controls were grouped into distinct edges (Fig. 3D). We further determined the hub genes in the PPI network

Fig. 1. Comparison of gene expression profiles of the iris between BU patients and healthy controls. (A) Principal component analysis results of all mRNA-sequencing
samples (n = 8).(B) The volcano plot of the DEGs in the BU group compared with healthy controls. (C) The heat map of the DEGs between these two groups. DEGs,
differentially expressed genes; BU, Behcet’s uveitis; B, the iris sample from the Behcet’s uveitis group; C, the iris sample from healthy controls.

3
Y. Deng et al. Journal of Autoimmunity 133 (2022) 102920

Fig. 2. Functional enrichment analyses. (A) The GO enrichment analysis of all DEGs. The bar graph shows the ten most significantly enriched GO items in each
category, including biological process (BP), cellular component (CC), and molecular function (MF). (B) The GSEA enrichment plot for the most significantly enriched
GO item marked by red rectangle in (A). The NES = 2.18, nominal P < 0.001, and FDR q-value<0.001. (C) The KEGG enrichment analysis of all DEGs. The bubble
plot shows all significantly enriched KEGG pathways. (D–F) The GSEA enrichment plots for the significantly enriched T cell-related KEGG pathways marked by red
rectangle in (C). The NES = 1.66, nominal P < 0.001, and FDR q-value = 0.009 in (D); The NES = 1.90, nominal P < 0.001, and FDR q-value<0.001 in (E); The NES
= 1.80, nominal P < 0.001, and FDR q-value = 0.002 in (F). DEGs, differentially expressed genes; GSEA, Gene Set Enrichment Analysis; NES, normalized enrichment
scores; FDR, false discovery rate. GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes.

by choosing overlapping genes obtained by 5 algorithms in the cyto­ phosphorylation level of LCK at Y394, an indicator of activation.
Hubba (Fig. 3E). A total of 7 hub genes, with 6 being upregulated, were Western blot results showed an increased phosphorylation level in the
identified. The Cytoscape was used to visualize these hub genes ac­ BU group (Fig. 4C–F), indicating the presence of abundantly activated
cording to their significance of differential expression between the BU LCK in the iris of BU patients. The LCK upregulation, including its
patients and healthy controls (Fig. 3F). Table 1 provides details on these mRNA, total protein, and active protein levels, was also observed in the
7 hub genes. We found that the top two hub genes, CD8A and iris from AAU and VKH patients (Supplemental Fig. 1). Additionally,
lymphocyte-specific protein tyrosine kinase (LCK), were closely related immunohistochemical staining results also showed a distinctly increased
to T cell functions [26], further suggesting an important role of T cells in LCK expression in the BD-related skin lesions (Supplemental Fig. 2).
BU pathogenesis. Finally, we chose LCK as the key hub gene for the
following studies because difference of LCK expression between these
two groups was the most significant among all identified hub genes. 3.5. Analysis on LCK combined with the scRNA-seq data

We analyzed the cellular expression patterns of DEGs in the healthy


3.4. Validation of the key hub gene LCK iris using the scRNA-seq data to determine which cell types constitu­
tively express LCK. The tSNE plots of the scRNA-seq data showed eight
To verify the reliability of the RNA-seq results, we performed RT- distinct iris cell clusters, including fibroblasts, melanocytes, smooth
qPCR and Western blot to detect LCK expression in the iris of another muscle cells, activated T cells, monocytes, Schwann cells, putative stem
cohort containing 10 BU patients and 5 healthy controls. Consistent with cells, and cytotoxic T cells (Fig. 5A). In these iris cell clusters, we found
the RNA-seq data, RT-qPCR results showed that LCK mRNA level in the that the DEGs mainly exhibited five types of distribution features. Type 1
BU group was substantially higher than that in the controls (Fig. 4A and referred to those genes that were not constitutively expressed in almost
B). Western blot analysis also confirmed an obviously increased LCK all clusters (Fig. 5B). Type 2 exhibited sporadic distribution of the DEGs
protein level in the BU group (Fig. 4C–F). Furthermore, we examined the (Fig. 5C). Type 3 showed a preferred distribution in one or more clusters

4
Y. Deng et al. Journal of Autoimmunity 133 (2022) 102920

Fig. 3. Analysis of PPI network of the DEGs. (A) The overview of the PPI network. Pink nodes represent up-regulated DEGs in the BU group, dark blue nodes
represent down-regulated DEGs in the BU group, and light blue nodes denote non-DEGs predicted to interact with the DEGs. (B) The overview of two modules in the
PPI network. All genes in the module 1 (left side) and module 2 (right side) are shown. (C–D) KEGG pathways enriched by genes in the module 1 (C) and module 2
(D), respectively. The size of the pathway node indicates the number of mapped genes, and its color denotes the significance of the pathway enrichment, with dark
brown representing a P value less than 0.0005 and red representing a P value less than 0.05 but greater than 0.005. (E) Venn diagram showing overlapping hub genes
identified by 5 algorithms in the cytoHubba. (F) PPI network of the 7 overlapping hub genes. The node size represents the significance level of the differential
expression of hub genes between the BU patients and healthy controls; the larger the node, the more significant it is. PPI, protein-protein interaction; DEGs,
differentially expressed genes; BU, Behcet’s uveitis.

(Fig. 5D). Type 4 referred to those genes expressed almost exclusively in 3.6. Screening of potential drugs that target LCK
one cluster (Fig. 5E). Type 5 represented widespread distribution in
almost all clusters (Fig. 5F). We found that LCK, a representative of type Given that LCK could be implicated in BU development, we therefore
4, was only expressed in clusters of activated T cells (Fig. 5E). searched the DGIdb database to explore whether it could be targeted by
drugs or belonged to putatively druggable genes. The result showed that

5
Y. Deng et al. Journal of Autoimmunity 133 (2022) 102920

Table 1
The information of seven hub genes.
Hub genes Degree Closeness EPC MNC Radiality Total Rank log2 Padj
Score Score Score Score Score Score Fold Change
CD8A 120 482.7 73.7 115 10.4 801.8 1 3.48 7.84E-09
LCK 109 480.1 68.4 109 10.4 776.9 2 4.08 1.68E-18
TNF 99 477.2 67.1 97 10.4 750.7 3 3.83 2.56E-05
EGFR 103 482.6 59.2 95 10.4 750.3 4 − 1.44 2.22E-05
MYC 83 469.0 58.8 76 10.4 697.2 5 4.09 0.0009
CXCL12 78 452.2 71.8 75 10.3 687.3 6 1.62 0.005
SYK 81 453.5 58.0 79 10.3 681.8 7 1.94 2.94E-05

Fig. 4. Validation of the key hub gene LCK. (A) LCK expression level based on mRNA sequencing data (n = 4).(B) Quantification of LCK mRNA level by RT-qPCR (n
= 5). (C) Representative Western blot images of LCK protein and its phosphorylation. (D–F) Quantification of the Western blot data of LCK protein level and its
phosphorylation level (n = 5). BU, Behcet’s uveitis; HC, healthy controls.

LCK could be a potentially druggable gene, with 59 drugs possibly tar­ suggesting numerous molecular pathological changes occurring in
geting it. Fig. 6 presents drugs having a well-defined interaction (in­ ocular lesions. The GO, KEGG, and PPI network analyses for these DEGs
hibitor) with LCK. A total of 16 drugs could act as its inhibitors, of which highlighted T cell-mediated biological processes, suggesting an active
JNJ-26483327 and NINTEDANIB were the top two with the highest participation of T cells in disease progression. The GSEA further indi­
interaction score. cated that these T cell-mediated immune responses were closely related
to the development of intraocular inflammation. These results are
4. Discussion principally consistent with histopathological findings reported by
George et al. They showed a predominant infiltration of T cells in the
This study, for the first time, presents comprehensive transcriptomic retina and uvea from an enucleated BU eye [28]. Previous studies also
profiling of the iris obtained from BU patients and healthy controls. Bulk reported that T cells were the main infiltrating cells in the aqueous
RNA-seq data showed 1633 dysregulated genes in the BU group. humor of BU patients [29,30]. Moreover, our previous studies revealed
Enrichment analyses (KEGG, GO, and GSEA) highlighted the involve­ that active BU patients had hyperactive Th1 and Th17 responses in their
ment of T cell-mediated biological processes and their association with peripheral blood [31,32]. Other studies of peripheral inflammatory sites
the BU phenotype. LCK was identified as the key hub gene in the PPI also underlined additional contributors to BU pathogenesis, such as
network, and its phosphorylation level at Y394 was increased in the BU CD68+ macrophages, neutrophils, and natural killer cells [33–35].
group. Additionally, we revealed five cellular expression patterns of These studies on different inflammatory sites collectively unravel the
DEGs in the healthy iris using the scRNA-seq data. LCK was expressed in complicated pathogenesis of BU.
clusters of activated T cells but not in other iris cell clusters, suggesting Compared with CD4+ T cells, the contribution of CD8+ T cells in BD
that LCK dysregulation was related to disturbance of T cell homeostasis. pathogenesis is less investigated. In agreement with previous studies
The BU is one of the most severe entities of uveitis, primarily [36,37], our PPI network analysis for DEGs also provided evidence for
affecting the retina and the uvea [27]. Considering the almost inacces­ the participation of CD8+ T cells in intraocular immunopathogenesis.
sibility of other intraocular tissues such as the retina, ciliary body, and We found that CD8A was the top-ranked hub gene in the PPI network,
choroid, we focused on the iris of BU patients to explore intraocular suggesting that CD8+ T cells may play a pivotal role in the development
immunopathogenesis of this disease. In this study, the bulk RNA-seq of of intraocular inflammation. One previous study also demonstrated that
the iris identified 1633 DEGs between the BU group and controls, CD8+ T cells were the predominant infiltrating cells in the aqueous

6
Y. Deng et al. Journal of Autoimmunity 133 (2022) 102920

Fig. 5. Constitutive expression pattern of the DEGs in the distinct iris cell clusters. (A) The tSNE plots of the scRNA-seq data of the healthy iris showing eight distinct
iris cell clusters. (B–F) Representative tSNE plots exhibiting five types of distribution features of the DEGs in the distinct iris cell clusters. Purple intensity denotes the
normalized gene expression level. Dark purple corresponds to high expression, grey indicates no expression. DEGs, differentially expressed genes.

and cytotoxic potential of CD8+ T cells might partially explain the severe
intraocular destructive property of BU as compared with other nonin­
fectious uveitis. Recently, Vural et al. reported that CD8+ T cells were
the predominant T-cell subset and the main IL-17A-producing cells in
papulopustular skin lesions of BD patients, contributing to recruitment
and activation of neutrophils [38]. However, the specific functions of
CD8+ T cells and their interactions with other inflammatory infiltrating
cells in the ocular lesions of BU patients remain unclear, which need to
be clarified in future studies.
Another intriguing result in our PPI network analysis was the iden­
tification of a previously undescribed LCK in the BU. LCK, a tyrosine
kinase belonging to the non-receptor Src family, has been shown to be
expressed in a variety of cell types, such as T cells, B cells, NK cells,
neurons, and tumor cells, and is essential for activating signaling cas­
cades downstream of multiple receptors [39–41]. LCK overexpression
has been demonstrated to be strongly correlated with the development
of other autoimmune or chronic inflammatory diseases, such as psori­
asis, Type 1 diabetes, asthma, rheumatoid arthritis, inflammatory bowel
diseases, atherosclerosis, systemic lupus erythematosus, and dermato­
myositis [42]. In this study, we also found that LCK expression was
evidently upregulated in the iris of BU patients, along with a consider­
Fig. 6. Drugs inhibiting LCK based on the Drug–Gene Interaction Database. able increase in its activity. Its upregulation was also observed in the iris
of AAU and VKH patients and in the BD-related skin lesions. These
findings indicate that LCK signaling may be a crucial molecular pathway
humor of patients with active BU, whereas CD4+ T cells were mostly
observed in patients with other noninfectious uveitis such as VKH and shared by the above diseases. Interestingly, our scRNA-seq results
revealed that LCK was expressed in clusters of activated T cells but not in
idiopathic uveitis [37]. They speculated that the distinct predominance

7
Y. Deng et al. Journal of Autoimmunity 133 (2022) 102920

other iris cell clusters, suggesting a close association between LCK dys­ Appendix A. Supplementary data
regulation and disturbance of T cell homeostasis in the inflamed iris. It is
worth pointing out that LCK has been widely recognized to have a Supplementary data to this article can be found online at https://doi.
critical role in T cell-mediated biological processes such as T cell org/10.1016/j.jaut.2022.102920.
development, differentiation, and function [42]. Moreover, a recent
study showed that suppression of LCK signaling improved psoriatic References
symptoms by reducing Th1/Th17 immune responses and increasing
Treg cells [43]. Likewise, Luo et al. reported that LCK inhibition [1] A. Hedayatfar, Behçet’s disease: autoimmune or autoinflammatory? J. Ophthalmic
Vis. Res. 8 (2013) 291–293.
significantly alleviated pulmonary inflammation and improved lung [2] A. Greco, A. De Virgilio, M. Ralli, A. Ciofalo, P. Mancini, G. Attanasio, M. de
function by lowering Th2 cytokines production in asthma mice [44]. Vincentiis, A. Lambiase, Behçet’s disease: new insights into pathophysiology,
These results further suggest that LCK blockade may also have beneficial clinical features and treatment options, Autoimmun. Rev. 17 (2018) 567–575,
https://doi.org/10.1016/j.autrev.2017.12.006.
effects on the reduction of BU intraocular inflammation by influencing [3] The International Criteria for Behçet’s Disease (ICBD), A collaborative study of 27
various T cell-mediated immune responses. Therefore, our study may countries on the sensitivity and specificity of the new criteria, J. Eur. Acad.
provide a new and promising target for the study of BU treatment, which Dermatol. Venereol. 28 (2014) 338–347, https://doi.org/10.1111/jdv.12107.
[4] E.T. Cunningham Jr., I. Tugal-Tutkun, M. Khairallah, A.A. Okada, B. Bodaghi,
deserves future confirmation. M. Zierhut, Behçet Uveitis. Ocul Immunol Inflamm. 25 (2017) 2–6, https://doi.
Our study has several limitations. Firstly, the sample size used in this org/10.1080/09273948.2017.1279840.
study was limited due to the small number of BU patients receiving the [5] T. Özgürtaş, Ç. Yücel, E. Sertoğlu, Y. Hayran, S. Çolak, E. Tekgöz, A. Omma, A.
H. Durukan, Evaluation of the relationship of lymphangiogenesis markers with
preventive peripheral iridectomy. Secondly, as this study only included
disease pathogenesis in patients with Behçet’s uveitis, Acta Clin. Belg. 77 (2022)
the patients with a relatively long course of disease, our results did not 387–395, https://doi.org/10.1080/17843286.2021.1890451.
address the immunopathogenesis of the entire disease course. Finally, [6] S. Hou, D. Liao, J. Zhang, J. Fang, L. Chen, J. Qi, Q. Zhang, Y. Liu, L. Bai, Y. Zhou,
these enrolled patients were on systemic corticosteroids and immuno­ A. Kijlstra, P. Yang, Genetic variations of IL17F and IL23A show associations with
Behçet’s disease and Vogt-Koyanagi-Harada syndrome, Ophthalmology 122 (2015)
suppressants before surgery to achieve a clinically quiescent status, 518–523, https://doi.org/10.1016/j.ophtha.2014.09.025.
which could have influenced our transcriptome data. [7] Z. Zhong, G. Su, A. Kijlstra, P. Yang, Activation of the interleukin-23/interleukin-
In conclusion, our study highlighted the involvement of T cell- 17 signalling pathway in autoinflammatory and autoimmune uveitis, Prog. Retin.
Eye Res. 80 (2021), 100866, https://doi.org/10.1016/j.preteyeres.2020.100866.
mediated immunity and LCK signaling in intraocular immunopatho­ [8] X.F. Huang, M.A. Brown, Progress in the genetics of uveitis, Gene Immun. 23
genesis of BU patients. Our results also provide evidence that LCK (2022) 57–65, https://doi.org/10.1038/s41435-022-00168-6.
upregulation may be related to T cell-mediated immune dysregulation. [9] J. Shan, P. Zhou, Z. Liu, K. Zheng, X. Jin, L. Du, Association of miRNA-146a gene
polymorphism Rs2910164 with Behcet’s disease: a meta-analysis, Ocul. Immunol.
Therefore, LCK inhibition might serve as a new therapeutic strategy to Inflamm. (2021) 1–7, https://doi.org/10.1080/09273948.2021.1968002.
improve intraocular inflammation for BU patients. [10] A.M. Abu El-Asrar, N. Berghmans, S.A. Al-Obeidan, P.W. Gikandi, G. Opdenakker,
J. Van Damme, S. Struyf, Local cytokine expression profiling in patients with
specific autoimmune uveitic entities, Ocul. Immunol. Inflamm. 28 (2020) 453–462,
Author contributions https://doi.org/10.1080/09273948.2019.1604974.
[11] J. Xu, Y. Qin, R. Chang, H. Tan, Q. Wang, G. Su, Q. Cao, A. Kijlstra, P. Yang,
Yang Deng: Conceptualization, Formal analysis, Investigation, Data Aqueous cytokine levels in four common uveitis entities, Int. Immunopharm. 78
(2020), 106021, https://doi.org/10.1016/j.intimp.2019.106021.
Curation, Writing - Original Draft, Writing - Review & Editing. Yinan
[12] L. Wildschütz, D. Ackermann, A. Witten, M. Kasper, M. Busch, S. Glander,
Zhang: Formal analysis, Investigation. Tao Cai: Data Curation, Writing - H. Melkonyan, K. Walscheid, C. Tappeiner, S. Thanos, A. Barysenka, J. Koch,
Review & Editing. Qingfeng Wang: Formal analysis, Writing - Review & C. Heinz, B. Laffer, D. Bauer, M. Stoll, S. König, A. Heiligenhaus, Transcriptomic
Editing. Wanyun Zhang: Formal analysis. Zhijun Chen: Investigation. and proteomic analysis of iris tissue and aqueous humor in juvenile idiopathic
arthritis-associated uveitis, J. Autoimmun. 100 (2019) 75–83, https://doi.org/
Xiang Luo: Investigation. Guannan Su: Writing - Review & Editing. 10.1016/j.jaut.2019.03.004.
Peizeng Yang: Conceptualization, Resources, Supervision, Writing - [13] M.H. Errera, A. Pratas, S. Fisson, T. Manicom, M. Boubaya, N. Sedira, E. Héron,
Review & Editing, Funding acquisition. L. Merabet, A. Kobal, V. Levy, J.M. Warnet, C. Chaumeil, F. Brignole-Baudouin, J.
A. Sahel, P. Goldschmidt, B. Bodaghi, C. Bloch-Queyrat, Cytokines, chemokines
and growth factors profile in human aqueous humor in idiopathic uveitis, PLoS
Funding One 17 (2022), e0254972, https://doi.org/10.1371/journal.pone.0254972.
[14] M. Bonacini, A. Soriano, L. Cimino, L. De Simone, E. Bolletta, F. Gozzi, F. Muratore,
M. Nicastro, L. Belloni, A. Zerbini, L. Fontana, C. Salvarani, S. Croci, Cytokine
This work was supported by the Natural Science Foundation Major profiling in aqueous humor samples from patients with non-infectious uveitis
International (Regional) Joint Research Project [grant numbers associated with systemic inflammatory diseases, Front. Immunol. 11 (2020) 358,
81720108009]; the National Natural Science Foundation Key Program https://doi.org/10.3389/fimmu.2020.00358.
[15] Y. Lei, R. Tang, J. Xu, W. Wang, B. Zhang, J. Liu, X. Yu, S. Shi, Applications of
[grant numbers 81930023]; the Chongqing Outstanding Science Project single-cell sequencing in cancer research: progress and perspectives, J. Hematol.
[grant numbers 2019]; the Chongqing Chief Medical Scientist Project Oncol. 14 (2021) 91, https://doi.org/10.1186/s13045-021-01105-2.
[grant numbers 2018]; the Chongqing Key Laboratory of Ophthal­ [16] X. Li, Z. Liao, Z. Deng, N. Chen, L. Zhao, Combining bulk and single-cell RNA-
sequencing data to reveal gene expression pattern of chondrocytes in the
mology [grant numbers CSTC, 2008CA5003]; the Chongqing Science &
osteoarthritic knee, Bioengineered 12 (2021) 997–1007, https://doi.org/10.1080/
Technology Platform and Base Construction Program [grant numbers 21655979.2021.1903207.
cstc2014pt-sy10002]; and the Key Project of Chongqing Science and [17] L. Liang, J. Yu, J. Li, N. Li, J. Liu, L. Xiu, J. Zeng, T. Wang, L. Wu, Integration of
Technology Bureau [grant numbers CSTC2021jscx-gksb-N0010]. The scRNA-seq and bulk RNA-seq to analyse the heterogeneity of ovarian cancer
immune cells and establish a molecular risk model, Front. Oncol. 11 (2021),
funding organization had no role in the design or conduct of this 711020, https://doi.org/10.3389/fonc.2021.711020.
research. [18] W. Zheng, X. Wang, J. Liu, X. Yu, L. Li, H. Wang, J. Yu, X. Pei, C. Li, Z. Wang,
M. Zhang, X. Zeng, F. Zhang, C. Wang, H. Chen, H.Z. Chen, Single-cell analyses
highlight the proinflammatory contribution of C1q-high monocytes to Behçet’s
Declaration of competing interest disease, Proc. Natl. Acad. Sci. U. S. A. 119 (2022), e2204289119, https://doi.org/
10.1073/pnas.2204289119.
No conflicting relationship exists for any author. [19] Criteria for diagnosis of behçet’s disease. International study group for behçet’s
disease, Lancet 335 (1990) 1078–1080.
[20] A. Subramanian, P. Tamayo, V.K. Mootha, S. Mukherjee, B.L. Ebert, M.A. Gillette,
Data availability A. Paulovich, S.L. Pomeroy, T.R. Golub, E.S. Lander, J.P. Mesirov, Gene set
enrichment analysis: a knowledge-based approach for interpreting genome-wide
expression profiles, Proc. Natl. Acad. Sci. U. S. A. 102 (2005) 15545–15550,
Data will be made available on request. https://doi.org/10.1073/pnas.0506580102.
[21] N. Fenouille, C.F. Bassil, I. Ben-Sahra, L. Benajiba, G. Alexe, A. Ramos, Y. Pikman,
A.S. Conway, M.R. Burgess, Q. Li, F. Luciano, P. Auberger, I. Galinsky, D.
J. DeAngelo, R.M. Stone, Y. Zhang, A.S. Perkins, K. Shannon, M.T. Hemann,

8
Y. Deng et al. Journal of Autoimmunity 133 (2022) 102920

A. Puissant, K. Stegmaier, The creatine kinase pathway is a metabolic vulnerability [34] K.M. Verrou, N.I. Vlachogiannis, G. Ampatziadis-Michailidis, P. Moulos, G.
in EVI1-positive acute myeloid leukemia, Nat. Med. 23 (2017) 301–313, https:// A. Pavlopoulos, P. Hatzis, G. Kollias, P.P. Sfikakis, Distinct transcriptional profile of
doi.org/10.1038/nm.4283. blood mononuclear cells in Behçet’s disease: insights into the central role of
[22] X. Li, X. Liu, X. Pang, Y. Yin, H. Yu, Y. Yuan, B. Li, Transcriptomic analysis reveals neutrophil chemotaxis, Rheumatology 60 (2021) 4910–4919, https://doi.org/
hub genes and subnetworks related to ROS metabolism in Hylocereus undatus 10.1093/rheumatology/keab052.
through novel superoxide scavenger trypsin treatment during storage, BMC [35] U.C. Kucuksezer, E. Aktas-Cetin, S. Bilgic-Gazioglu, I. Tugal-Tutkun, A. Gül,
Genom. 21 (2020) 437, https://doi.org/10.1186/s12864-020-06850-1. G. Deniz, Natural killer cells dominate a Th-1 polarized response in Behçet’s
[23] S.L. Freshour, S. Kiwala, K.C. Cotto, A.C. Coffman, J.F. McMichael, J.J. Song, disease patients with uveitis, Clin. Exp. Rheumatol. 33 (2015) S24–S29.
M. Griffith, O.L. Griffith, A.H. Wagner, Integration of the drug-gene interaction [36] J.K. Ahn, H.G. Yu, H. Chung, Y.G. Park, Intraocular cytokine environment in active
database (DGIdb 4.0) with open crowdsource efforts, Nucleic Acids Res. 49 (2021) Behçet uveitis, Am. J. Ophthalmol. 142 (2006) 429–434, https://doi.org/10.1016/
D1144–d1151, https://doi.org/10.1093/nar/gkaa1084. j.ajo.2006.04.016.
[24] P. Gautam, K. Hamashima, Y. Chen, Y. Zeng, B. Makovoz, B.H. Parikh, H.Y. Lee, K. [37] H.G. Yu, D.S. Lee, J.M. Seo, J.K. Ahn, Y.S. Yu, W.J. Lee, H. Chung, The number of
A. Lau, X. Su, R.C.B. Wong, W.K. Chan, H. Li, T.A. Blenkinsop, Y.H. Loh, Multi- CD8+ T cells and NKT cells increases in the aqueous humor of patients with
species single-cell transcriptomic analysis of ocular compartment regulons, Nat. Behçet’s uveitis, Clin. Exp. Immunol. 137 (2004) 437–443, https://doi.org/
Commun. 12 (2021) 5675, https://doi.org/10.1038/s41467-021-25968-8. 10.1111/j.1365-2249.2004.02536.x.
[25] Y. Deng, Y. Fu, L. Sheng, Y. Hu, L. Su, J. Luo, C. Yan, W. Chi, The regulatory NOD- [38] S. Vural, K. Kerl, P. Ertop Doğan, S. Vollmer, U. Puchta, M. He, Y. Arakawa, A.
like receptor NLRC5 promotes ganglion cell death in ischemic retinopathy by O. Heper, A. Karal-Öktem, D. Hartmann, A. Boyvat, J.C. Prinz, A. Arakawa,
inducing microglial pyroptosis, Front. Cell Dev. Biol. 9 (2021), 669696, https:// Lesional activation of T(c) 17 cells in Behçet disease and psoriasis supports HLA
doi.org/10.3389/fcell.2021.669696. class I-mediated autoimmune responses, Br. J. Dermatol. 185 (2021) 1209–1220,
[26] C.E. Rudd, How the discovery of the CD4/CD8-p56(lck) complexes changed https://doi.org/10.1111/bjd.20643.
immunology and immunotherapy, Front. Cell Dev. Biol. 9 (2021), 626095, https:// [39] U. Bommhardt, B. Schraven, L. Simeoni, Beyond TCR signaling: emerging functions
doi.org/10.3389/fcell.2021.626095. of lck in cancer and immunotherapy, Int. J. Mol. Sci. 20 (2019), https://doi.org/
[27] P. Yang, Z. Zhang, H. Zhou, B. Li, X. Huang, Y. Gao, L. Zhu, Y. Ren, J. Klooster, 10.3390/ijms20143500.
A. Kijlstra, Clinical patterns and characteristics of uveitis in a tertiary center for [40] S.J. Bozso, J.J.H. Kang, J. Nagendran, The role of competing mechanisms on Lck
uveitis in China, Curr. Eye Res. 30 (2005) 943–948, https://doi.org/10.1080/ regulation, Immunol. Res. 68 (2020) 289–295, https://doi.org/10.1007/s12026-
02713680500263606. 020-09148-2.
[28] R.K. George, C.C. Chan, S.M. Whitcup, R.B. Nussenblatt, Ocular immunopathology [41] A. Elkamhawy, E.M.H. Ali, K. Lee, New horizons in drug discovery of lymphocyte-
of Behçet’s disease, Surv. Ophthalmol. 42 (1997) 157–162, https://doi.org/ specific protein tyrosine kinase (Lck) inhibitors: a decade review (2011-2021)
10.1016/s0039-6257(97)00026-x. focussing on structure-activity relationship (SAR) and docking insights, J. Enzym.
[29] R. Belfort Jr., N.C. Moura, N.F. Mendes, T and B lymphocytes in the aqueous humor Inhib. Med. Chem. 36 (2021) 1574–1602, https://doi.org/10.1080/
of patients with uveitis, Arch. Ophthalmol. 100 (1982) 465–467, https://doi.org/ 14756366.2021.1937143.
10.1001/archopht.1982.01030030467019. [42] P. Kumar Singh, A. Kashyap, O. Silakari, Exploration of the therapeutic aspects of
[30] H. Keino, J. Sakai, K. Nishioka, T. Sumida, M. Usui, Clonally accumulating T cells Lck: a kinase target in inflammatory mediated pathological conditions, Biomed.
in the anterior chamber of Behçet disease, Am. J. Ophthalmol. 130 (2000) Pharmacother. 108 (2018) 1565–1571, https://doi.org/10.1016/j.
243–245, https://doi.org/10.1016/s0002-9394(00)00498-0. biopha.2018.10.002.
[31] W. Chi, X. Zhu, P. Yang, X. Liu, X. Lin, H. Zhou, X. Huang, A. Kijlstra, Upregulated [43] N.O. Al-Harbi, S.F. Ahmad, M. Almutairi, A.Z. Alanazi, K.E. Ibrahim, S.A. Alqarni,
IL-23 and IL-17 in Behçet patients with active uveitis, Invest. Ophthalmol. Vis. Sci. F. Alqahtani, K. Alhazzani, M. Alharbi, F. Alasmari, A. Nadeem, Lck signaling
49 (2008) 3058–3064, https://doi.org/10.1167/iovs.07-1390. inhibition causes improvement in clinical features of psoriatic inflammation
[32] Z. Ye, B. Deng, C. Wang, D. Zhang, A. Kijlstra, P. Yang, Decreased B and T through reduction in inflammatory cytokines in CD4+ T cells in imiquimod mouse
lymphocyte attenuator in Behcet’s disease may trigger abnormal Th17 and Th1 model, Cell. Immunol. 376 (2022), 104531, https://doi.org/10.1016/j.
immune responses, Sci. Rep. 6 (2016), 20401, https://doi.org/10.1038/srep20401. cellimm.2022.104531.
[33] S. Cho, J. Kim, S.B. Cho, Z. Zheng, M.J. Choi, D.Y. Kim, D. Bang, [44] S. Zhang, R. Yang, Y. Zheng, The effect of siRNA-mediated lymphocyte-specific
Immunopathogenic characterization of cutaneous inflammation in Behçet’s protein tyrosine kinase (Lck) inhibition on pulmonary inflammation in a mouse
disease, J. Eur. Acad. Dermatol. Venereol. 28 (2014) 51–57, https://doi.org/ model of asthma, Int. J. Clin. Exp. Med. 8 (2015) 15146–15154.
10.1111/jdv.12054.

You might also like