You are on page 1of 11

Clinical Immunology 245 (2022) 109184

Contents lists available at ScienceDirect

Clinical Immunology
journal homepage: www.elsevier.com/locate/yclim

Single-cell mapping reveals dysregulation of immune cell populations and


VISTA+ monocytes in myasthenia gravis
Rui Fan a, 1, Wenjun Que a, b, 1, Zhuoting Liu a, Wei Zheng a, Xia Guo a, c, Linqi Liu a, Fei Xiao a, *
a
Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
b
Department of Blood Transfusion, the First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing 400016, China
c
Department of Neurology, the Second Affiliated Hospital of Zunyi Medical University, The Intersection of Xinlong Avenue and Xinpu Avenue in New Pu District, Zunyi
563000, China

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

Keywords: The pathogenesis and progression of myasthenia gravis (MG), an autoimmune disease, involve abnormal func­
Myasthenia gravis tion and composition of several immune cell populations. However, details of this dysregulation remain unclear.
Cohort We performed a cross-section analysis using cytometry time-of-flight on blood samples from 12 generalized MG
Immune
without glucocorticoid or other immunosuppressant treatment, and 10 sex- and age-matched healthy controls.
monocyte
NK cell
Combining data from an external validation cohort (MG n = 38, control n = 21), bulk-RNA sequencing and
Mass cytometry single-cell RNA sequencing, alterations in immune cell populations and differential expression of immune check
Immune check point point were revealed. Several switched memory B cell subsets (CD3- CD19+ CD27+ IgD- CD38+/− ) were
increased in MG patients. The number of HLA- DQ- CD38+ naïve B cells was higher in MG patients and
correlated with the quantitative MG score (QMG). Among NK cells, the number of CD56+ CD16+ NK cells and
CD56+ CD16+ CD8+ NK cells were decreased in MG patients and positively correlated with QMG. VISTA+
monocytes were increased in MG patients. Classical T cell subsets showed no significant change; however, the
expression of VISTA, LAG3, CTLA4, and CXCR5 was higher in T cells from MG patients. The expression of CD38
was higher in neutrophils from MG patients. The external validation cohort validated the dysregulation of NK cell
subtypes, and differences were also observed in subgroups of patients. Bulk-RNA sequencing also revealed
increased mRNA expression of VSIR in monocytes of MG patients compared to those from healthy controls, and
the antigen presentation and processing pathway was identified as enriched in the functional characterization of
VISTA+ monocytes via single-cell RNA sequencing. Our study revealed alterations in several immune cell subsets
and identified potential cellular biomarkers for MG diagnosis and disease severity assessment. In addition, the
abnormal expression of multiple immune checkpoints in MG provides further rationale for the investigation of
immune-checkpoint-related therapy.

1. Introduction that include ptosis, dysphagia, limb weakness, and even dyspnea. The
incidence rate of MG is 1.7 to 21.3 cases per million person-years, and
Myasthenia gravis (MG) is a typical T cell-dependent autoimmune the prevalence rate is 15 to 179 cases per million persons [2]. The
disease mediated by a series of autoantibodies, including anti- Myasthenia Gravis Foundation of America's clinical classification is
acetylcholine receptor (AchR), anti-muscle-specific receptor tyrosine based on disease severity; with class I corresponding to ocular MG
kinase, anti-lipoprotein-receptor-related protein 4, anti-agrin, anti-rya­ (OMG), and classes II to V to generalized MG (GMG) [3]. More than half
nodine receptor, and anti-titin. [1] These antibodies are produced from of OMG cases will progress to GMG within three years, and about 20% of
autoreactive B cells activated by CD4+ T cells and bind to functional all MG cases progress to crisis within 1–3 years for unknown reasons.
molecules in the postsynaptic membrane, causing impairment of signal Although anti-AchR is a pathogenic antibody in over 80% of MG
transmission in synapses and resulting in a series of clinical symptoms cases and is involved in the pathogenesis of MG via multiple

* Corresponding author at: Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing
400016, China.
E-mail address: feixiao81@126.com (F. Xiao).
1
These authors contributed equally

https://doi.org/10.1016/j.clim.2022.109184
Received 4 September 2022; Received in revised form 21 October 2022; Accepted 5 November 2022
Available online 11 November 2022
1521-6616/© 2022 Elsevier Inc. All rights reserved.
R. Fan et al. Clinical Immunology 245 (2022) 109184

mechanisms, such as complement-mediated damage to the postsynaptic 2. Materials and methods


membrane, diminishing Ach-AchR binding, and increasing AchR
degradation [4], there is no significant correlation between antibody 2.1. Ethics statement
titer and disease severity, and the underlying cause is still unclear.
Recently, several studies have indicated that specific immune cell sub­ This project was approved by the Ethics Committee of the First
sets, including follicular helper T cells (Tfh) and ThCD103, are different in Affiliated Hospital of Chongqing Medical University (2020–382). Writ­
MG and healthy controls (HC), and their relative frequencies are ten informed consent was obtained from all subjects.
significantly associated with disease severity [5,6]. However, most
studies have focused on T and B cell subsets, whereas few have focused 2.2. Clinical features of enrolled participants
on the dysregulation of natural killer (NK) or myeloid cell subsets via an
unbiased analysis method. The V-domain immunoglobulin suppressor of Patients with MG, and age- and sex-matched healthy controls (MG
T cell activation (VISTA), also known as PD-1H, is a novel immune group, n = 12; HC group, n = 10) (Table 1) were recruited from the
checkpoint (ICP) molecule mainly expressed on myeloid and T cells [7] Department of Neurology of the First Affiliated Hospital of Chongqing
that is involved in multiple diseases such as cancer and systemic lupus Medical University. Peripheral blood leukocytes were isolated within 12
erythematosus (SLE) [8]. However, the role of VISTA in myasthenia h after blood collection, and antigen expression was detected using mass
gravis remains largely unknown. cytometry. All enrolled patients satisfied the following conditions: 1)
Mass cytometry/cytometry time-of-flight (CyTOF) is a rapidly Generalized myasthenia gravis; 2. Duration of the disease >3 months; 3)
evolving technique that combines mass spectrometry and flow cytom­ Not receiving glucocorticoids or other immunosuppressant treatments,
etry, and provides significant advantages for the discovery of new cell or treatment having been discontinued for >3 months; 4) Not affected
subsets, revealing abnormal immune statuses in autoimmune diseases by any other autoimmune, inflammatory, or infectious diseases, or
and oncologic disorders by simultaneous quantitative detection of more pregnancy; 5) Not affected by thymoma. Data from the external vali­
than 40 proteins in a single cell. Recently, Ingelfinger et al. [6] dation cohort (MG, n = 38; Control (CTRL), n = 21) were downloaded
demonstrated via CyTOF that the main population of immune cells was from Mendeley Data (https://data.mendeley.com/datasets/nkcb8nc
similar in MG patients and in healthy controls, and described an alter­ 7w8/1) [6].
ation in the ThCD103 and ThGM cells from the blood and thymus in MG
patients. However, an in-depth investigation of other immune cell sub­ 2.3. Mass cytometry
sets and novel ICP expression in MG is lacking at present. Moreover,
multiple studies have indicated that immune status, as well as the status Metal-tagged antibodies (Supplementary Table 1) were purchased
of the thymus and potential treatment strategies, are different in OMG from Fluidigm Corporation (CA, USA). Cell labeling was performed as
and in GMG, and we considered these factors to be potential con­ previously described [9]. Briefly, after peripheral blood leukocytes were
founders that may lead to biased results. We ruled out potential con­ isolated, single-cell suspensions were stained for 5 min with a 5 μM
founders in this study by recruiting 12 GMG patients and 10 age- and solution of 194Pt cisplatin (Fluidigm) to distinguish between live and
sex-matched HCs. Phenotypic alterations of immune cell populations dead cells, then blocked with phosphate buffered saline containing 5%
were evaluated via CyTOF using 42 markers. The main altered cell goat serum and 30% bovine serum albumin (BSA) for 30 min at 4 ◦ C.
populations were validated by external validation cohort, main altered Next, they were stained with different surface markers on ice for 30 min
ICP in mRNA level and functional characterization of novel cell popu­ at 4 ◦ C. The Foxp3 Fixation and Permeabilization kit (eBioscience) was
lation revealed by bulk-RNA-sequencing and single-cell RNA- used according to the instructions of the manufacturer. After washing,
sequencing. resuspension, and centrifugation, cells were incubated overnight. Next,
after being washed twice with Foxp3 permeabilization and fluorescence-
activated cell sorting (FACS) buffer, cells were incubated with

Table 1
Information of enrolled participants.
Participants Gender Age MGFA Thymoma Anti-AchR Anti-Musk Anti-LRP4 Course (Month)
(M/F) (Y) classification

MG1 F 72 IIIB None Positive Negative Negative 41


MG2 F 47 IIIA None Positive Negative Negative 19
MG3 M 40 IIIA None Positive Negative Negative 60
MG4 F 50 IIA None Positive Negative Negative 163
MG5 F 49 IIB None Positive Negative Negative 223
MG6 F 53 IIB None Positive Negative Negative 27
MG7 F 21 IIIA None Positive Negative Negative 10
MG8 F 48 IIA None Positive Negative Negative 225
MG9 M 57 IIIB None Positive Negative Negative 34
MG10 M 24 IIIB None Positive Negative Negative 21
MG11 M 44 IIA None Positive Negative Negative 18
MG12 M 27 IIIA None Positive Negative Negative 24
HC1 F 66 NA None ND ND ND NA
HC2 F 47 NA None ND ND ND NA
HC3 F 52 NA None ND ND ND NA
HC4 F 25 NA None ND ND ND NA
HC5 M 34 NA None ND ND ND NA
HC6 F 58 NA None ND ND ND NA
HC7 M 24 NA None ND ND ND NA
HC8 F 58 NA None ND ND ND NA
HC9 F 72 NA None ND ND ND NA
HC10 F 48 NA None ND ND ND NA

AchR: acetylcholine receptors; F, female; HC: healthy controls; LRP4: lipoprotein-receptor-related protein 4; M, male; MG: myasthenia gravis; MGFA: Myasthenia
Gravis Foundation of America; MuSK: muscle-specific kinase; NA, not applicable; ND, not done; Y, years.

2
R. Fan et al. Clinical Immunology 245 (2022) 109184

intracellular antibodies in permeabilization buffer for 30 min at 4 ◦ C. significantly different cell subsets (Fig. 1A). CD45+ live cells and other
Finally, after washing and resuspension, cells were analyzed within 12 h immune cell populations were defined by loading data into FlowJo (http
on a Helios CyTOF System (Fluidigm Corporation) at an event rate of s://www.flowjo.com/). The .fcs file was then loaded into R software
<500 events/s. (version 4.3.1, The R Foundation, Vienna, Austria)) using the FlowCore
R package (http://bioconductor.org/packages/flowCore/). The unsu­
2.4. Data analysis pervised clustering algorithm (PhenoGraph) and data transformation
were performed using the cytofkit R package [11], the tSNE algorithm
After data acquisition, .fcs and normalized files were obtained using was performed using the Rtsne package (https://github.com/jkrijth
a Helio3 CyTOF mass cytometer (Fluidigm Corporation) and bead-based e/Rtsne/), and the visualization was performed using the ggplot2
normalization software [10]. CD45+ cells were gated after the elimi­ (https://CRAN.R-project.org/package=ggplot2/) and the ggpubr pack­
nation of calibration beads and cell fragments, and multi-omics analyses age (https://CRAN.R-project.org/package=ggpubr).
were conducted to explore the potential biological function of novel

Fig. 1. The composition of the population of the main immune cells differed in MG patients and HCs. A. Flowchart of experimental design. B. Heatmap showing
lineage markers among the main immune cell populations. C. tSNE plot of immune cell populations in MG patients and HCs. D. Differences in the population of the
main immune cells between MG patients and HCs. Statistical analysis was performed using unpaired t-tests (significance level: NS >0.05; *0.01 < p < 0.05; **0.001
< p < 0.01; *** < 0.001). DC: dendritic cells; HC: healthy controls; MG: myasthenia gravis; NK: natural killer cells; NKT: natural killer T cells. tSNE: t-distributed
Stochastic Neighbor Embedding.

3
R. Fan et al. Clinical Immunology 245 (2022) 109184

2.5. Bulk-RNA-sequencing and single-cell RNA-sequencing data 2.6. Semi-supervised clustering algorithm, differential expression gene
analysis and functional enrichment
The gene expression profiles of bulk-RNA-sequencing and single-cell
RNA-sequencing were obtained from the Gene Expression Omnibus Semi-supervised Category Identification and Assignment (SCINA)
database (http://www.ncbi.nlm.nih.gov/geo) (GSE85452 and [12] was conducted using the SCINA package to identify VISTA+
GSE103544). In the GSE85452 dataset, CD14 + monocytes were iso­ monocytes using single-cell RNA sequencing. The Seurat package was
lated from 13 patients with MG and 12 HC. In the GSE103544 dataset, used to construct a single-cell analysis file and identify differentially-
we obtained 856 RNA-sequencing data of CD14 + monocytes from two expressed genes (DEGs) using standard MAST and DESeq2 methods.
healthy donors. Four pseudobulk-based algorithms were applied using the Libra package

Fig. 2. Phenotypic alterations exhibited by B cells and T cells in MG. A. tSNE plot of B cell clusters corresponding to MG patients and HCs. B. Heatmap showing the
markers among B cell clusters identified by the PhenoGraph algorithm. C. Differences in the two main classical B cell subsets between MG patients and HCs. D.
Annotation of B cell clusters. E. F. Five significantly different subsets of B cells. G. Correlation between the frequency of HLA- DQ- CD38+ naïve B cells and QMG.
Statistical analysis performed using unpaired t-tests (significance level: NS >0.05; *0.01 < p < 0.05; **0.001 < p < 0.01; *** < 0.001) and Pearson correlation
analysis. HC: healthy controls; MG: myasthenia gravis. tSNE: t-distributed Stochastic Neighbor Embedding.

4
R. Fan et al. Clinical Immunology 245 (2022) 109184

[13]. Transcription factor enrichment analysis was performed using 3.3. Phenotypic alteration of NK cells in MG
CHEA3 (https://maayanlab.cloud/chea3/). Gene ontology (GO) and
Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrich­ In addition to T and B cells, other immune cells contribute to the
ment were performed using ClusterProfiler (https://guangchuangyu. regulation of immune responses in direct or indirect ways. Approxi­
github.io/software/clusterProfiler), and visualization was performed mately 33 clusters were identified by PhenoGraph in CD3- CD19- cells,
using the GOplot R package (https://cran.r-project.org/web/packa and CD14+ monocytes, neutrophils, CD56+ NK cells, and dendritic cells
ges/GOplot/citation.html). (DCs) were identified using classical lineage markers (Fig. 3A and B,
Supplementary Table 2). Although the total frequency of neutrophils,
2.7. Statistical analysis CD56+ NK cells, and DCs was not significantly different between the HC
and the MG groups, the tSNE map showed heterogeneity of these cell
Statistical analysis was performed using R software, and differences populations between the groups (Fig. 3A). To explore the cell pop­
between the two groups were evaluated using either an unpaired Stu­ ulations that showed significant differences and were associated with
dent's t-test or a Mann–Whitney U test. Correlations were assessed using disease severity, a series of statistical analyses was conducted. Inter­
Spearman's analysis. estingly, the main significantly different cell populations between the
HC and MG were NK cell subsets and it correlated with QMG. The fre­
2.8. Role of the funders quencies of cluster27 and cluster29 were increased in the MG group,
those of cluster25 and cluster26 were decreased in the MG group. The
The funders had no role in the study design, data collection, data frequency of cluster29 was negatively correlated with QMG; whereas
analyses, interpretation, or writing of this report. those of cluster7, 22, and 26 were positively correlated with QMG.
Notably, cluster27 and cluster29 corresponded to CD56+ CD16- NK
3. Results cells, and cluster25 and clutser26 to CD56+ CD16+ NK cells (Fig. 3B).
CD56+ NK cells were clustered by PhenoGraph and visualized by tSNE,
3.1. Composition of main immune cell populations differed in HC and MG and the results showed that the position of CD56+ CD16+ NK cells was
almost empty in the MG group; conversely, the position of CD56+ CD16-
B cells, T cells, monocytes, NK, natural killer T cells (NKT cells), NK cells was almost empty in the HC group (Fig. 3C-E). The frequency of
neutrophils, and dendritic cells (DC) were identified by PhenoGraph CD56+ CD16+ NK cells was positively correlated with QMG. In addi­
based on the expression of lineage markers, including CD3, CD19, CD14, tion, the frequency of CD56+ CD16+ CD8+ NK cells was positively
CD56, CD66b, and HLA-DR (Fig. 1B), and the dimensionality reduction correlated to QMG and was significantly lower in in the MG group
of these cell populations was assessed by t-distributed stochastic compared to that in HCs (Fig. 3E and F). Five thousand cells were
neighbor embedding (tSNE) (Fig. 1C). Statistical analysis of the fre­ randomly extracted from each sample in the external validation cohort,
quencies of these main immune cell populations indicated that the fre­ and 25 clusters were identified by PhenoGraph based on the expression
quencies of B cells and monocytes were significantly higher in MG of CD3, CD19, CD14, CD56, CD16, and CD8. Cluster8 and cluster19
patients compared to those those in HCs, but were not significantly were identified as CD56+ CD16+ NK cells, and cluster19 as CD56+
associated with quantitative MG score (QMG). No significant differences CD16+ CD8+ NK cells (Fig. S2A and B). We found that the frequency of
were observed in T cells and other immune cell populations between the CD56+ CD16+ NK cells was significantly lower in the MG group
two groups (Fig. 1D). However, tSNE plots revealed heterogeneity in NK compared to that in the CTRL group, which validated the reliability of
cells, monocytes, and neutrophils between the MG and HC groups. our results. In addition, the results of the subgroup analysis showed that
the frequency of CD56+ CD16+ NK cells was lower in early-onset MG
3.2. Frequency of switched-memory B cells increased in MG (EOMG) than in late-onset MG (LOMG) cases, lower in patients with
azathioprine therapy than in patients without immunosuppressive
To further explore the heterogeneity of B cells in HCs and MG pa­ therapy, lower in patients with thymectomy than in those without thy­
tients, and to identify the main B cell subsets that were increased in MG mectomy, and lower in GMG patients than in OMG patients (Fig. S2C).
patients, B cells were further divided into 18 clusters by unsupervised CD56+ CD16+ CD8+ NK cells showed a similar trend (Fig. S2D). The
clustering algorithms using PhenoGraph and reduced dimensionality by frequencies of CD56+ CD16+ and CD56+ CD16+ CD8+ NK cells also
tSNE (Fig. 2A and B). These clusters could then be classified into four showed a high area under the curve for the diagnosis of MG (73% and
classical subsets according to the expression of IgD and CD27: switched 72%, respectively) (Fig. S2F). However, there was no significant corre­
memory B cells (CD27 + IgD-), non-switched B cells (CD27 + IgD+), lation between the frequencies of NK cell subsets and the titer of the anti-
naïve B cells (CD27-IgD+), and double negative B cells (CD27-IgD-). AchR antibody (Fig. S2E).
Switched memory B cells and non-switched B cells were significantly
increased in MG patients (Fig. 2C). These four classical subsets of B cells 3.4. VISTA+ monocytes were the mainly increased monocyte subset in
can be subdivided into 12 subsets in combination with other cell MG
markers (Fig. 2D), and we found that five B cell subsets were signifi­
cantly different between the HC and the MG groups (Fig. 2E, F). Cluster9 Nine clusters were identified as monocytes (Supplementary Table 2):
(HLA-DQ-CD38+ naïve B cells) was increased in MG patients and cluster6, cluster8, and cluster9 comprised approximately 94% of the
significantly associated with QMG (Fig. 2G). Using the same approach, T total monocyte population. Cluster8 and cluster9 were significantly
cells were subdivided in 30 clusters (Fig. S1A), but the frequencies of decreased and increased, respectively, in the MG group (Fig. 4A).
classical subpopulations, including naïve T cells, effector memory T cells Cluster9 was identified as CD14+ VISTA+ monocytes using the tSNE
(TEM), central memory T cells (TCM), and effector T cells (Teff), were plot (Fig. 4B). High-throughput microarray data from the GSE database
not significantly different between the HC and the MG group (Fig. S1B, (GSE:85452, isolated CD14+ monocytes from 13 MG patients and 12
S1C). Interestingly, the expression of immune checkpoints in T cells, HCs) were analyzed for VISTA mRNA expression. The results showed
including VISTA, CTLA4, and LAG3 (but not PD-1), was significantly that VISR/C10orf54 mRNA was significantly increased in monocytes
higher in MG patients, (Fig. S1D). Previous studies suggested that the from MG patients (Fig. 4C). Although VISTA is thought to serve pri­
number of Tfh was increased in MG patients and correlated with QMG marily as an inhibitory protein that limits immune responses and is most
[14], and our result showed that the expression of CXCR5 (a charac­ highly expressed in monocytes and other potential antigen-presening
teristic marker for Tfh), was significantly increased in MG patients cells (APCs) [15], a recent study indicated that VISTA antibodies can
(Fig. S1D). activate CD14+ monocytes and promote inflammation responses [16].

5
R. Fan et al. Clinical Immunology 245 (2022) 109184

Fig. 3. Phenotypic alterations of NK cells in MG. A. tSNE plot of CD3-CD19- cells corresponding to MG patients and HCs. B. Heatmap showing the markers among
clusters divided by the PhenoGraph algorithm. C. tSNE plot of CD56+ NK cells corresponding to MG patients and HCs. D. tSNE plot of CD16 expression in CD56+ NK
cells. E. Differences in NK subsets between MG patients and HCs. F. Correlation between frequencies of CD56+ CD16+ or CD56+ CD16+ CD8+ NK cells and QMG.
Statistical analysis performed using unpaired t-tests (significance level: ns >0⋅05; *0⋅01 < p < 0⋅05; **0⋅001 < p < 0⋅01; *** < 0⋅001) and Pearson correlation
analysis (significant level: ns >0.05; #0.01 < p < 0.05; ##0.001 < p < 0.01; ### < 0.001). HC: healthy controls; MG: myasthenia gravis. tSNE: t-distributed
Stochastic Neighbor Embedding.

To further explore the biological features of VISTA, identifying the gene and PSME, were included in antigen processing and presentation
signature and performing the functional characterization of VISTA+ pathway (Fig. 5D). From our mass cytometry data, we found that HLA-
CD14+ monocytes was crucial. To achieve this, we used CD14+ DR expression was indeed increased in CD14+ VISTA+ monocytes
monocyte single-cell RNA sequencing datasets from GSE103544 (856 (Fig. 5E). Transcription factor enrichment analysis was performed on the
CD14+ monocytes RNA-seq data), VSIR+ monocytes (VSIR+ Mo), and 37 gene signatures, and the results showed that IRF8 and IRF5 were the
VSIR- monocytes (VSIR-Mo), which were clearly identified by a semi- most enriched transcription factors (TFs), and that VSIR may be regu­
supervised clustering algorithm known as SCINA (Fig. 4D and E). Four lated by TFEB (Fig. S3, Supplementary File2). In addition, although the
pseudobulk-based algorithms (DESeq2-Wald, DESeq2-LRT, edgeR-LRT, frequencies of neutrophil subsets were not significantly different, the
and edgeR-QLF) and two classical algorithms (MAST and DESeq2) were tSNE plot revealed heterogeneity in neutrophils from the MG and the HC
used to identify significantly differentially expressed genes (DEGs). group. We found that CD38+ neutrophils were enriched in MG patients
VSIR+ Mo gene signatures were identified as intersections of the results (Fig. 5F), and the expression of CD38 in neutrophils from these patients
of the six algorithms. In total, 37 genes were upregulated and identified was significantly increased (Fig. 5G).
as gene signatures, but no common downregulated genes were observed
(Fig. 4F-G and Fig. 5A, Supplementary File 1). Next, The 37 gene sig­ 4. Discussion
natures were subjected to KEGG pathway enrichment analysis and GO
functional enrichment analysis. Antigen processing and presentation, Our study provides an expression landscape of multiple immune
and related functional pathways were the most significantly enriched markers in peripheral immune cells using CyTOF. We observed that the
pathways (Fig. 5B and C). Eight genes, including HLA gene family CD74 frequencies of CD56+ CD16+ and CD56+ CD16+ CD8+ NK cells

6
R. Fan et al. Clinical Immunology 245 (2022) 109184

Fig. 4. Alterations in CD14+ VISTA+ monocytes and identified gene signatures via single-cell RNA sequencing. A. Frequencies of cluster8 and cluster9 were
decreased and increased in MG, respectively. B. tSNE plot showing the alteration of cluster8 and cluster9; cluster9 was identified as VISTA+ CD14+ monocytes. C.
mRNA expression of VSIR in monocytes from MG patients (n = 13) and HCs (n = 12). D. Density plot showing the expression of VSIR in VSIR+ and VSIR- Mo. E.
Violin plot showing the expression of VSIR in VSIR+ and VSIR- Mo. F. Interaction of up-regulated genes identified by six algorithms. G. Interaction of down-regulated
genes identified by six algorithms. Statistical analysis performed using unpaired t-tests (significance level: NS >0.05; *0.01 < p < 0.05; **0.001 < p < 0.01; *** <
0.001). HC: healthy controls; MG: myasthenia gravis; p_DGEseq2_LRT: pseudobulk DESeq2 LRT; p_DGEseq2_wald: pseudobulk DESeq2 Wald; p_edgeR_LRT: pseu­
dobulk edgeR LRT; p_edgeR_QLF: pseudobulk edgeR QLF; VSIRneg_Mo: VSIR-Monocytes; VSIRpos_Mo: VSIR+Monocytes.

decreased and were significantly associated with the QMG score. In the potential of CD56+ CD16+ and CD56+ CD16+ CD8+ NK cells as
external validation cohort, we validated the differences exhibited by diagnostic cellular markers and indicators of disease severity.
these two NK cell subsets in the MG and the CTRL groups, and showed NK cells are innate lymphoid cells that are generally identified as
significant differences between GMG and OMG patients, between pa­ CD56+ CD3- in humans. Human NK cells can be classified into two
tients treated with and without thymectomy, between patients treated subsets (CD56bright and CD56dim) based on the density of the cell
with and without immunosuppressive therapy, and between early onset surface marker CD56. Most human NK cells (approximately 90%) ex­
and late-onset patients. From a clinical perspective, our results show the press CD56 at a relatively low-density (CD56dim), and this is associated

7
R. Fan et al. Clinical Immunology 245 (2022) 109184

Fig. 5. Functional characterization of VSIR+ monocytes. A. Heatmap of gene signatures in VSIR+Mo and VSIR-Mo. B. Network graph of top10 GO enriched terms. C.
Network graph of top 10 KEGG enriched terms. D. Gene signatures enriched in the antigen processing and presentation pathway. E. Mass cytometry data showing the
expression of HLA-DR in VSIR+/− monocytes. F. tSNE analysis revealed heterogeneity of neutrophils in MG patients and HCs, and distinguished clusters of CD38+
cells. G. Expression of CD38 in neutrophils from MG patients and HCs. Statistical analysis performed using unpaired t-tests (significance level: ns >0.05; *0.01 < p <
0.05; **0.001 < p < 0.01; *** < 0.001). HC: healthy controls; MG: myasthenia gravis; VSIRneg_Mo: VSIR-Monocytes; VSIRpos_Mo: VSIR+Monocytes.

with high cytotoxicity. In contrast, the other human NK cell type CD19- CD14- CD56+ cells from human blood and subjected them to
(approximately 10%) expresses CD56 at a relatively high density single-cell RNA sequencing. The results showed that NK cells were
(CD56bright) in the peripheral blood. This cell type is less cytotoxic but divided into two clusters by an unsupervised clustering algorithm. One
more efficient in cytokine and chemokine production compared to the was characterized by FCGR3A (encoding CD16), with gene signatures of
CD56dim subset [17]. Notably, recent studies have shown that classi­ a CD56+ FCGR3A+ NK cell cluster enriched in cytolysis function, while
fying the cytotoxicity and cytokine-producing ability of NK cells by the gene signature of the other NK cell cluster was enriched in response
expression of CD56 is not a strict method; for example, cytokine- to cytokines and regulation of cytokine production functions. CD16
producing CD56bright NK cells may acquire cytotoxicity equal to (FCGR3A) is a low-affinity FcyRIII expressed on a small portion of
CD56dim upon specific stimulation by cytokines or receptor activation, CD56bright (50–70%) and in the majority of CD56dim cells (95%) [19],
and CD56dim NK cells may produce IFN-γ or other functional cytokines and mediates antibody-dependent cellular cytotoxicity (ADCC) by
under specific conditions [17]. Crinier et al. [18] isolated CD45+ CD3- binding to IgG-coated targets [21,22]. In our study, CD56+ CD16+ NK

8
R. Fan et al. Clinical Immunology 245 (2022) 109184

cell populations were decreased in MG patients, which may imply that were identified and a functional characterization was performed using
cytotoxicity and ADCC are decreased in NK cells from these patients. Liu single-cell RNA sequencing. Our results showed that 37 genes, including
et al. [22] evaluated the killing activity of NK cells from MG patients and the HLA gene family, were upregulated in VISTA+ monocytes, and these
reported the killing ability of these cells on CD4+ T cells was impaired. gene signatures were enriched in antigen presentation/processing in GO
Chien et al. [23] evaluated NK cell cytotoxicity in MG patients treated and KEGG. Moreover, IRF8 and IRF5 were the most enriched factors
with or without plasmapheresis, and with or without immunosuppres­ according to the transcription factor enrichment analysis. To the best of
sants, and reported that it was decreased after plasmapheresis and in the our knowledge, this study is the first to identify VISTA+ monocytes and
group treated with immunosuppressants. Our study further reveals that reveal their gene signatures.
the lower cytotoxicity of NK cells was secondary to the reduction in the Our results suggest the potential value of cellular therapies related to
number of CD56+ CD16+ NK cells. Reportedly, cytokines are closely VISTA, such as downregulating VISTA+ monocytes to reduce their an­
related to the phenotype and function of NK cells, and cytokine dysre­ tigen processing/presentation ability in autoimmune diseases, and
gulation has been observed in MG patients [25,5]. Previous research adoptive VISTA+ monocyte therapy to enhance the anti-tumor ability of
demonstrated concentration-dependent downregulation of CD16 the immune system. In addition, expression of ICPs (including VISTA,
expression at membrane surface of NK cells induced by IL-18 [26] and LAG3, and CTLA4) was also higher in T cells from MG patients, sug­
IL-18 levels were reported to be higher in MG patients than in HCs [27]. gesting that the expression of these ICPs is increased by some unknown
Tfh cells were significantly increased in MG patients, and IL-21 is the mechanism to regulate immune response, but the feedback is not suffi­
characteristic cytokine produced by these cells. Kasaian et al. [28] cient to elicit disease remission. This potential mechanism needs to be
indicated the production of IL-21 by activated T cells may limit the NK explored in further research.
cell response. We suggest that the lower number of CD56+ CD16+ NK Activation of functional populations of T and B cells is the typical
cells in MG patients could be due to regulation of cytokines related to NK mechanism of MG pathogenesis, but several studies have indicated that
function, and the frequency of CD56+ CD16+ NK cells may reflect the percentage of classical T cell subsets, including Th1/2/9/17, TEM,
regulation ability, with a higher number of CD56+ CD16+ NK cells and TCM [5,6] are similar to those observed in HCs, while the Tfh subset
indicating stronger regulation ability and relatively mild disease is significantly altered [14]. In our study, a marked alteration in T cell
severity, and vice versa. In addition, an abnormal percentage of NK cells subsets was not observed; however, CXCR5 expression was higher in MG
was also observed in other diseases, such as SLE and rheumatoid patients than in HCs. Upregulation of the expression of multiple ICPs in
arthritis (RA) (both the frequency of CD3- CD56+ NK cells and their the T cells of MG patients may imply functional dysregulation of T cells.
cytotoxicity were lower than in the control group) [29], type 1 diabetes B cells play a fundamental role in multiple autoimmune diseases, and
(the frequency of CD3- CD56+ NK cells was lower in recent-onset T1D B cell populations undergo a wide range of alterations (including
cases compared to those of long-standing T1D and to controls) [30], and transformation into memory B cells and transitional B cells) before
neuromyelitis optica syndrome disease (the frequency of CD3- CD56+ giving rise to antibody-secreting cells (ASCs) at different stages of
CD16+ NK cells was lower than in the control group) [31]. CD56+ autoimmune diseases. In SLE, IgD- CD27- memory B cells [35,36],
CD16+ CD8+ NK cells were also identified in our study, and showed CD27- IgD- CD95+ memory B cells [38], and CD23- IgD+ CD27- acti­
trend that was similar to that of CD56 + CD16+ cells. McKinney et al. vated naïve B cells [39] are increased and correlate with disease
[32] suggested that CD8+ NK cells are associated with the outcome of severity. In MG, CD19- CD138- ASCs are also increased and correlate
relapsing/remitting multiple sclerosis as well as with the regulation of with disease severity [40], and memory B cells (CD19 + CD27 + CD38-)
CD4+ T cell activation and proliferation. In HIV, the frequency of highly are increased in GMG and OMG compared with HCs [41]. Our results
functional CD8+ NK cells is negatively correlated with HIV-related demonstrate that switched memory B cells (CD19+ CD27+ IgD-),
disease markers [33]. Recent research on CD8+ NK cells mainly including CD38+ and CD38-, are significantly increased, but this in­
focused on infectious diseases, and few studies have focused on auto­ crease did not correlate with disease severity. CD38 is a type II glyco­
immune diseases. Therefore, the role of CD8+ NK cells in autoimmune protein widely expressed in almost all developmental stages of B cells,
diseases is still poorly understood and needs to be further explored. and is highly expressed in early B cells [42], suggesting that it may be
We also found that the frequency of VISTA+CD14 + monocytes in involved in developmental pathways, including activation and prolif­
the MG group was markedly increased, and the mRNA level of VSIR was eration [43]. CD38 is also expressed in plasmablasts, and in short-lived
also significantly increased in monocytes from MG patients, suggesting and long-lived plasma cells, implying that CD38 may be an interesting
that increased VISTA protein levels may be due to increased levels of target for targeted therapy aimed at depleting ASCs. In addition, the
mRNA. VISTA is a novel immune checkpoint that is highly expressed on percentage of HLA- DQ- CD38+ naïve B cells was higher in MG patients
myeloid cells and T cells [34] and is considered to have therapeutic and was correlated with disease severity. The expression of CD38 was
potential because of its suppressive ability on the immune system, as significantly increased in MG patients; however, the expression of CD38
immune response improved after anti-VISTA therapy or in cases of in total T cells and B cells was not significantly different, and functional
VISTA deficiency. Han et al. [35] suggested that the expression of VISTA characterization of HLA- DQ- CD38+ naïve B cells through future
is increased in SLE and discoid lupus erythematosus; VISTA knockout research is needed. CD38 is required for migration of neutrophils to the
mice with a BALB/c background spontaneously develop autoimmune site of infection or inflammation [44], and CD38 expression is closely
diseases resembling human lupus. Furthermore, the immune response correlated with chemotactic migration to formylmethionine leucyl-
was reduced in MRL/lpr mice after treatment with monoclonal antibody phenylalanine in localized aggressive periodontitis patients [45].
against VISTA, which suggests a therapeutic potential of VISTA in Neutrophil numbers are significantly higher in the spleens and livers of
autoimmune diseases. However, no study has explored the relationship wild type BALB/c mice upon Listeria infection, but lower in the spleen of
between MG and VISTA. Although emerging data has revealed an CD38 knockdown mice [46]. Neutrophil migration can mediate joint
important role for VISTA in the regulation of innate immune cell func­ damage and persistent inflammation by migrating into synovial joints
tion, there is still controversy on whether VISTA is an activating or an and triggering Rheumatoid arthritis [47]. We suggest that MG neutro­
inhibitory receptor in monocytes. In myeloid cells, overexpression of phils may preferentially migrate to the neuromuscular junction and
VISTA reduced MHC-II expression, and anti-VISTA (a VISTA agonist) enhance the inflammatory response due to their high levels of expres­
downregulates MHC-II genes in monocytes [15]; however, another sion of CD38; however, the regulatory mechanism needs to be further
monoclonal antibody against VISTA upregulates MHC-II genes [16]. explored. The primary limitation of our research is the limited sample
These contradictory results have limited the development of anti-ICP size due to the strict inclusion criteria adopted. To address this short­
therapies based on VISTA. To further explore the biological function coming, we validated our results with an external validation cohort. In
of VISTA in monocytes, gene signatures for VISTA+ CD14+ monocytes addition, this study was a cross-sectional study, which could not

9
R. Fan et al. Clinical Immunology 245 (2022) 109184

dynamically assess the immune changes, especially the crosstalk be­ N.G. Núñez, T. Marafioti, D. Schneiter, I. Opitz, A. Lanzavecchia, H.H. Jung, D. De
Feo, S. Mundt, B. Schreiner, B. Becher, Single-cell profiling of myasthenia gravis
tween different immune cells. Therefore, future longitudinal follow-up
identifies a pathogenic T cell signature, Acta Neuropathol. 141 (2021) 901–915,
studies are necessary. https://doi.org/10.1007/s00401-021-02299-y.
[7] S. Qin, L. Xu, M. Yi, S. Yu, K. Wu, S. Luo, Novel immune checkpoint targets: moving
beyond PD-1 and CTLA-4, Mol. Cancer 18 (2019) 155, https://doi.org/10.1186/
5. Conclusion
s12943-019-1091-2.
[8] G. Wang, R. Tai, Y. Wu, S. Yang, J. Wang, X. Yu, L. Lei, Z. Shan, N. Li, The
In summary, we revealed the dysregulation of multiple immune cell expression and immunoregulation of immune checkpoint molecule VISTA in
populations, identified several cellular biomarkers related to MG disease autoimmune diseases and cancers, Cytokine Growth Factor Rev. 52 (2020) 1–14,
https://doi.org/10.1016/j.cytogfr.2020.02.002.
severity, and highlighted large alterations in NK cell subsets. Further­ [9] Z. Wu, M. Wang, G. Liang, P. Jin, P. Wang, Y. Xu, Y. Qian, X. Jiang, J. Qian,
more, we discovered alterations in VISTA in MG patients and identified M. Dong, Pro-Inflammatory Signature in decidua of recurrent pregnancy loss
the characteristics of VISTA+ monocytes. regardless of embryonic chromosomal abnormalities, Front. Immunol. 12 (2021),
772729, https://doi.org/10.3389/fimmu.2021.772729.
Supplementary data to this article can be found online at https://doi. [10] R. Finck, E.F. Simonds, A. Jager, S. Krishnaswamy, K. Sachs, W. Fantl, D. Peer, G.
org/10.1016/j.clim.2022.109184. P. Nolan, S.C. Bendall, Normalization of mass cytometry data with bead standards,
Cytometry A. 83 (2013) 483–494, https://doi.org/10.1002/cyto.a.22271.
[11] H. Chen, M.C. Lau, M.T. Wong, E.W. Newell, M. Poidinger, J. Chen, Cytofkit: A
Funding bioconductor package for an integrated mass cytometry data analysis pipeline,
PLoS Comput. Biol. 12 (2016), e1005112, https://doi.org/10.1371/journal.
This work was supported by Future Medical Youth Innovation Team pcbi.1005112.
[12] Z. Zhang, D. Luo, X. Zhong, J.H. Choi, Y. Ma, S. Wang, E. Mahrt, W. Guo, E.
Program of Chongqing Medical University (No. W0043), the Fifth Senior W. Stawiski, Z. Modrusan, S. Seshagiri, P. Kapur, G.C. Hon, J. Brugarolas, T. Wang,
Medical Talents Program of Chongqing for Young and Middle-aged, SCINA: a semi-supervised subtyping algorithm of single cells and bulk samples,
Middle-aged Medical Excellence Team Program of Chongqing, and Genes (Basel) 10 (2019) E531, https://doi.org/10.3390/genes10070531.
[13] J.W. Squair, M. Gautier, C. Kathe, M.A. Anderson, N.D. James, T.H. Hutson,
Chongqing chief expert studio project, China. R. Hudelle, T. Qaiser, K.J.E. Matson, Q. Barraud, A.J. Levine, G.L. Manno, M.
A. Skinnider, G. Courtine, Confronting false discoveries in single-cell differential
Data statement expression, Nat. Commun. 12 (2021), https://doi.org/10.1038/s41467-021-
25960-2.
[14] S. Ashida, H. Ochi, M. Hamatani, C. Fujii, K. Kimura, Y. Okada, Y. Hashi,
Data are available on request to the authors. K. Kawamura, H. Ueno, R. Takahashi, T. Mizuno, T. Kondo, Immune skew of
circulating follicular helper T cells associates with myasthenia gravis severity,
Neurol Neuroimmunol Neuroinflamm. 8 (2021), e945, https://doi.org/10.1212/
Contributors NXI.0000000000000945.
[15] E. Ma, Z. Y, S. E, B. Cm, M. R, C. C, N. Rj, VISTA: A Target to Manage the Innate
The study was conceived, designed by F.X., R.F. and W.J.Q.; Z.T.L., Cytokine Storm, Front. Immunol. 11 (2021), https://doi.org/10.3389/
fimmu.2020.595950.
W.Z., X.G., L.Q.L. contributed to sample collection and clinical assess­ [16] B.M. Rogers, L. Smith, Z. Dezso, X. Shi, E. DiGiammarino, D. Nguyen,
ments; R.F. performed mass cytometry data analysis and bioinformatic S. Sethuraman, P. Zheng, D. Choi, D. Zhang, A. Nguyen, K. McGuire, W. Liu,
analysis; R.F. and W.J.Q. drafted the manuscript and all authors dis­ N. Chung, D.T. Chao, S. Ye, G.R. Starbeck-Miller, VISTA is an activating receptor in
human monocytes, J. Exp. Med. 218 (2021), https://doi.org/10.1084/
cussed the results and contributed to the writing of the paper. R.F and W.
jem.20201601.
J.Q. contributed equally to this work as co-first authors. [17] B. Zitti, Y.T. Bryceson, Natural killer cells in inflammation and autoimmunity,
Cytokine Growth Factor Rev. 42 (2018) 37–46, https://doi.org/10.1016/j.
cytogfr.2018.08.001.
Declaration of Competing Interest [18] C. A, M. P, E. B, P. C, G. J, B. A, S. L, C.-M. I, E. M, G.-M. M, J. S, B. E, H. S, H. J, U.
S, V. F, N.-M. E, V. E, High-dimensional single-cell analysis identifies organ-specific
The authors declare no conflicts of interest. signatures and conserved NK Cell subsets in humans and mice, Immunity 49
(2018), https://doi.org/10.1016/j.immuni.2018.09.009.
[19] M.A. Cooper, T.A. Fehniger, M.A. Caligiuri, The biology of human natural killer-
Data availability cell subsets, Trends Immunol. 22 (2001) 633–640, https://doi.org/10.1016/s1471-
4906(01)02060-9.
[21] K. Kitaya, Accumulation of uterine CD16(− ) natural killer (NK) cells: friends, foes,
Data will be made available on request. or Jekyll-and-Hyde relationship for the conceptus? Immunol. Investig. 37 (2008)
467–481, https://doi.org/10.1080/08820130802191292.
Acknowledgments [22] R.-T. Liu, W. Li, D. Guo, C.-L. Yang, J. Ding, J.-X. Xu, R.-S. Duan, Natural killer cells
promote the differentiation of follicular helper T cells instead of inducing apoptosis
in myasthenia gravis, Int. Immunopharmacol. 98 (2021), 107880, https://doi.org/
We greatly appreciate the all participating patients and thank the 10.1016/j.intimp.2021.107880.
mass cytometry facility (PLT Tech, China) for mass cytometry experi­ [23] P.J. Chien, J.H. Yeh, H.C. Chiu, Y.M. Hsueh, C.T. Chen, M.C. Chen, C.M. Shih,
Inhibition of peripheral blood natural killer cell cytotoxicity in patients with
ment and single-cell sample preparation. myasthenia gravis treated with plasmapheresis, Eur. J. Neurol. 18 (2011)
1350–1357, https://doi.org/10.1111/j.1468-1331.2011.03424.x.
References [25] X. Huan, S. Luo, H. Zhong, X. Zheng, J. Song, L. Zhou, J. Lu, Y. Wang, Y. Xu, J. Xi,
Z. Zou, S. Chen, C. Zhao, In-depth peripheral CD4+ T profile correlates with
myasthenic crisis, Ann Clin Transl Neurol. 8 (2021) 749–762, https://doi.org/
[1] N.E. Gilhus, J.J. Verschuuren, Myasthenia gravis: subgroup classification and
10.1002/acn3.51312.
therapeutic strategies, Lancet Neurol. 14 (2015) 1023–1036, https://doi.org/
[26] C.M. Nielsen, A.-S. Wolf, M.R. Goodier, E.M. Riley, Synergy between Common γ
10.1016/S1474-4422(15)00145-3.
Chain Family Cytokines and IL-18 Potentiates Innate and Adaptive Pathways of NK
[2] A.S. Carr, C.R. Cardwell, P.O. McCarron, J. McConville, A systematic review of
Cell Activation, Front. Immunol. 7 (2016) 101, https://doi.org/10.3389/
population based epidemiological studies in Myasthenia Gravis, BMC Neurol. 10
fimmu.2016.00101.
(2010) 46, https://doi.org/10.1186/1471-2377-10-46.
[27] S. Jander, G. Stoll, Increased serum levels of the interferon-gamma-inducing
[3] J.M. Ponseti, E. Espín, M. Armengol, Diagnóstico y tratamiento de la miastenia
cytokine interleukin-18 in myasthenia gravis, Neurology. 59 (2002) 287–289,
grave, Med. Clin. 115 (2000) 264–270, https://doi.org/10.1016/S0025-7753(00)
https://doi.org/10.1212/wnl.59.2.287.
71529-6.
[28] M.T. Kasaian, M.J. Whitters, L.L. Carter, L.D. Lowe, J.M. Jussif, B. Deng, K.
[4] M.I. Leite, S. Jacob, S. Viegas, J. Cossins, L. Clover, B.P. Morgan, D. Beeson,
A. Johnson, J.S. Witek, M. Senices, R.F. Konz, A.L. Wurster, D.D. Donaldson,
N. Willcox, A. Vincent, IgG1 antibodies to acetylcholine receptors in ‘seronegative’
M. Collins, D.A. Young, M.J. Grusby, IL-21 limits NK cell responses and promotes
myasthenia gravis†, Brain. 131 (2008) 1940–1952, https://doi.org/10.1093/
antigen-specific T cell activation: a mediator of the transition from innate to
brain/awn092.
adaptive immunity, Immunity. 16 (2002) 559–569, https://doi.org/10.1016/
[5] X. Huan, S. Luo, H. Zhong, X. Zheng, J. Song, L. Zhou, J. Lu, Y. Wang, Y. Xu, J. Xi,
s1074-7613(02)00295-9.
Z. Zou, S. Chen, C. Zhao, In-depth peripheral CD4+ T profile correlates with
[29] Y.-W. Park, S.-J. Kee, Y.-N. Cho, E.-H. Lee, H.-Y. Lee, E.-M. Kim, M.-H. Shin, J.-
myasthenic crisis, Ann Clin Transl Neurol. 8 (2021) 749–762, https://doi.org/
J. Park, T.-J. Kim, S.-S. Lee, D.-H. Yoo, H.-S. Kang, Impaired differentiation and
10.1002/acn3.51312.
cytotoxicity of natural killer cells in systemic lupus erythematosus, Arthritis
[6] F. Ingelfinger, S. Krishnarajah, M. Kramer, S.G. Utz, E. Galli, M. Lutz, P. Zwicky, A.
Rheum. 60 (2009) 1753–1763, https://doi.org/10.1002/art.24556.
U. Akarca, N.P. Jurado, C. Ulutekin, D. Bamert, C.C. Widmer, L. Piccoli, F. Sallusto,

10
R. Fan et al. Clinical Immunology 245 (2022) 109184

[30] M. Rodacki, B. Svoren, V. Butty, W. Besse, L. Laffel, C. Benoist, D. Mathis, Altered by expression of CD27, IgD, and CD95, Arthritis Rheum. 58 (2008) 1762–1773,
natural killer cells in type 1 diabetic patients, Diabetes. 56 (2007) 177–185, https://doi.org/10.1002/art.23498.
https://doi.org/10.2337/db06-0493. [39] C.M. Tipton, C.F. Fucile, J. Darce, A. Chida, T. Ichikawa, I. Gregoretti, S. Schieferl,
[31] L. Khani, M.H. Jazayeri, R. Nedaeinia, M. Bozorgmehr, S.M. Nabavi, G.A. Ferns, J. Hom, S. Jenks, R.J. Feldman, R. Mehr, C. Wei, F.E.-H. Lee, W.C. Cheung, A.
The frequencies of peripheral blood CD5+CD19+ B cells, CD3-CD16+CD56+ NK, F. Rosenberg, I. Sanz, Diversity, cellular origin and autoreactivity of antibody-
and CD3+CD56+ NKT cells and serum interleukin-10 in patients with multiple secreting cell population expansions in acute systemic lupus erythematosus, Nat.
sclerosis and neuromyelitis optica spectrum disorder, Allergy, Asthma Clin. Immunol. 16 (2015) 755–765, https://doi.org/10.1038/ni.3175.
Immunol. 18 (2022) 5, https://doi.org/10.1186/s13223-021-00596-5. [40] C.-J. Zhang, Y. Gong, W. Zhu, Y. Qi, C.-S. Yang, Y. Fu, G. Chang, Y. Li, S. Shi,
[32] M. Ef, C. I, H. Km, S. De, C. C, M. G, C. Ej, Z. Ss, S. Kgc, A CD8 + NK cell K. Wood, S. Ladha, F.-D. Shi, Q. Liu, Y. Yan, Augmentation of Circulating Follicular
transcriptomic signature associated with clinical outcome in relapsing remitting Helper T Cells and Their Impact on Autoreactive B Cells in Myasthenia Gravis,
multiple sclerosis, Nature, Communications. 12 (2021), https://doi.org/10.1038/ J. Immunol. 197 (2016) 2610–2617, https://doi.org/10.4049/jimmunol.1500725.
s41467-020-20594-2. [41] Y. Hu, J. Wang, J. Rao, X. Xu, Y. Cheng, L. Yan, Y. Wu, N. Wu, X. Wu, Comparison
[33] F. Ahmad, H.S. Hong, M. Jäckel, A. Jablonka, I.-N. Lu, N. Bhatnagar, J. of peripheral blood B cell subset ratios and B cell-related cytokine levels between
M. Eberhard, B.A. Bollmann, M. Ballmaier, M. Zielinska-Skowronek, R.E. Schmidt, ocular and generalized myasthenia gravis, Int. Immunopharmacol. 80 (2020),
D. Meyer-Olson, High frequencies of polyfunctional CD8+ NK cells in chronic HIV- 106130, https://doi.org/10.1016/j.intimp.2019.106130.
1 infection are associated with slower disease progression, J. Virol. 88 (2014) [42] R.-R. H, M.-G. Mt, P. R, L.-S. R, S.-A. L, CD38 expression in early B-cell precursors
12397–12408, https://doi.org/10.1128/JVI.01420-14. contributes to extracellular signal-regulated kinase-mediated apoptosis,
[34] I. Le Mercier, W. Chen, J.L. Lines, M. Day, J. Li, P. Sergent, R.J. Noelle, L. Wang, Immunology. 144 (2015), https://doi.org/10.1111/imm.12370.
VISTA regulates the development of protective antitumor immunity, Cancer Res. [43] M.-O. N, M.-G. Me, F. K, S.-A. L, CD38 cross-linking enhances TLR-induced B cell
74 (2014) 1933–1944, https://doi.org/10.1158/0008-5472.CAN-13-1506. proliferation but decreases IgM plasma cell differentiation, Eur. J. Immunol. 37
[35] X. Han, M.D. Vesely, W. Yang, M.F. Sanmamed, T. Badri, J. Alawa, F. López- (2007), https://doi.org/10.1002/eji.200636453.
Giráldez, P. Gaule, S.W. Lee, J.-P. Zhang, X. Nie, A. Nassar, A. Boto, D.B. Flies, [44] F.E. Lund, D.A. Cockayne, T.D. Randall, N. Solvason, F. Schuber, M.C. Howard,
L. Zheng, T.K. Kim, G.W. Moeckel, J.M. McNiff, L. Chen, PD-1H (VISTA)-mediated CD38: a new paradigm in lymphocyte activation and signal transduction, Immunol.
suppression of autoimmunity in systemic and cutaneous lupus erythematosus, Sci. Rev. 161 (1998) 79–93, https://doi.org/10.1111/j.1600-065x.1998.tb01573.x.
Transl. Med. 11 (2019) eaax1159, https://doi.org/10.1126/scitranslmed.aax1159. [45] T. Fujita, A. Kantarci, M.L. Warbington, K.H. Zawawi, H. Hasturk, H. Kurihara, T.
[36] C. Wei, J. Anolik, A. Cappione, B. Zheng, A. Pugh-Bernard, J. Brooks, E.-H. Lee, E. E. Van Dyke, CD38 expression in neutrophils from patients with localized
C.B. Milner, I. Sanz, A new population of cells lacking expression of CD27 aggressive periodontitis, J. Periodontol. 76 (2005) 1960–1965, https://doi.org/
represents a notable component of the B cell memory compartment in systemic 10.1902/jop.2005.76.11.1960.
lupus erythematosus, J. Immunol. 178 (2007) 6624–6633, https://doi.org/ [46] T. Lischke, K. Heesch, V. Schumacher, M. Schneider, F. Haag, F. Koch-Nolte, H.-
10.4049/jimmunol.178.10.6624. W. Mittrücker, CD38 controls the innate immune response against Listeria
[38] A.M. Jacobi, K. Reiter, M. Mackay, C. Aranow, F. Hiepe, A. Radbruch, A. Hansen, monocytogenes, Infect. Immun. 81 (2013) 4091–4099, https://doi.org/10.1128/
G.-R. Burmester, B. Diamond, P.E. Lipsky, T. Dörner, Activated memory B cell IAI.00340-13.
subsets correlate with disease activity in systemic lupus erythematosus: delineation [47] W. Hl, M. Rj, E. Sw, The multifactorial role of neutrophils in rheumatoid arthritis,
Nat. Rev. Rheumatol. 10 (2014), https://doi.org/10.1038/nrrheum.2014.80.

11

You might also like